ArticlePDF Available

Abstract and Figures

A long-pursued goal in cancer treatment is to deliver a therapy specifically to metastases. As a result of the disseminated nature of the metastatic disease, carrying the therapeutic agent to the sites of tumor growth represents a major step for success. We hypothesized that tumor cells injected intravenously (i.v.) into an animal with metastases would respond to many of the factors driving the metastatic process, and would target metastases. Using a model of spontaneous metastases, we report here that i.v. injected tumor cells localized on metastatic lesions. Based on this fact, we used genetically transduced tumor cells for tumor targeting of anticancer agents such as a suicide gene or an oncolytic virus, with evident antitumoral effect and negligible systemic toxicity. Therefore, autologous tumor cells may be used as cellular vehicles for systemic delivery of anticancer therapies to metastatic tumors.
Content may be subject to copyright.
Tumor cells as cellular vehicles to deliver gene therapies
to metastatic tumors
Javier Garcı
´
a-Castro,
1,5
Jesu
´
s Martı
´
nez-Palacio,
1
Rosa Lillo,
2
Fe
´
lix Garcı
´
a-Sa
´
nchez,
2
Ramo
´
n Alemany,
3
Luis Madero,
4
Juan A Bueren,
1
and Manuel Ramı
´
rez
4
1
Unidad de Hematopoyesis y Terapia Ge
´
nica, CIEMAT/Fundacio
´
n Marcelino Botı
´
n, Madrid, Spain;
2
Centro
de Transfusio
´
n, Madrid, Spain;
3
Institut Catala
`
d’Oncologı
´
a, Barcelona, Spain; and
4
Oncologı
´
a Pedia
´
trica,
Hospital Universitario Nin
˜
o Jesu
´
s, Madrid, Spain.
A long-pursued goal in cancer treatment is to deliver a therapy specifically to metastases. As a result of the disseminated nature of
the metastatic disease, carrying the therapeutic agent to the sites of tumor growth represents a major step for success. We
hypothesized that tumor cells injected intravenously (i.v.) into an animal with metastases would respond to many of the factors
driving the metastatic process, and would target metastases. Using a model of spontaneous metastases, we report here that i.v.
injected tumor cells localized on metastatic lesions. Based on this fact, we used genetically transduced tumor cells for tumor
targeting of anticancer agents such as a suicide gene or an oncolytic virus, with evident antitumoral effect and negligible systemic
toxicity. Therefore, autologous tumor cells may be used as cellular vehicles for systemic delivery of anticancer therapies to
metastatic tumors.
Cancer Gene Therapy advance online publication, 14 January 2005; doi:10.1038/sj.cgt.7700801
Keywords: neoplasm metastasis; delivery system; oncolytic virus; NOD SCID mice
C
ancer is often diagnosed when the disease has already
disseminated and most of the patient deaths are
related to metastatic disease. Current treatment options
for metastatic tumors lack efficacy and metastases
targeting remains a major challenge for curing cancer.
Novel cell- and gene-based therapies have been developed
aimed to deliver an antitumor effect specifically to
metastases. Some strategies rely on the immune system
for selectivity, such as the stimulation of effector immune
cells
1
or the use of monoclonal antibodies that recognize
tumor antigens.
2
Others target angiogenic
3–5
or anaerobic
signals
6,7
that arise at the metastases. Recently, onco-
tropic viruses have been used to selectively kill the tumor
cells with the advantage that once targeting is achieved
the propagation of the virus amplifies the therapy.
8,9
These promising new therapies have fallen behind
expectations mainly because of their limited capacity for
effectively targeting in vivo.
2,10
The process of metastasis is a turning point in the
progression of malignant solid tumors. Million tumor
cells leave the primary tumor, reach the bloodstream and
disseminate through the body.
11
Only a minority will
eventually survive to become the origin of a new cancer
nodule
12
at anatomical sites that are specific for the tumor
type.
13
The dissemination of cancer cells from the primary
tumor and their homing in specific organs involve several
steps: invasion, detachment, circulation, cell adhesion,
motility and invasion again. Cells must express a specific
receptor molecule repertoire (cell adhesion molecules,
chemokine receptors or integrin ligands among others) to
complete this metastatic process.
14–17
We hypothesized that cancer cells may be good
candidates to target established metastases in vivo because
they express the receptor and effector molecules involved
in the metastatic process. Autologous intravenously (i.v.)
injected tumor cells should respond to metastasis-related
cues as the cells leaving the primary tumor did when the
latter formed the metastases. In the present work, we
demonstrated that i.v. injected tumor cells localized on
established metastases and were able of carrying genetic-
based antitumor agents such as suicide genes or oncolytic
viruses to the metastases, delivering the therapy in a
localized and toxic-tempered fashion.
Methods
Cell lines
The human breast cancer cell line MDA-MB-231 and the
human prostate cancer cell line PC3 were maintained in
DMEM supplemented with 10% fetal bovine serum
Received May 31, 2004.
Address correspondence and reprint requests to: Dr Manuel
Ramı
´
rez, MD, PhD, Oncologı
´
a Pedia
´
trica, Hospital Universitario
Nin
˜
o Jesu
´
s, Avenida Mene
´
ndez Pelayo, 65, 28009-Madrid, Spain.
E-mail: mramirezo.hnjs@salud.madrid.org
5
Present address: Servicio de Oncologı
´
a, Hospital Nin
˜
o Jesu
´
s,
Madrid.
Cancer Gene Therapy (2005), 19
r
2005 Nature Publishing Group All rights reserved 0929-1903/05 $30.00
www.nature.com
/
cgt
(FBS), glutamine and antibiotics (Life Technologies, Inc.,
Grand Island, NY).
Transduction of tumor cells with adenovectors or
oncolytic adenoviruses
MDA-MB-231 or PC3 cells were transduced with
different adenoviral vectors or oncolytic adenoviruses
depending on the experiments. The adenovectors had
either the lac-Z gene (AdLacZ), or the cytosine deaminase
(CD) gene (AdCD
18
), under the control of the cytome-
galovirus (CMV) promoter. The oncolytic adenovirus
(AdwtFi-GFP) contains the wild-type E1 region and,
after the fiber sequence (L5), the GFP coding sequence
has been inserted with a splicing acceptor from the IIIa
adenovirus gene. In this way, the adenovirus major late
promoter regulates GFP, and viral replication can be
followed by detection of green fluorescent cells.
The cells were transduced in DMEM, under conditions
optimized so that 100% of the cells were infected: 1hour
infection with 100 pfu/cell.
18
Evaluation of bystander effect in vitro
Tumor cells were mixed with different proportions of
AdCD transduced cells, and cultured in the presence of
400 mM 5-fluorocytosine (5-FC, Sigma Chemical Co., St
Louis, MO). The cell viability was evaluated after 4 days
by Trypan-blue (Sigma) exclusion of viable cells and
crystal violet staining. In different experiments, tumor
cells were mixed with different proportions of Ad-
wtFiGFP-infected cells. The cell viability was evaluated
after 6 days by Trypan-blue exclusion of viable cells and
green fluorescence determination.
In vivo studies
We used 6–8-week-old NOD.CB17-Prkdcscid/J (NOD
scid) mice, bred at the CIEMAT Laboratory Animals
Facility (Registration Number 28079-21 A) from breeding
pairs originally obtained from Jackson Lab (Bar Harbor,
Maine). The mice were routinely screened for pathogens,
in accordance with FELASA (Federation of European
Laboratory Animal Science Associations) procedures.
They were housed in microisolator individually ventilated
cages, and allowed water and food ad libitum. All
experimental procedures were carried out according to
European and Spanish laws and regulations and internal
biosafety and bioethics guidelines.
MDA-MB-231 cells were implanted in the mammary
fat pad of female NOD/SCID mice while PC3 cells were
implanted in the subcutaneous flank of male NOD/SCID
mice. At indicated time points, the animals were
euthanized by CO
2
inhalation. Samples from different
organs were collected in 10% buffered formalin, Tissue-
Tek (O.C.T. Compound, Zoeterwoude, The Netherlands)
or immediately frozen.
Detection of tumor cells by RT-PCR
RNA was purified using the TRIzol reagent (Life
Technologies). RT-PCR amplification of a human cyto-
keratin-19 sequence was carried out as published else-
where.
18
b-gal þ cells were detected using the following
lac-Z primers (sense: 5
0
CCGATCGCGTCACACTAC3
0
and antisense: 5
0
CAGATGATCACACTCGGG 3
0
).
Histology and immunohistochemistry
Sections of paraffin-embedded organs were stained with
hematoxilin–eosin, or with the CAM5.2 monoclonal
antibody (anti-human pancytokeratin, Becton Dickinson,
San Jose, CA) using a standard indirect avidin–biotin
horseradish peroxidase method (Vectastain ABC kit,
Vector Laboratories Inc., Burlingame, CA). Color was
developed with diaminobenzidine (DAB, Vector Labora-
tories Inc.) and sections were counterstained with
hematoxylin. Transduced tumor cells were stained with
the anti-b-galactosidase Ab1 antibody (Oncogene, Cam-
bridge, MA) and counterstained with eosin. GFP
þ
cells
were stained with a rabbit antiGFP IgG fraction
(Molecular Probes, Eugene, OR) and counterstained with
hematoxylin.
Cryosections were stained with X-gal (5-bromo-4-
chloro-3-indolyl-b-
D-galactoside, United States Biochem-
ical Corp., Cleveland, OH) and then with the CAM 5.2
antibody, and counterstained with eosin.
Evaluation of lung area, lung metastases and
localization of the i.v. injected tumor cells
Images of the lung cryosections stained with the CAM 5.2
antibody (n ¼ 10, two animals) were digitalized and
analyzed with the IMAGE-PRO Plus 4.5 software (Media
Cybernetics Inc., Carlsbad, CA). Different colors were
assigned to healthy and to metastatic tissues, and the total
lung and metastatic areas were calculated in square pixels
for each color, by an independent researcher (Dr Jose
´
M
Martı
´
nez, departamento de Ciencias de Materiales,
Universidad Polite
´
cnica de Madrid). The associated error
was estimated as less than 4% in triplicate measurements.
The number of b-gal þ cells and its localization in the
lungs were directly scored under a light microscope on
lung cryosections stained with X-gal and the CAM 5.2
antibody (n ¼ 10, two animals). Colocalization was
defined as the presence of a b-gal þ cell directly in
contact with a cytokeratin-positive nodule. All other
situations were not considered as colocalization.
Evaluation of bystander effect in vivo
(a) Cells transduced with the AdCD vector. Metastatic
tumors were generated as explained before. The primary
tumors were excised and a group of mice received five
doses of 3 10
5
transduced cancer cells, two doses per
week. An osmotic pump (Alza Corporation, Palo Alto,
CA) delivering 5-FC at a rate of 0.25 mL/hour 5-FC
(10 mg/mL) was subcutaneously implanted in the animals.
At 10 days after the last i.v. injection, the mice were killed,
the diameters of the remaining primary tumors were
scored and the organs were recovered for histology and
quantification studies. Metastatic burden was evaluated
on DNA samples from the organs by dot blot hybridiza-
Delivering antitumor therapies using tumor cells
J Garcı´a-Castro et al
2
Cancer Gene Therapy
tion with a human specific probe (a kind gift of Dr John
Dick).
(b) Cells infected with the AdwtFiGFP virus. Metastatic
tumors were generated as explained before. The primary
tumors were excised and a group of mice received five
doses of 3 10
5
transduced cancer cells, two doses per
week. At 10 days after the last i.v. injection, the mice were
killed, the fresh organs were analyzed under a fluorescent
light and the lungs were weighed and recovered for
immunohistochemical analysis.
Statistical analysis
Statistical analysis was performed using the Stata
software program (Stata Press, College Station, TX).
Results are expressed as mean7standard error (SE).
Results were considered significant if the P value was
equal to or less than .05. We used the two-sample
Wilcoxon rank-sum test (Mann–Whitney two-sample
statistic) for comparisons.
Results
Tumor cells colocalized on pre-existing metastases
We generated metastatic cancer in mice by implanting
MDA-MB-231 human breast cancer cells in the mam-
mary fat of female NOD/SCID mice. In this model,
human breast cancer cells formed a primary tumor in the
mammary fat, and metastasized preferentially to the local
lymph nodes, lungs, kidneys, liver and other organs (Fig
S1). In a different experiment, we determined whether i.v.
injected tumor cells would localize on pre-existing
metastases. In mice with metastatic disease, we injected
i.v. a single dose of MDA-MB-231 cells transduced with
an adenoviral vector carrying the lac-Z transgene.
Transduction conditions were optimized so that all the
injected cells expressed the transgene.
18
At 24 hours after
i.v. injection, we determined the localization of the
transduced cancer cells. Immunohistochemical analysis
showed b-gal þ cells in micrometastatic nodules of the
lungs and kidneys (Fig 1). Microscopic examination
(serial sections of 5 mm) showed that 46% of the b-gal þ
cells in the lungs colocalized with micrometastases. A
total of 19% of the total lung area was tumoral and an
average of 20% of the lung metastases had b-gal þ cells
after a single i.v. injection of 3 10
5
transduced tumor
cells. We did not detect b-galactosidase activity (X-gal
staining) in the preparations we studied from the kidney
(10 sections), liver (10 sections) and cerebrum (10
sections). Using a different detection technique, we
confirmed the presence of b-gal þ cells in the primary
tumor and in organs with metastases by RT-PCR of the
lac-Z transgene (Fig S1). These experiments demonstrated
that i.v. injected tumor cells targeted metastatic lesions. In
addition, injected tumor cells carried a heterologous
molecule to the metastases. Preliminary experiments using
the same approach with PC3 (human prostate) cancer
cells in male NOD/SCID mice confirmed that i.v. infused
cancer cells localized on established metastases.
Ex vivo manipulated tumor cells exerted a bystander
toxic effect in vitro and in vivo on the metastases
In the light of these findings, we used the tumor cells for
cancer treatment. MDA-MB-231 or PC3 cancer cells were
transduced with an adenoviral vector carrying the
Escherichia coli enzyme cytosine deaminase (CD)
(AdCD). The cells expressing the CD transform the
prodrug 5-FC into the anticancer drug 5-fluoruracil (5-
FU), which eliminates the transduced cells and the cells
around them (bystander effect). Transduced cells were
mixed with different proportions of untransduced cells
and cultured in the presence of 5-FC. The viability of the
mixture decreased when 5% of cells in the starting
cultures were transduced (Fig S2). We next studied
whether the ex vivo manipulated tumor cells have
therapeutic effect in vivo. We generated metastatic cancer
in mice (as explained) and, when the primary tumor
reached a major axis of 10 mm, we surgically eliminated as
much tumor as possible before delivering the treatment.
For treatment, we implanted an osmotic pump delivering
0.25 mL/hour 5-FC (10 mg/mL) for 4 weeks in a first group
of mice, and i.v. injected them with five doses of MDA-
MB-231 cells transduced with the AdCD vector (two
doses per week, 3 10
5
cells per dose). At 10 days after
the last i.v. injection, the mice were killed and analyzed.
The macroscopic examination showed that the treated
mice had a significant smaller primary tumor volume than
the untreated group (received only surgery). Large
infiltrated lymph nodes, local and distant to the primary
tumor, could be seen in the animals of the control group
but were absent in the treated mice. Histological
examination of the lungs showed abundant large cancer
nodules in the untreated mice while the treated mice had
no visible nodules (Fig 2a). Moreover, we quantified the
amount of cancer in the organs prone to metastasis and
found that the treatment had diminished the metastatic
burden in the lungs and in kidneys when compared with
the control group (Fig 2b). We did not detect tumor cells
in the control mice that received only transduced tumor
cells i.v. and the implantation of the 5-FC osmotic pump
(not shown).
Ex vivo manipulated tumor cells carried an oncolytic
adenovirus to the metastases
Aiming to validate this experimental approach with a
different antitumor agent, the i.v. injected cancer cells
were infected with oncolytic adenoviruses. We used a
replication-competent adenovirus, AdwtFiGFP. Follow-
ing the sequence encoding the fiber (L5), this vector
contains an adenovirus IIIa splicing acceptor and the
GFP coding sequence (such an L6 unit). Therefore, GFP
is expressed as another late protein upon replication. We
expect this adenovirus to replicate in tumor cells, lyse
them and spread to the surrounding tumor cells. Viral
replication can be followed by detection of green
fluorescent cells. We used it as a proof of concept that
tumor cells may transport oncolytic viruses to the
metastases. In our model, the AdwtFiGFP cannot
replicate in murine cells but it does in the human tumor
Delivering antitumor therapies using tumor cells
J Garcı´a-Castro et al
3
Cancer Gene Therapy
cells. In preliminary in vitro experiments to assess the
bystander effects of this strategy, MDA-MB-231 and
PC3-infected cells were mixed with different proportions
of uninfected cells. The infection kinetic was followed by
periodical analyses under a fluorescence microscope. The
viability of the mixtures was determined after 6 days and
decreased when as low as 5% of the infected cells were
present in the culture (not shown). Longer culture times
ended up with a complete mortality of the mixtures. In a
set of mice with metastatic cancer, after surgical elimina-
tion of their mammary fat pad tumors (as above), we
injected i.v. cells carrying the AdwtFiGFP adenovirus.
The mice received five doses of infected cells (two doses
per week, 3 10
5
cells per dose) and 10 days after the last
dose were killed and their fresh organs examined under
fluorescent light (Fig 3a). We found nodules expressing
the GFP in the lung and liver of three out of seven mice,
and confirmed that the images corresponded to tumor
Figure 1 I.V. injected tumor cells localized on established metastases. (a) Lung cryosections stained with X-gal and the CAM5.2 antibody (anti-
human pancytokeratin) showed b-gal þ cells (i.v. injected) in or by established metastases. (b) Above: a low-power view showing the
hematoxilyn–eosin and CAM 5.2 antibody stain for the tumor nodule. Below: three different paraffin-embedded sections of the metastasis stained
with an anti-b-galactosidase antibody. Positive cells were detected at different levels within the tumor nodule.
Delivering antitumor therapies using tumor cells
J Garcı´a-Castro et al
4
Cancer Gene Therapy
nodules by RT-PCR of the human cytokeratin-19 gene.
Green nodules were not seen in control mice (received
surgical treatment) or mice injected only with the infected
cells (the AdwtFiGFP does not replicate in murine cells),
indicating that the green nodules corresponded to
metastases where AdwtFiGFP was actively replicating.
Immunohistochemical staining with an anti-GFP anti-
body revealed the presence of GFP-positive tumor cells in
the middle of GFP-negative micrometastatic nodules (Fig
S3). These results showed that i.v. injected tumor cells
initiated an oncolytic infection and that the lytic process
propagated to the surrounding tumor mass. This onco-
lytic infective process was active long after the last i.v.
injected cells had died (in vitro cell lysis happened within
96 hours), and resulted in a significant reduction in the
lung metastases of the treated mice compared to the
control mice (Fig 3b). Our results indicate that effective
targeting could be achieved with this delivery system and
virus replication in metastases was revealed by the
presence of GFP.
Figure 2 Ex vivo manipulated tumor cells exerted a bystander toxic effect on the metastases. (a) Hematoxilin–eosin colored sections of paraffin-
embedded lungs showed the presence of abundant metastases in the control mice but not in the treated ones. (b) Tumor volumes were
estimated by the formula V ¼ (p/6)a
2
b, where ‘‘a’’ was the short axis and ‘‘b’’ the long axis (left). Metastatic burden was quantitated as percentage
of human DNA by dot blot with a human-specific probe on DNA samples from the lungs and kidneys (right) (n ¼ 5 mice per group).
Delivering antitumor therapies using tumor cells
J Garcı´a-Castro et al
5
Cancer Gene Therapy
The therapeutical effect of i.v. injected tumor cells has
no detectable systemic toxicities
Microscopic examination of histological sections of the
organs from the treated mice did not show evidence that
the treatment had been toxic to the animals. We further
evaluated the toxicity associated to the treatment with i.v.
injected tumor cells by analyzing the renal and hepatic
functions. For this experiment, we did not generate any
Figure 3 Ex vivo manipulated tumor cells carried an oncolytic adenovirus to the metastases. (a) Upper: a cancer nodule in the lung surface of a
treated mouse is shown (left), and the same nodule is under fluorescent light (right). Bottom: a cancer nodule in the lung surface of a control
mouse (left). The same nodule under fluorescent light (right). (b) Lungs were weighed after killing. Weight increase was calculated in comparison
to healthy mice (n ¼ 7 mice per group).
Delivering antitumor therapies using tumor cells
J Garcı´a-Castro et al
6
Cancer Gene Therapy
metastatic cancer in the mice before the i.v. injections, but
mice received the same treatments as above. Biochemical
parameters of both renal (urea and creatinine) and
hepatic (transaminase enzymes, total proteins, bilirrubin
and gGT enzyme) functions were not affected by the
treatment (Table 1).
Discussion
We show here that i.v. injected tumor cells home in
established metastases. Our preliminary quantification
indicated that half of the b-gal þ cells in the lungs
localized in established metastases. Given that only 19%
of the total lung area were occupied by metastases, this
number suggests that tumor targeting was not a random
process. It has already been reported that intraperitone-
ally injected ovarian carcinoma cells preferentially loca-
lized at sites of locoregional metastatic ovarian cancer,
19
showing that injected cancer cells interacted with estab-
lished tumors. In addition to the lungs, we found b-gal þ
cells in different sections of a kidney metastasis (Fig 1).
Thus, localization cannot be solely explained by anato-
mical factors (route of injection and first pass effect). We
foresee three steps at which targeting may take place.
First, i.v. injected tumor cells may respond to the
chemokines involved in the preferential homing of
metastatic cells in organs.
16
Second, i.v. injected tumor
cells may attach to the vascular endothelium similarly to
intravascular micrometastases.
20
Last, i.v. injected tumor
cells should be able to participate in the crosstalk between
the cancer cells and factors from the host tissues that
governs the extravasation of the metastases.
21,22
Support-
ing this, in vitro experiments have shown that noninvasive
cancer cells can progress through three-dimensional
matrices following the track of invasive tumor cells,
which suggests that tumor cell migration and invasion
may be modulated by signals generated by other tumor
cells.
23
Infiltrating central nervous system tumors give rise
to signals, which can be read and followed by specific
neural cell types,
24
underscoring the role of tumor–host
signals for recruiting other cells into the metastases
environment. It is reasonable to assume that transducing
tumor cells with genes implicated in the metastatic process
would likely enhance the targeting efficacy.
Different cell types have been used as vector carriers for
systemic cancer gene therapies.
25
These cellular vehicles
act like shuttles for delivering vectors to the sites of tumor
growth much more efficiently than the vectors per se
regularly accomplish. Injection of oncolytic adenoviruses
to the circulation is followed by viral sequestration in the
liver.
8
The effective targeting of metastases when injecting
monoclonal antibodies is just 0.001–0.01% of the total
injected dose.
2
Therefore, there is a growing need of
vehicles that enhance the efficacy of new oncolytic viruses
or monoclonal antibodies.
2,10
Allogeneic tumor cells have
been tested as vehicles in locoregional tumors
19
but not as
systemic carriers. We present here two examples of how
ex vivo manipulated autologous tumor cells may deliver a
localized anticancer effect in vivo after systemic delivery.
Using autologous rather than allogeneic tumor cells will
prevent an immune rejection against the anticancer
cellular vehicles. In the case of AdCD-transduced cells,
locally produced 5-FU would be responsible for the
killing of the metastases, although a systemic effect of 5-
FU may also participate. The highest levels of the drug
would be reached at the site where it is produced, that is,
the injected cells. High levels of 5-FU from transduced
tumor cell should reach the metastases immediately after
being produced. The CD/5-FC system does not require
intercellular gap junctions, often lost in tumor cells,
26
for
bystander effect. This gives advantage over other suicide
gene therapy systems, such as the HSV-TK. Antitumor
immunity has been associated with the destruction of
tumor cells with the CD/5-FC gene therapy
27
and may
enhance the therapeutic benefits. As we used immunode-
ficient mice we could not evaluate this possibility. We
used the AdwtFiGFP adenovirus as a model of oncolytic
virus. Most of the advances on oncolytic adenoviruses
have been related to replication selectivity through
mutations or promoter insertions but a major hurdle
remains with regard to tumor targeting.
10
Our studies
focus on the delivery and detection of viral replication in
the targeted metastases and we used this kind of virus, a
replicating vector with no such mutations or promoter
insertions, as a proof of concept. Tumor cells present a
major advantage as vehicles for oncolytic viruses since
they are the primary cell types where the viruses replicate.
Candidate anticancer agents that would fit in our strategy
are suicide genes, antiangiogenic genes, oncolytic viruses,
monoclonal antibodies or drugs.
Safety is likely the most important concern when
considering new therapies. Our preliminary results about
Table 1 Biochemical evaluation of liver and kidney functions after treatment
ALT gGT Bilirubin Protein Creatinine Urea
(a) acute phase (10 days after last i.v. injection) and (b) chronic phase (50 days after last i.v. injection)
(a) Untreated 18.776 4.773 0.4470.1 4.7470.2 0.3170.05 42.771.1
AdCD treated 1477 5.374 0.3470.2 4.1270.6 0.370.02 38.372.9
(b) Untreated 13.471.3 4.471.9 0.570.2 570.4 0.370.02 4975.3
AdCD treated 9.576.6 7.271.9 0.470.2 4.770.3 0.470.05 39.374.2
ALT: alanine transaminase (U/L); gGT: gamma glutamil transferase (U/L); bilirubin (mg/dL); protein: (g/mL); creatinine: (mg/dL); urea:
(mg/dL).
Delivering antitumor therapies using tumor cells
J Garcı´a-Castro et al
7
Cancer Gene Therapy
systemic toxicities are encouraging, however, the potential
danger related to the infusion of live tumor cells back into
the patient needs to be addressed in further research. It is
well known that metastasis is a very inefficient pro-
cess,
12,22
even so, injected tumor cells may cause an
unwanted bystander effect on healthy cells, since targeting
is not totally specific. Risks can be greatly minimized
before infusionly optimizing the transduction efficiency
up to 100% as shown here, selecting the transduced cells
and inactivating the cells (lethal irradiation, chemicals).
Loading cancer cells with selective antitumor agents will
also enhance safety, that is, oncolytic viruses provide a
self-destructive mechanism in this strategy.
We consider our strategy as a treatment for the
metastatic disease rather than for the primary tumor,
even though we detected that i.v. injected tumor cells
localized in the primary tumor. Surgery of the primary
tumor is a standard first-line treatment for patients with
cancer, although it stimulates neoangiogenesis and
growth of metastases.
28
In this sense, our model resembles
the clinical situation of many patients with solid tumors.
The strategy shown is rationally different from the actual
anticancer therapies (surgery, chemoradiotherapies) and
they could be employed synergistically. We anticipate that
tumor cells may be loaded with different anticancer
agents, as unique or combined therapeutic elements, and
deliver its beneficial effect locally with less toxicity. In
summary, we show here that autologous tumor cells may
be used as a flexible tool for the treatment of the
metastatic disease. Further research is needed for addres-
sing crucial points before its use in patients. We have
designed experiments to rule out the ability of lethally
irradiated MDA-MB-231 cells to target metastatic le-
sions. Chasing experiments with a different tumor cell line
will help in clarifying the target specificity. In addition,
the experiments will be performed in immunocompetent
models with murine tumors in order to determine the role
of the immune system.
Acknowledgments
We thank Isabel de los Santos, Pilar Herna
´
ndez and
Sergio Garcı
´
a for technical assistance, and Dr Jose
´
M.
Martı
´
nez for his help with analyzing image files. This
work was supported in part by Fundacio
´
n Leucemia
Linfoma (MR) and Fundacio
´
n Oncohematologı
´
a Infantil
(JGC, LM and MR).
References
1. Rosenberg SA. Progress in human tumour immunology and
immunotherapy. Nature. 2001;411:380–384.
2. Carter P. Improving the efficacy of antibody-based cancer
therapies. Nat Rev Cancer. 2001;1:118–129.
3. Arap W, Pasqualini R, Ruoslahti E. Cancer treatment by
targeted drug delivery to tumor vasculature in a mouse
model. Science. 1998;279:377–380.
4. Hood JD, Bednarski M, Frausto R, et al. Tumor regression
by targeted gene delivery to the neovasculature. Science.
2002;296:2404–2407.
5. Lyden D, Hattori K, Dias S, et al. Impaired recruitment
of bone-marrow-derived endothelial and hematopoietic
precursor cells blocks tumor angiogenesis and growth.
Nat Med. 2001;7:1194–1201.
6. Sznol M, Lin SL, Bermudes D, et al. Use of preferentially
replicating bacteria for the treatment of cancer. J Clin
Invest. 2000;105:1027–1030.
7. Dang LH, Bettegowda C, Huso DL, et al. Combination
bacteriolytic therapy for the treatment of experimental
tumors. Proc Natl Acad Sci USA. 2001;98:15155–15160.
8. Alemany R, Balague C, Curiel DT. Replicative adenoviruses
for cancer therapy. Nat Biotechnol. 2000;18:723–727.
9. Bell JC, Garson KA, Lichty BD, et al. Oncolytic viruses:
programmable tumour hunters. Curr Gene Ther. 2002;2:
243–254.
10. Shayakhmetov DM, Li ZY, Ni S, et al. Targeting of
adenovirus vectors to tumor cells does not enable efficient
transduction of breast cancer metastases. Cancer Res.
2002;62:1063–1068.
11. Liotta LA, Kleinerman J, Saidel GM. Quantitative relation-
ships of intravascular tumor cells, tumor vessels, and
pulmonary metastases following tumor implantation.
Cancer Res. 1974;34:997–1004.
12. Fidler IJ. Metastasis: quantitative analysis of distribution
and fate of tumor embolilabeled with 125 I-5-iodo-2’-
deoxyuridine. J Natl Cancer Inst. 1970;45:773–782.
13. Nicolson GL. Organ specificity of tumor metastasis: role of
preferential adhesion, invasion and growth of malignant
cells at specific secondary sites. Cancer Metastasis Rev.
1988;7:143–188.
14. Liao F, Li Y, O’Connor W, et al. Monoclonal antibody to
vascular endothelial-cadherin is a potent inhibitor of
angiogenesis, tumor growth, and metastasis. Cancer Res.
2000;60:6805–6810.
15. Ruoslahti E, Giancotti FG. Integrins and tumor cell
dissemination. Cancer Cells. 1989;1:119–126.
16. Muller A, Homey B, Soto H, et al. Involvement of
chemokine receptors in breast cancer metastasis. Nature.
2001;410:50–56.
17. Felding-Habermann B, O’Toole TE, Smith JW, et al.
Integrin activation controls metastasis in human breast
cancer. Proc Natl Acad Sci USA. 2001;98:1853–1858.
18. Lillo R, Ramirez M, Alvarez A, et al. Efficient and non-
toxic adenoviral purging method for autologous transplan-
tation in breast cancer patients. Cancer Res. 2002;62:
5013–5018.
19. Coukos G, Makrigiannakis A, Kang EH, et al. Use of
carrier cells to deliver a replication-selective herpes simplex
virus-1 mutant for the intraperitoneal therapy of epithelial
ovarian cancer. Clin Cancer Res. 1999;5:1523–1537.
20. Al-Mehdi AB, Tozawa K, Fisher AB, et al. Intravascular
origin of metastasis from the proliferation of endothelium-
attached tumor cells: a new model for metastasis. Nat Med.
2000;6:100–102.
21. Liotta LA, Kohn EC. The microenvironment of the
tumour–host interface. Nature. 2001;411:375–379.
22. Cameron MD, Schmidt EE, Kerkvliet N, et al. Temporal
progression of metastasis in lung: cell survival, dormancy,
and location dependence of metastatic inefficiency. Cancer
Res. 2000;60:2541–2546.
23. Horino K, Kindezelskii AL, Elner VM, et al. Tumor cell
invasion of model 3-dimensional matrices: demonstration of
Delivering antitumor therapies using tumor cells
J Garcı´a-Castro et al
8
Cancer Gene Therapy
migratory pathways, collagen disruption, and intercellular
cooperation. FASEB J. 2001;15:932–939.
24. Aboody KS, Brown A, Rainov NG, et al. Neural stem cells
display extensive tropism for pathology in adult brain:
evidence from intracranial gliomas. Proc Natl Acad Sci
USA. 2000;97:12846–12851.
25. Harrington K, Alvarez-Vallina L, Crittenden M, et al. Cells
as vehicles for cancer gene therapy: the missing link between
targeted vectors and systemic delivery? Hum Gene Ther.
2002;13:1263–1280.
26. Holder JW, Elmore E, Barrett JC. Gap junction function
and cancer. Cancer Res. 1993;53:3475–3485.
27. Mullen CA, Coale MM, Lowe R, et al. Tumors expressing
the cytosine deaminase suicide gene can be eliminated in vivo
with 5-fluorocytosine and induce protective immunity to
wild type tumor. Cancer Res. 1994;54:1503–1506.
28. O’Reilly MS, Holmgren L, Shing Y, et al. Angiostatin: a
novel angiogenesis inhibitor that mediates the suppression
of metastases by a Lewis lung carcinoma. Cell. 1994;79:
315–328.
Supplementary information accompanies the paper on Cancer Gene Therapy website (http://www.nature.com/cgt).
Delivering antitumor therapies using tumor cells
J Garcı´a-Castro et al
9
Cancer Gene Therapy
... In the last two decades, several groups have exploited self-homing CTCs as "self-targeted" delivery vehicles for ex vivo loaded anti-cancer therapeutic cargo [30][31][32][33][34][35]. Cargo has included oncolytic viruses such as the H-1 parvovirus and vesicular stomatitis virus (VSV), prodrug converting enzyme genes including herpes simplex virus thymidine kinase (HSV-TK) and cytosine deaminase (CD), transgenes that target the tumor microenvironment such as tumor necrosis factor (TNF), and the secretory version of TNF-related apoptosisinducing ligand (S-TRAIL). ...
... Cargo has included oncolytic viruses such as the H-1 parvovirus and vesicular stomatitis virus (VSV), prodrug converting enzyme genes including herpes simplex virus thymidine kinase (HSV-TK) and cytosine deaminase (CD), transgenes that target the tumor microenvironment such as tumor necrosis factor (TNF), and the secretory version of TNF-related apoptosisinducing ligand (S-TRAIL). Additionally, a few groups have co-engineered the therapeutic CTCs and/or their viral cargo with optical or positron emission tomography (PET) imaging reporter genes to enable the fate of the cells/cargo to be noninvasively monitored with reporter gene imaging [31][32][33]35]. Importantly, while the ability to target, visualize, and treat singular pre-established subcutaneous tumors as well as orthotopic or metastatic lesions in a singular organ (e.g., lungs [31] or brain [35]) has been demonstrated, to the best of our knowledge, the ability of self-homing CTCs to migrate into and be used to visualize and treat spontaneous multi-organ metastatic disease is largely unknown. ...
... Self-homing has been largely attributed to an established tumor microenvironment that is considered highly permissive soil for the survival and growth of recruited CTCs [26]. Numerous studies have shown self-homing of systemically administered CTCs to an established primary tumor or singular metastatic lesion [27,[30][31][32][33][34][35]. Importantly, our study here demonstrates that CTCs administered intracardially are also efficient at homing to metastases that have spontaneously disseminated throughout the body (Figure 2). ...
Article
Full-text available
New ways to target and treat metastatic disease are urgently needed. Tumor “self-homing” describes the recruitment of circulating tumor cells (CTCs) back to a previously excised primary tumor location, contributing to tumor recurrence, as well as their migration to established metastatic lesions. Recently, self-homing CTCs have been exploited as delivery vehicles for anti-cancer therapeutics in preclinical primary tumor models. However, the ability of CTCs to self-home and treat metastatic disease is largely unknown. Methods: Here, we used bioluminescence imaging (BLI) to explore whether systemically administered CTCs home to metastatic lesions and if CTCs armed with both a reporter gene and a cytotoxic prodrug gene therapy can be used to visualize and treat metastatic disease. Results: BLI performed over time revealed a remarkable ability of CTCs to home to and treat tumors throughout the body. Excitingly, metastatic tumor burden in mice that received therapeutic CTCs was lower compared to mice receiving control CTCs. Conclusion: This study demonstrates the noteworthy ability of experimental CTCs to home to disseminated breast cancer lesions. Moreover, by incorporating a prodrug gene therapy system into our self-homing CTCs, we show exciting progress towards effective and targeted delivery of gene-based therapeutics to treat both primary and metastatic lesions.
... It is currently administered in weekly doses with no certainty as to the optimal time between doses or even the viral load required for cancer regression. 26,27,29,55,73 Regarding neuroblastoma, it plays a pivotal role in our study, signifying the pioneering onset of Celyvir application in pediatric cancer treatment. Since the innovative treatment began in October 2005 in children with neuroblastoma, simultaneously, numerous children were treated under compassionate use, with authorization from the Medicines Agency in Spain. ...
Article
Celyvir is an advanced therapy medicine, consisting of mesenchymal stem cells (MSCs) containing the oncolytic virus ICOVIR 5. This paper sets out a dynamic system which attempts to capture the fundamental relationships between cancer, the immune system and adenoviruses. Two forms of treatment were studied: continuous and periodic, the second being closer to the real situation. In the analysis of the first model, in addition to identifying the critical points, their properties and bifurcation points, a number of numerical simulations were carried out. It has thus been shown that there are bistability regimes in which Celyvir can produce an equilibrium of tumor progression, or even freedom from tumor. A sensitivity analysis was also performed to determine which parameters are most important in the system. Subsequently, an optimal control problem with nonlinear objective functional has been formulated, where the therapeutic goal is not only to minimize the size of the tumor cell population and the total cost of treatment, but also to prevent the tumor from reaching a critical size. It has been shown that the optimal control is bang–bang. With the second model, a threshold value of viral load has been identified at which the success of the treatment could be ensured. It is clear in both models that a low viral load would lead to relapse of the disease. Finally, it is shown that a periodic bang–bang regime should be used to optimize treatment with Celyvir.
... Although the best way to administer this treatment is unknown, it is currently administered in periodic doses, with no known optimal time between doses or viral load required for neuroblastoma regression. [9,10,11,12,13]. ...
Article
Full-text available
Celyvir viro-immunotherapy - a therapy using mesenchymal stem cells (MSCs) infected with the oncolytic adenovirus ICOVIR 5, an innovative oncolytic adenovirus - is a new therapeutic approach in cancer treatment. The purpose of this study is to create a mathematical model describing the relationships between immune, cancer and viral cells in pediatric patients with neuroblastoma. This was done by studying two different ways of applying the treatment: continuous and periodic therapy. Analysis of the first mathematical model identifies equilibrium points, their stability properties and bifurcation points. It can also identify bistability regimes in which therapy can induce either tumour-free equilibrium or tumour progression, depending only on the initial conditions. The second model reveals the existence of a threshold value of viral load, beyond which the patient’s recovery could be assured. The models make it clear that both the intensity of the viral load and the time over which treatment is given must be sufficient to ensure the success of the therapy. Low levels of treatment may fail to eliminate neuroblastoma, while stopping therapy early could lead to recurrence.
... Cancer cells themselves have been utilized as cellular vehicles, though largely in regional delivery studies. Tumor carrier cells were infected with oncolytic parvovirus and then inactivated through gamma irradiation, quite elegantly creating a microscopic "Trojan horse" capable of infecting tumors with oncolytic viruses [299,300], with the potential to localize to metastatic locations when introduced intravenously [301]. Tumorigenic cells are well characterized to affect the surrounding immune environments, including the potential to secrete immune cell recruitment chemokines [301]. ...
Article
Full-text available
While many classes of chemotherapeutic agents exist to treat solid tumors, few can generate a lasting response without substantial off-target toxicity despite significant scientific advancements and investments. In this review, the paths of development for nanoparticles, oncolytic viruses , and oncolytic bacteria over the last 20 years of research towards clinical translation and acceptance as novel cancer therapeutics are compared. Novel nanoparticle, oncolytic virus, and onco-lytic bacteria therapies all start with a common goal of accomplishing therapeutic drug activity or delivery to a specific site while avoiding off-target effects, with overlapping methodology between all three modalities. Indeed, the degree of overlap is substantial enough that breakthroughs in one therapeutic could have considerable implications on the progression of the other two. Each onco-therapeutic modality has accomplished clinical translation, successfully overcoming the potential pitfalls promising therapeutics face. However, once studies enter clinical trials, the data all but disappears , leaving pre-clinical researchers largely in the dark. Overall, the creativity, flexibility, and innovation of these modalities for solid tumor treatments are greatly encouraging, and usher in a new age of pharmaceutical development.
... In terms of early or late transgene expression, it was reported that insertions driven by the major late promoter (MLP) confer higher expression than E3 promoters [20]. The reported insertion sites in non-E3 deleted viruses driven by the MLP are downstream of L3 [20] and downstream of L5 (After-fiber), generating a new transcription unit, also known as L6 [14,16]. In this study, we compared the After-fiber insertion site (proximal site) against the After-E4 insertion site (distal site), which has been described only in E3-deleted viruses [18,19]. ...
Article
Full-text available
Oncolytic adenoviruses (OAds) present limited efficacy in clinics. The insertion of therapeutic transgenes into OAds genomes, known as “arming OAds”, has been the main strategy to improve their therapeutic potential. Different approaches were published in the decade of the 2000s, but with few comparisons. Most armed OAds have complete or partial E3 deletions, leading to a shorter half-life in vivo. We generated E3+ OAds using two insertion sites, After-fiber and After-E4, and two different splice acceptors linked to the major late promoter, either the Ad5 protein IIIa acceptor (IIIaSA) or the Ad40 long fiber acceptor (40SA). The highest transgene levels were obtained with the After-fiber location and 40SA. However, the set of codons of the transgene affected viral fitness, highlighting the relevance of transgene codon usage when arming OAds using the major late promoter.
Article
Full-text available
Lung cancer’s intractability is enhanced by its frequent resistance to (chemo)therapy and often high relapse rates that make it the leading cause of cancer death worldwide. Improvement of therapy efficacy is a crucial issue that might lead to a significant advance in the treatment of lung cancer. Oncolytic viruses are desirable combination partners in the developing field of cancer immunotherapy due to their direct cytotoxic effects and ability to elicit an immune response. Systemic oncolytic virus administration through intravenous injection should ideally lead to the highest efficacy in oncolytic activity. However, this is often hampered by the prevalence of host-specific, anti-viral immune responses. One way to achieve more efficient systemic oncolytic virus delivery is through better protection against neutralization by several components of the host immune system. Carrier cells, which can even have innate tumor tropism, have shown their appropriateness as effective vehicles for systemic oncolytic virus infection through circumventing restrictive features of the immune system and can warrant oncolytic virus delivery to tumors. In this overview, we summarize promising results from studies in which carrier cells have shown their usefulness for improved systemic oncolytic virus delivery and better oncolytic virus therapy against lung cancer.
Article
This work presents oncolytic adenovirus (OA)-loaded Janus tumor cell-based microrobots fabricated by asymmetrically coating OA-infected tumor cells with Fe 3 O 4 nanoparticles (NPs), capable of homologous adhesion, active virotherapy, and magnetic navigation in complex topography. The cellular resources allow the infected OA to replicate inside the host cell, further inducing cytolysis to release the viral progeny for persistent and specific tumor infection and killing. The resulting cell robots preserve the intrinsic feature of tumor cells with cognate adhesion to homologous cancerous cells. The effective magnetic propulsion coupling with homologous binding of Janus cell robots greatly enhance their binding and penetration to three-dimensional (3D) homologous tumor cell spheroid with improved cell-killing efficacy. Moreover, cell robots can be navigated across the confined space and migrate from the bladder to the renal area through the ureter in the excised rabbit urinary system upon magnetic manipulation. Imparting tumor cells with magnetic propulsion force and virus cargos generates a motile therapeutic platform with the capability of tumor-specific killing and binding, and robust and steerable motion, holding great potential to cross the inherent barriers inside living body and achieve efficient drug delivery toward hard-to-reach sites upon minimized invasion and toxicity to healthy tissues.
Article
Efficacy of oncolytic, conditionally-replicating adenovirus (CRAd) vectors can be enhanced by “arming” the vector with therapeutic transgenes. We examined whether inclusion of an intact early region 3 (E3) and the reptilian reovirus fusogenic p14 fusion-associated small transmembrane (FAST) protein enhanced vector efficacy. The p14 FAST transgene was cloned between the fiber gene and E4 region, with an upstream splice acceptor for replication-dependent expression from the major late promoter. In A549 cells, this vector expressed p14 FAST protein at very low levels, and showed a poor ability to mediate cell-cell fusion, relative to a similar vector encoding p14 FAST within the E3 deletion. Although expression of E3 proteins from the CRAd increased plaque size, poor expression of p14 FAST protein compromised the fusogenic capacity of the vector. Thus, location of a therapeutic transgene within a CRAd can significantly impact expression of the transgene and is an important consideration in vector design.
Article
The incidence of hematological malignancies such as multiple myeloma, leukemia, and lymphoma has increased over time. Although bone marrow transplantation, immunotherapy and chemotherapy have led to significant improvements in efficacy, poor prognosis in elderly patients, recurrence and high mortality among hematological malignancies remain major challenges, and innovative therapeutic strategies should be explored. Besides directly lyse tumor cells, oncolytic viruses can activate immune responses or be engineered to express therapeutic factors to increase antitumor efficacy, and have gradually been recognized as an appealing approach for fighting cancers. An increasing number of studies have applied oncolytic viruses in hematological malignancies and made progress. In particular, strategies combining immunotherapy and oncolytic virotherapy are emerging. Various phase I clinical trials of oncolytic reovirus with lenalidomide or programmed death 1(PD-1) immune checkpoint inhibitors in multiple myeloma are ongoing. Moreover, preclinical studies of combinations with chimeric antigen receptor T (CAR-T) cells are underway. Thus, oncolytic virotherapy is expected to be a promising approach to cure hematological malignancies. This review summarizes progress in oncolytic virus research in hematological malignancies. After briefly reviewing the development and oncolytic mechanism of oncolytic viruses, we focus on delivery methods of oncolytic viruses, especially systemic delivery that is suitable for hematological tumors. We then discuss the main types of oncolytic viruses applied for hematological malignancies and related clinical trials. In addition, we present several ways to improve the antitumor efficacy of oncolytic viruses. Finally, we discuss current challenges and provide suggestions for future studies.
Article
Due to their innate tumour homing capabilities, in recent years, circulating tumour cells (CTCs) have been engineered to express therapeutic genes for targeted treatment of primary and metastatic lesions. Additionally, previous studies have incorporated optical or PET imaging reporter genes to enable noninvasive monitoring of therapeutic CTCs in preclinical tumour models. An alternative method for tracking cells is to pre-label them with imaging probes prior to transplantation into the body. This is typically more sensitive to low numbers of cells since large amounts of probe can be concentrated in each cell. The objective of this work was to evaluate magnetic particle imaging (MPI) for the detection of iron-labeled experimental CTCs. CTCs were labeled with micro-sized iron oxide (MPIO) particles, administered via intra-cardiac injection in tumour bearing mice and were detected in the tumour region of the mammary fat pad. Iron content and tumour volumes were calculated. Ex vivo MPI of the tumours and immunohistochemistry were used to validate the imaging data. Here, we demonstrate for the first time the ability of MPI to sensitively detect systemically administered iron-labeled CTCs and to visualize tumour self-homing in a murine model of human breast cancer.
Article
Full-text available
The role of bone marrow (BM)-derived precursor cells in tumor angiogenesis is not known. We demonstrate here that tumor angiogenesis is associated with recruitment of hematopoietic and circulating endothelial precursor cells (CEPs). We used the angiogenic defective, tumor resistant Id-mutant mice to show that transplantation of wild-type BM or vascular endothelial growth factor (VEGF)-mobilized stem cells restore tumor angiogenesis and growth. We detected donor-derived CEPs throughout the neovessels of tumors and Matrigel-plugs in an Id1 +/- Id3 -/- host, which were associated with VEGF-receptor-1−positive (VEGFR1+) myeloid cells. The angiogenic defect in Id-mutant mice was due to impaired VEGF-driven mobilization of VEGFR2+ CEPs and impaired proliferation and incorporation of VEGFR1+ cells. Although targeting of either VEGFR1 or VEGFR2 alone partially blocks the growth of tumors, inhibition of both VEGFR1 and VEGFR2 was necessary to completely ablate tumor growth. These data demonstrate that recruitment of VEGF-responsive BM-derived precursors is necessary and sufficient for tumor angiogenesis and suggest new clinical strategies to block tumor growth.
Article
Full-text available
Metastasis is a frequent complication of cancer, yet the process through which circulating tumor cells form distant colonies is poorly understood. We have been able to observe the steps in early hematogenous metastasis by epifluorescence microscopy of tumor cells expressing green fluorescent protein in subpleural microvessels in intact, perfused mouse and rat lungs. Metastatic tumor cells attached to the endothelia of pulmonary pre-capillary arterioles and capillaries. Extravasation of tumor cells was rare, and it seemed that the transmigrated cells were cleared quickly by the lung, leaving only the endothelium-attached cells as the seeds of secondary tumors. Early colonies were entirely within the blood vessels. Although most models of metastasis include an extravasation step early in the process, here we show that in the lung, metastasis is initiated by attachment of tumor cells to the vascular endothelium and that hematogenous metastasis originates from the proliferation of attached intravascular tumor cells rather than from extravasated ones. Intravascular metastasis formation would make early colonies especially vulnerable to intravascular drugs, and this possibility has potential for the prevention of tumor cell attachment to the endothelium.
Article
Full-text available
Metastasis is the primary cause of death in human breast cancer. Metastasis to bone, lungs, liver, and brain involves dissemination of breast cancer cells via the bloodstream and requires adhesion within the vasculature. Blood cell adhesion within the vasculature depends on integrins, a family of transmembrane adhesion receptors, and is regulated by integrin activation. Here we show that integrin αvβ3 supports breast cancer cell attachment under blood flow conditions in an activation-dependent manner. Integrin αvβ3 was found in two distinct functional states in human breast cancer cells. The activated, but not the nonactivated, state supported tumor cell arrest during blood flow through interaction with platelets. Importantly, activated αvβ3 was expressed by freshly isolated metastatic human breast cancer cells and variants of the MDA-MB 435 human breast cancer cell line, derived from mammary fat pad tumors or distant metastases in severe combined immunodeficient mice. Expression of constitutively activated mutant αvβ3D723R, but not αvβ3WT, in MDA-MB 435 cells strongly promoted metastasis in the mouse model. Thus breast cancer cells can exhibit a platelet-interactive and metastatic phenotype that is controlled by the activation of integrin αvβ3. Consequently, alterations within tumors that lead to the aberrant control of integrin activation are expected to adversely affect the course of human breast cancer.
Article
Full-text available
The locations of distant secondary tumors in many clinical cancers and animal tumors are nonrandom, and their distributions cannot be explained by simple anatomical or mechanical hypotheses based on the simple lodgment or trapping of tumor cell emboli in the first capillary bed encountered. Evidence from certain experimental tumor systems supports Paget's 'seed and soil' hypothesis on the nonrandom distributions of metastases, in which the unique properties of particular tumor cells ('seeds') and the different characteristics of each organ microenvironment ('soil') collectively determine the organ preference of metastasis. Experimentally, differential tumor cell adhesion to organ-derived microvessel endothelial cells and organ parenchymal cells, differential invasion of basement membranes and organ tissues, and differential responses to organ-derived growth-stimulatory and -inhibitory factors all appear to be important determinants in explaining the organ preference of metastasis. Each tumor system may achieve organ specificity because of its own unique set of multiple metastasis-associated properties and responses to host microenvironments. As neoplasms progress to more highly malignant states multisite metastases are more likely and organ-specific metastases may be masked or circumvented owing to stochastic events, tumor cell diversification, host selection processes, and increased production of tumor autocrine molecules that may modulate adhesion, invasion, growth, and other properties important in metastasis. The importance of each of these properties, however, appears to vary considerably among different metastatic tumor systems. These and other tumor cell and host properties may eventually be used to predict and explain the unique metastatic distributions of certain human malignancies.
Article
The phenomenon of inhibition of tumor growth by tumor mass has been repeatedly studied, but without elucidation of a satisfactory mechanism. In our animal model, a primary tumor inhibits its remote metastases. After tumor removal, metastases neovascularize and grow. When the primary tumor is present, metastatic growth is suppressed by a circulating angiogenesis inhibitor. Serum and urine from tumor-bearing mice, but not from controls, specifically inhibit endothelial cell proliferation. The activity copurifies with a 38 kDa plasminogen fragment that we have sequenced and named angiostatin. A corresponding fragment of human plasminogen has similar activity. Systemic administration of angiostatin, but not intact plasminogen, potently blocks neovascularization and growth of metastases. We here show that the inhibition of metastases by a primary mouse tumor is mediated, at least in part, by angiostatin.
Article
We report a novel 3-dimensional model for visualizing tumor cell migration across a nylon mesh-supported gelatin matrix. To visualize migration across these model barriers, cell proteolytic activity of the pericellular matrix was detected using Bodipy-BSA (fluorescent upon proteolysis) and DQ™ collagen (fluorescent upon collagenase activity). For 3-dimensional image reconstruction, multiple optical images at sequential z axis positions were deconvoluted by computer analysis. Specificity was indicated using well-known inhibitors. Using these fluorescent proteolysis markers and imaging methods, we have directly demonstrated proteolytic and collagenolytic activity during tumor cell invasion. Moreover, it is possible to visualize migratory pathways followed by tumor cells during matrix invasion. Using cells of differing invasive potentials (uPAR-negative T-47D wild-type and uPAR-positive T-47D A2–1 cells), we show that the presence of the T-47D-A2–1 cells facilitates the entry of T-47D wild-type cells into the matrix. In some cases, wild-type cells follow T-47D A2–1 cells into the matrix whereas other T-47D-wild-type cells appear to enter without the direct intervention of T-47D A2–1 cells. Thus, we have developed a new 3-dimensional model of tumor cell invasion, demonstrated protein and collagen disruption, mapped the pathways followed by tumor cells during migration through an extracellular matrix, and illustrated cross-talk among tumor cell populations during invasion.—Horino, K., Kindezelskii, A. L., Elner, V. M., Hughes, B. A., Petty, H. R. Tumor cell invasion of model 3-dimensional matrices: demonstration of migratory pathways, collagen disruption, and intercellular cooperation.
Article
The delineation of the genetic etiology of cancer makes gene therapy a rational approach for the molecular treatment of cancer. Many gene delivery systems have been developed, with viral vectors being the most effective. Underlying cancer gene therapy protocols is the recognition that quantitative tumor transduction cannot be achieved with the vector systems available at the present time. One way to overcome this problem could be to amplify the transduction efficiency through the use of vectors capable of replicating specifically in tumor cells. We are currently developing an adenoviral vector in which viral replication will be restricted to the target tumor cells by limiting the expression of viral genes essential for the virus replication only to the tumor cells of interest.
Article
The ability of a tumor cell to metastasize is determined, in part, by its adhesive interactions with other cells and with components of the extracellular matrix. An ever-expanding family of cell surface receptors, the integrins, is an essential player in these interactions. Research efforts aimed at understanding the precise role of individual integrins in normal and transformed cells may offer a general framework for future therapeutic approaches. Here we review recent progress toward this goal.
Article
SUMMARY An experimental model has been developed to quantify some of the major processes initiated by tumor transplanta tion and culminating in pulmonary métastases. The T241 fibrosarcoma, chosen because of its high hematogenous metastatic propensity and reproducible biological behavior, is transplanted into the femoral region of the C57BL mouse. Experiments are performed at specified times after trans plantation to determine the dynamics of tumor growth, density, and size distribution of perfused tumor vessels, entry rate of tumor cells into the circulation, and the number of pulmonary métastases. Estimates of the entry rate of tumor cells into the circulation are obtained by perfusing tumors with an oxygenated, cell-free medium to allow the counting of single tumor cells and tumor cell clumps collected in the venous effluent. The tumor vascular network appears at approximately Day 4, while tumor cells are first observed in the perfusate approximately 5 days after transplantation. The concentration of effluent tumor cells, singly and in clumps, increases rapidly until Day 10, after which the rate of cell entry into the perfusion is diminished. A linear relation is found between the density of perfused vessels and the concentration of effluent tumor cells. Métastases, first observed on Day 10, increase rapidly with time, at a rate similar to that of the concentration of effluent tumor cell clumps containing 4 or more cells. These studies suggest that dynamics of hematogenously initiated métastases depend strongly on the entry rate of tumor cell clumps into the circulation, which in turn is intimately linked to tumor vascularization.