ArticlePDF Available

An orthotopic metastatic prostate cancer model in SCID mice via grafting of a transplantable human prostate tumor line

Authors:

Abstract and Figures

Metastasis is the major cause of prostate cancer deaths and there is a need for clinically relevant in vivo models allowing elucidation of molecular and cellular mechanisms underlying metastatic behavior. Here we describe the development of a new in vivo model system for metastatic prostate cancer. Pieces of prostate cancer tissue from a patient were grafted in testosterone-supplemented male NOD-SCID mice at the subrenal capsule graft site permitting high tumor take rates. After five serial transplantations, the tumor tissues were grafted into mouse prostates. Resulting tumors and suspected metastatic lesions were subjected to histopathological and immunohistochemical analysis. Samples of metastatic tissue were regrafted in mouse anterior prostates and their growth and spread examined, leading to isolation from lymph nodes of a metastatic subline, PCa1-met. Orthotopic grafting of PCa1-met tissue in 47 hosts led in all cases to metastases to multiple organs (lymph nodes, lung, liver, kidney, spleen and, notably, bone). Histopathological analysis showed strong similarity between orthotopic grafts and their metastases. The latter were of human origin as indicated by immunostaining using antibodies against human mitochondria, androgen receptor, prostate-specific antigen and Ki-67. Spectral karyotyping showed few chromosomal alterations in the PCa1-met subline. This study indicates that transplantable subrenal capsule xenografts of human prostate cancer tissue in NOD-SCID mice can, as distinct from primary cancer tissue, be successfully grown in the orthotopic site. Orthotopic xenografts of the transplantable tumor lines and metastatic sublines can be used for studying various aspects of metastatic prostate cancer, including metastasis to bone.
Content may be subject to copyright.
An orthotopic metastatic prostate cancer
model in SCID mice via grafting of a
transplantable human prostate tumor line
Yuwei Wang
1
, Hui Xue
1
, Jean-Claude Cutz
1,2
, Jane Bayani
3
, Nasrin R Mawji
4
,
Wilfred G Chen
5
, Lester J Goetz
6
, Simon W Hayward
7
, Marianne D Sadar
4
,
C Blake Gilks
8
, Peter W Gout
1
, Jeremy A Squire
3
, Gerald R Cunha
9
and Yu-Zhuo Wang
1
1
Department of Cancer Endocrinology, BC Cancer Agency, Vancouver, British Columbia, Canada;
2
Department of Anatomical Pathology, St Joseph’s Hospital—Hamilton Health Sciences, Hamilton, Ontario,
Canada;
3
Department of Laboratory Medicine and Pathobiology, Ontario Cancer Institute, Toronto, Ontario,
Canada;
4
Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada;
5
Urology Research Unit, Carlton Centre, San Fernando, Trinidad;
6
Department of Urology, Gulf View Medical,
La Romaine, Trinidad;
7
Departments of Urologic Surgery and Cancer Biology, Vanderbilt University Medical
Center, Nashville, TN, USA;
8
Department of Pathology, Vancouver General Hospital, University of British
Columbia, Vancouver, British Columbia, Canada and
9
Departments of Anatomy and Urology, University
of California San Francisco, San Francisco, CA, USA
Metastasis is the major cause of prostate cancer deaths and there is a need for clinically relevant in vivo models
allowing elucidation of molecular and cellular mechanisms underlying metastatic behavior. Here we describe
the development of a new in vivo model system for metastatic prostate cancer. Pieces of prostate cancer tissue
from a patient were grafted in testosterone-supplemented male NOD-SCID mice at the subrenal capsule graft
site permitting high tumor take rates. After five serial transplantations, the tumor tissues were grafted into
mouse prostates. Resulting tumors and suspected metastatic lesions were subjected to histopathological and
immunohistochemical analysis. Samples of metastatic tissue were regrafted in mouse anterior prostates and
their growth and spread examined, leading to isolation from lymph nodes of a metastatic subline, PCa1-met.
Orthotopic grafting of PCa1-met tissue in 47 hosts led in all cases to metastases to multiple organs (lymph
nodes, lung, liver, kidney, spleen and, notably, bone). Histopathological analysis showed strong similarity
between orthotopic grafts and their metastases. The latter were of human origin as indicated by
immunostaining using antibodies against human mitochondria, androgen receptor, prostate-specific antigen
and Ki-67. Spectral karyotyping showed few chromosomal alterations in the PCa1-met subline. This study
indicates that transplantable subrenal capsule xenografts of human prostate cancer tissue in NOD-SCID mice
can, as distinct from primary cancer tissue, be successfully grown in the orthotopic site. Orthotopic xenografts
of the transplantable tumor lines and metastatic sublines can be used for studying various aspects of
metastatic prostate cancer, including metastasis to bone.
Laboratory Investigation (2005) 85, 1392–1404. doi:10.1038/labinvest.3700335; published online 12 September 2005
Keywords:
human prostate cancer; metastatic model; orthotopic; xenograft; subrenal capsule
Metastatic prostate cancers have a poor prognosis
with limited responses to palliative therapies such
as androgen ablation and chemotherapy.
1,2
The
development of new, effective therapies for meta-
static prostate cancer is considered to be critically
dependent on the availability of clinically relevant
in vivo models.
3
Current models used for studying
prostate cancer metastasis and drug evaluation
generally consist of xenografts in immuno-deficient
mice of well-established human prostate cancer cell
lines that have adapted to in vitro growth, for
example, LNCaP and PC-3.
4–6
Such models have
been useful for identifying cellular and molecular
mechanisms underlying metastasis and develop-
ment of new therapeutics. However, they have
profound shortcomings: the highly anaplastic
Received 3 March 2005; revised 8 July 2005; accepted 11 July
2005; published online 12 September 2005
Correspondence: Dr Y-Z Wang, PhD, Department of Cancer
Endocrinology, BC Cancer Agency—Research Centre, 675 West
10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3.
E-mail: ywang@bccrc.ca
Laboratory Investigation (2005) 85, 1392 1404
&
2005 USCAP, Inc All rights reserved 0023-6837/05
$30.00
www.laboratoryinvestigation.org
prostate cancer cells used represent the extreme end
of highly advanced cancers and are not associated
with original tumor stroma, now recognized as a
crucial factor in the pathogenesis of cancer meta-
stasis.
4,5
These limitations severely restrict the
predictive power of such models with regard to
responses of patients’ tumors to anticancer drugs in
the clinic.
6
In view of this, various groups have
embarked on developing more relevant models
based on xenografting of primary prostate cancer
tissue in immuno-deficient mice, mainly using
advanced tumors and the subcutaneous graft
site.
3,7–11
These xenograft models retain growth and
histopathological features characteristic of the ori-
ginal cancers and have been used for rapid screening
of potential therapeutics. Unfortunately, xenograft-
ing to the subcutaneous site was associated with
exceedingly low tumor take rates and only success-
ful in case of highly advanced malignancies.
Furthermore, the subcutaneous compartment is a
nonorthotopic graft site. It does not mimic the
original tumor microenvironment, nor does it allow
full expression of metastatic potential. These defi-
ciencies reduce the usefulness of such models for
studies of prostate cancer metastasis.
12
Recently, we have developed a procedure for
successfully grafting and serially transplanting
primary human prostate cancer tissues in SCID
mice.
13
Using the subrenal capsule graft site, we
have consistently achieved high tumor take rates
(495%). This was a consequence of utilizing: (i)
SCID mouse hosts, (ii) adjustment of the hormonal
status of the host, if required and (iii) meticulous
surgical technique. In the present study, we found
that transplants of subrenal capsule xenografts of
human prostate cancer tissue, as distinct from
primary tumor tissue, were able to grow in the
orthotopic site (prostate) of SCID mice. The meta-
static behavior of the grafts at this site led to
isolation of a transplantable, metastatic tumor sub-
line, designated PCa1-met. Orthotopic xenografts in
SCID mice of the transplantable tumor and its
metastatic subline led to tumors showing histo-
pathological, tissue invasive and metastatic features,
which are similar to those normally encountered in
the clinic. Such xenografts could therefore be useful
as orthotopic models for human metastatic prostate
cancer.
Materials and methods
Materials and Animals
Chemicals, stains, solvents and solutions were
obtained from Sigma-Aldrich Canada Ltd, Oakville,
ON, Canada, unless otherwise indicated. Male 6- to
8-week old nonobese diabetic, severe combined
immuno-deficient (NOD-SCID) mice were bred by
the BC Cancer Research Centre Animal Resource
Centre, BC Cancer Agency, Vancouver, Canada.
Cell Cultures
The human prostate cancer cell line, LNCaP, was
obtained from the American Type Culture Collection
(Manassas, VA, USA). Cells were maintained in
RPMI-1640 medium (Stem Cell Technologies, Van-
couver, BC, Canada), supplemented with 10% fetal
bovine serum (Gibco-BRL, Burlington, ON, Canada),
penicillin (50 units/ml) and streptomycin (50 mg/ml)
(Stem Cell Technologies) in a humidified atmo-
sphere of 95% air and 5% CO
2
at 371C. Subculturing
was carried out using standard techniques, includ-
ing trypsinization using 0.25% trypsin/1.0 mM
EDTA.
Prostate Cancer Tissue Acquisition
Prostate cancer tissue was obtained via prostatect-
omy from a 75-year-old male, with informed
consent, at the Urology Research Unit, Carlton
Centre, San Fernando, Trinidad. The patient, diag-
nosed with advanced prostate cancer, had not
received neoadjuvant therapy prior to prostatect-
omy. The specimens were examined, sectioned and
selected by pathologists for histological analysis and
xenografting. The tumor sections selected for graft-
ing were shipped overnight, immersed in cold
Hanks’ balanced salt solution supplemented with
antibiotics to Vancouver, Canada.
Subrenal Capsule Grafting and Development of
Transplantable Tumor Lines
Within 24 h of its arrival, a minor portion of the
tumor was fixed for histological analysis. The
remainder of the tumor was cut into approximately
300 pieces with each piece about 1 3 3mm
3
in
size. A total of 70 NOD-SCID mice were supple-
mented with testosterone (25 mg) via subcuta-
neously implanted testosterone pellets.
14,15
The
tumor pieces were grafted to the subrenal capsule
sites of the mice. Under sterile conditions, a skin
incision of approximately 2 cm was made along the
dorsal midline of an anesthetized mouse. With the
animal lying on its side, an incision was then made
in the body wall slightly shorter than the long axis of
the kidney. The kidney was slipped out of the body
cavity by applying pressure on the other side of the
organ using forefinger and thumb. After exterioriza-
tion of the kidney, #5 fine forceps were used to
gently pinch and lift the capsule from the renal
parenchyma to allow a 2–4 mm incision in the
capsule using fine spring-loaded scissors. A pocket
between the kidney capsule and the parenchyma
was then created by blunt dissection. Care was taken
not to damage the parenchyma and thus prevent
bleeding. The graft was transferred to the surface of
the kidney using blunt-ended forceps. The cut edge
of the renal capsule was lifted with fine forceps, and
the graft was inserted into the pocket under the
A new orthotopic prostate cancer model
Y Wang et al
1393
Laboratory Investigation (2005) 85, 13921404
capsule using a fire-polished glass pipette. Two or
three grafts per kidney could be placed under the
renal capsule. The kidney was then gently eased
back into the body cavity and the body wall and skin
incisions sutured. Mice were housed in groups of
three in microisolators with free access to food and
water and their health was monitored daily. Animal
care and experiments were carried out in accor-
dance with the guidelines of the Canadian Council
on Animal Care.
After 60 or 90 days of growth (or earlier if required
by the health status of the hosts), the animals were
killed in a CO
2
chamber for necropsy. Tumors were
harvested, measured, photographed and fixed for
histopathological analysis. Some of the rapidly
growing tumors were maintained for up to eight
transplant generations by serial subrenal capsule
transplantation into testosterone-supplemented
male NOD-SCID mice. One of these transplantable
lines, PCa1, was used for the study presented here.
Orthotopic Grafting and Isolation of a Metastatic
Tumor Subline, PCa1-met
Tumor tissue of the transplantable PCa1 line was
harvested from a kidney and cut into pieces of about
2mm
3
. These were grafted into the anterior prostates
of 12 male, testosterone-supplemented NOD-SCID
mice (two per mouse). Briefly, a transverse incision
was made in the lower abdomen and the bladder,
seminal vesicles and prostate were partially pulled
out of the abdominal cavity to expose the anterior
prostate. A 2–3 mm incision was made through the
capsule of the anterior prostate between the two
main ducts. A 2 mm
3
PCa1 tumor graft was inserted
into the pocket created under the prostatic capsule.
The exteriorized organs were then returned into the
body cavity, and the incisions in the body wall and
skin closed using a running suture of 4-0 silk. After
6 weeks of growth, the host mice were killed for
gross examination of lymph nodes. Swollen lymph
nodes were bisected, and one-half subjected to
histological examination as frozen sections. Con-
firmed lymph node metastases were designated
‘PCa1-met’. Pieces of fresh lymph node containing
metastatic deposits were transplanted into prostates
of new hosts and the emerging PCa1-met line was
subsequently maintained by serial transplantation
both in prostates and under renal capsules of NOD-
SCID mice. The procedure for selecting PCa1-met is
illustrated in Figure 1.
Development of Multiorgan Metastases
PCa1-met tissue, freshly isolated from subrenal
capsule sites of NOD-SCID mice, was regrafted into
anterior prostates of 47 male, testosterone-supple-
mented NOD-SCID mice (divided into five groups).
At intervals of 2, 3, 4, 6 and 8 weeks after grafting,
mice were killed and lymph nodes, lungs, livers,
kidneys, spleens and bone (femur) were fixed for
histological and immunohistochemical (IHC) exam-
ination or frozen in liquid nitrogen for RT-PCR
analysis.
Histological and IHC Staining
The original tumor specimen, its transplants and
metastases were fixed in 10% neutral-buffered
formalin and embedded in paraffin; for bone
metastases, specimens were treated, after fixing,
with a sterile decalcification solution (15% EDTA/
0.5% paraformaldehyde in PBS, pH 8.0) for 3 weeks
prior to paraffin embedding. Serial sections (5 mm
thick) were cut on a microtome and mounted on
glass slides. Approximately 80 sections were cut
from each paraffin block. For histopathological
examination, every fourth section was de-waxed in
Histoclear (National Diagnostic, Atlanta, GA, USA)
and hydrated in graded alcohol solutions and
distilled water for H&E staining and examined
under a light microscope. For IHC staining, endo-
genous peroxidase activity was blocked with 0.5%
hydrogen peroxide in methanol for 30 min followed
by washing in PBS pH 7.4. In all, 5% normal goat or
donkey serum in PBS was applied to the sections for
30 min to block nonspecific sites. The sections were
then incubated with primary antibodies overnight at
41C or with control IgG from nonimmunized mice.
Rabbit polyclonal anti-androgen receptor (AR) anti-
bodies (PA1-111A) were purchased from Affinity
BioReagents (Golden, CO, USA) and mouse anti-
human mitochondria monoclonal antibodies (MAB-
1273) from Chemicon International (Temecula, CA,
USA). An anti-CK8 mouse monoclonal antibody
preparation (LE41) was generously provided by
Dr EB Lane, University of Dundee, UK. Polyclonal
rabbit antibodies against human prostate specific
antigen (PSA) and mouse anti-human Ki-67 anti-
bodies (m7240) were purchased from Dako (Carpin-
teria, CA, USA); these two antibodies reacted
with human, but not with mouse tissues. Purified
rabbit and mouse IgGs were obtained from Zymed
Corp. (So. San Francisco, CA, USA). Biotinylated
anti-rabbit and anti-mouse IgGs were obtained
from Amersham International (Arlington Heights,
IL, USA). Biotinylated anti-goat antibodies were
purchased from Sigma. Peroxidase-linked avidin/
biotin complex reagents were obtained from Vector
Laboratories (Burlingame, CA, USA). Following
incubation with the primary antibodies, sections
were washed with PBS and incubated for 30 min at
room temperature with the appropriate biotinylated
secondary anti-mouse immunoglobulins diluted
with PBS 1:200. After incubation with the secondary
antibodies, sections were washed in PBS (three
10-min washes), and then incubated with avidin–
biotin complex (Vector Laboratories, Foster City, CA,
USA) for 30 min at room temperature. Following a
further 30 min of washing in PBS, immunoreactivity
was visualized using 3,3
0
-diaminobenzidine tetra-
A new orthotopic prostate cancer model
Y Wang et al
1394
Laboratory Investigation (2005) 85, 13921404
hydrochloride (DAB) in PBS and 0.03% hydrogen
peroxide. Sections were counterstained with hema-
toxylin and dehydrated in graded alcohols. Control
sections were processed in parallel with mouse or
rabbit nonimmune IgG (Dako) used at the same
concentrations as the primary antibodies.
RT-PCR
Total RNA was isolated from tumor tissue using
TRIZOL reagent (Invitrogen, Carlsbad, CA, USA)
according to the manufacturer’s instructions. A 1 mg
quantity was used for one-step RT-PCR (Invitrogen)
with the following thermal cycling conditions: one
cycle at 501C for 30 min, 941C for 2 min with an
additional 29 cycles for both GAPDH and mouse b2-
microglobulin at 941C for 15 s, 621C for 30 s and 721C
for 1 min and a final extension at 721C for 5 min. For
the nested PSA RT-PCR, 1 mg of total RNA was
subjected to one-step RT-PCR (Invitrogen) using the
external primers under the following conditions:
one cycle at 501C for 30 min, at 941C for 2 min and 25
cycles at 941C for 15 s, at 621C for 30 s and 721C for
1 min and a final extension at 721C for 5 min. In all,
1 ml from this PCR product was subjected to a second
PCR run using the internal primers with the
following conditions: 941C for 2 min and 25 cycles
at 941C for 15 s, 621C for 30 s and 721C for 1 min and
a final extension at 721C for 5 min. Sequences of the
primers used are as follows. External primers for
PSA: sense-GAT GAC TCC AGC CAC GAC CT,
antisense-CAC AGA CAC CCC ATC CTA TC; inner
primers for PSA: sense-GCA AGT TCA CCC TCA
GAA GG, antisense-GAT ATG TCT CCA GGC ATG
GC; primers for human GAPDH: sense-CCG AGC
CAC ATC GCT CAG A, antisense-CCC AGC CTT
CTC CAT GGT G; primers for mouse b2-microglo-
bulin: sense-CAC GCC ACC CAC CGC AGA ATG
GGA AGC C, antisense-CTG CGT GCA TAA ATT
GTA TAG CAT ATT AG.
Western Blot Analysis
The mouse and xenograft tissues were ground up
with the aid of mortar and pestle using liquid
nitrogen and then homogenized in ice-cold RIPA
Figure 1 Establishing an orthotopic metastatic prostate cancer model. Primary human prostate cancer tissue (a) was grafted under renal
capsules of testosterone-supplemented male NOD-SCID mice (b). After successful serial subrenal capsule transplantations of tumor grafts
(up to 8 times), tumor tissue was regrafted into anterior prostates of NOD-SCID mice (c, arrows). These orthotopic grafts gave rise to
lymph node metastases (d, arrow), which were successfully regrafted into mouse prostates, giving rise to metastases (e) in the lymph
node (H&E), lung (Ki-67 staining), liver (H&E) and bone (Ki-67 staining).
A new orthotopic prostate cancer model
Y Wang et al
1395
Laboratory Investigation (2005) 85, 13921404
buffer (40 mM Tris-HCl, pH 7.0/1 mM EDTA/4%
glycerol/10 mM DTT/0.2% SDS/20 mM Na molyb-
date/50 mM NaF/complete protease inhibitors
(Roche)) using a polytron homogenizer. Protein
concentrations were determined by RC DC assay
(BioRad). In all, 50 mg of protein was resolved on
an 8.5% SDS-polyacrylamide gel and transferred
to nitrocellulose membrane. The membrane was
blocked for 1 h in 5% (w/v) nonfat dry milk in
20 mM Tris (7.6), 150 mM NaCl and 0.1% Tween-20
and then probed with anti-AR441 at 1:500 (Santa
Cruz) overnight and subsequently with goat anti-
mouse HRP-conjugated secondary antibody. The
protein bands were detected using the enhanced
chemiluminescence kit (Amersham).
Spectral Karyotyping Analysis
PCa1-met tumor tissue was harvested to produce
metaphase spreads via primary cell cultures. To this
end, tumor tissue fragments (about 3–5 mm in
greatest dimension) were finely minced into a paste
and incubated in tissue culture medium at 371C, as
described above. When cultures grew to about 40%
confluency, cells were incubated with 0.1 mg/ml
colcemid (Gibco/BRL) for 30 min followed by 10%
trypsin (Invitrogen) for 5–10 min. Cell swelling, cell
fixation, metaphase slide preparation and pretreat-
ment were performed as previously described.
16,17
Spectral karyotyping (SKY) painting and detection
were performed using the SKYt kit probe cocktail
according to the manufacturer’s recommendations
(Applied Spectral Imaging, Carlsbad, CA, USA).
Pretreated slides were denatured in 70% forma-
mide/2 SSC at 751C for 1–2 min. The SKY probe
was denatured for 7 min at 751C and reannealed at
371C for 1 h. After hybridization for 16–40 h at 371C
in a humid dark chamber, posthybridization washes
and detection were performed as per the manufac-
turer’s directions. Acquisition of hybridization sig-
nals and corresponding DAPI-stained chromosome
spreads was carried out using the SD 200 bio-
imaging system and software (ASI Ltd, MigdalHae-
mek, Israel) attached to a Zeiss Axioplan-2 Micro-
scope (Carl Zeiss, Canada). SKY was performed
using SKYview software version 1.6.2 (ASI, Ltd) as
previously described and in keeping with ISCN
conventions.
16,17
Results
Characterization of a Patient-Derived Prostate Cancer
Line, PCa1
The transplantable PCa1 tumor line was derived
from a subrenal capsule xenograft of primary
prostate cancer tissue in a NOD-SCID mouse (see
Figure 1), supplemented with testosterone to over-
come xenograft atrophy due to intrinsically low
levels of the hormone in SCID mice.
13
It was
propagated by serial subrenal capsule xenografting
in testosterone-supplemented male NOD-SCID mice
for up to eight transplant generations. The tumor
volume doubling time in such mice was approxi-
mately 7 days. Similar to the primary tumor, the
PCa1 xenografts were poorly differentiated, gener-
ally lacking glandular differentiation (Figures 2a
and d). The primary tumor cells expressed human
AR and PSA as indicated by IHC staining using anti-
human AR and PSA antibodies (Figures 2b and c)
with high specificity as indicated by positive and
negative controls (Figures 2h and j); likewise, the
PCa1 xenografts expressed human AR and PSA,
although weakly but still detectable by IHC (Figures
2e and f), thus confirming their human prostatic
origin. Taken together, the data indicate that the
PCa1 tumor line represents an actively growing
human prostatic adenocarcinoma.
Development of a Metastatic PCa1 Subline, PCa1-met
PCa1 tissue could be grown successfully in anterior
prostates of testosterone-supplemented NOD-SCID
mice, in contrast to primary prostatic cancer tissue
whose grafting at this site was less effective.
13
At 6
weeks after orthotopic grafting of PCa1 tissue,
lymph nodes of two out of 12 mice were found to
contain metastatic foci. Metastatic cancer in one of
the hosts, designated PCa1-met, was further deve-
loped by grafting metastases-containing lymph node
tissue in anterior prostates and subsequent trans-
plantation of PCa1-met tissue in prostates or under
the renal capsule (see Figure 1). Figure 3 shows
lymph node metastases of PCa1-met at gross (Figure
3a) and microscopic (Figure 3b) levels. The meta-
static cells stained positively for human mitochon-
dria (Figure 3c) and human Ki-67 (Figure 3d). The
metastatic deposits also weakly expressed AR
(Figure 3e) and PSA (Figure 3f). Detection of
expression of human Ki-67, PSA and mitochondria
provide evidence that the PCa1-met cells are of
human prostatic origin.
Development of Multiorgan Metastases from
Orthotopic PCa1-met Tissue Xenografts
Grafting of PCa1-met tissue into anterior prostates of
NOD-SCID mice produced tumor take rates 495%
(260 out of 270 grafts). The grafts showed poorly
differentiated features and local invasion of host’s
prostate tissue (see Figure 4a). An adjacent section
(Figure 4b) stained positively with anti-human
mitochondria antibody. Furthermore, the orthotopic
grafts weakly expressed AR as observed with PCa1
cells (data not shown). After approximately 2 weeks
of grafting the mice developed multiorgan meta-
stases. As shown in Table 1, the animals developed
metastases in lymph nodes, lung, liver, kidney,
spleen and bone. The histology (H&E staining) of
the metastases in the lung (Figure 4c), liver (Figure
A new orthotopic prostate cancer model
Y Wang et al
1396
Laboratory Investigation (2005) 85, 13921404
5a), kidney (data not shown) and spleen (Figure 6a)
was similar to that of the original PCa1-met (Figure
3). Metastases in the lung (Figure 4d), liver (Figure
5b), kidney (data not shown) and spleen (Figure 6b)
were positively stained with anti-human mitochon-
dria antibodies. Similarly, human-specific Ki-67
staining was detected in metastases to lung (Figure
4e), liver (Figure 5c), kidney (data not shown) and
spleen (Figure 6c). This demonstrates that the
metastases were highly proliferative. IHC analyses
confirmed weak expression of AR in metastases in
the lung (Figure 4f), liver (Figure 5d), kidney (data
not shown) and spleen (Figure 6d); in contrast, AR
expression was negative in host tissues.
Using light microscopy of sections of host’s femur,
micrometastases of human origin in the marrow
were detected via staining with specific anti-human
mitochondria antibody (Figure 7a, arrows). Gross
bone metastases, however, were not observed in any
of the engrafted animals. The human origin of the
micro-metastases was also confirmed by RT-PCR for
human GAPDH (data not shown). Supporting
evidence for their human prostatic origin was
obtained via nested RT-PCR showing that human
PSA mRNA was associated with PCa1-met tissue as
well as with bone marrow (Figure 7d). Similarly,
human AR protein was found to be associated with
PCa1-met tissue (Figure 7e). At high magnification,
H&E-staining (Figure 7b) shows that the metastatic
human prostate cancer cells in a bone lesion had
histologic and cytologic features of the original
tumor cells, including a solid growth pattern,
moderately pale to clear cytoplasm, oval nuclei
and nucleoli and, importantly, produced osteolytic
lesions as demonstrated by replacement of marrow
and resorption of boney trabeculae (Figure 7b). An
adjacent section, stained with anti-human mito-
chondria antibody (Figure 7c), shows cytoplasmic
immunoreactivity within metastatic tumor cells
whereas the remaining bone tissue is negative.
Figure 2 Sections of human primary prostate cancer before grafting (ac) and of the transplantable PCa1 tumor line developed by serial
subrenal capsule transplantation (df). The primary cancer is poorly differentiated and lacks glandular differentiation (a, H&E staining),
but is positive for AR (b) and PSA (c). The transplantable PCa1 tumor line retains poor differentiation and lack of glandular structure (d),
and is weakly positive for ARs (e, arrows) and PSA (f, arrowheads), indicating that it was derived from human prostate cancer. All IHC
staining experiments were carried out using both positive (g, AR; i, PSA) and negative (h, AR; j, PSA) controls using benign human
prostate tissue.
A new orthotopic prostate cancer model
Y Wang et al
1397
Laboratory Investigation (2005) 85, 13921404
SKY Analysis of PCa1-met
Primary PCa1-met tissue cultures were harvested after
5 days of incubation and subsequent 30-min treat-
ment with colcemid. The number of metaphases
obtained was very low (o1% metaphase count), but
sufficient to obtain a consensus SKY karyotype. PCa1-
met cells were diploid with a modal chromosome
Figure 3 Lymph node metastases from orthotopic xenografts of the PCa1-met prostate cancer tissue subline. Gross metastasis (a, arrow)
and microscopic sections of PCa1-met lymph node metastases (b, H&E), showing histological similarity with the parental subrenal
capsule xenografts (see Figure 2). Immunostaining is positive for human mitochondria (c), Ki-67 (d), AR (e) and PSA (f), indicating that
the lymph node metastases are derived from human prostate cancer.
A new orthotopic prostate cancer model
Y Wang et al
1398
Laboratory Investigation (2005) 85, 13921404
number of 45 and loss of the intact Y chromosome.
Sporadic (ie nonclonal, nonrecurring) chromosomal
aberrations were infrequent and the number of
clonal abnormalities was small. Figure 8 shows the
PCa1-met karyotype which is identical to that of PCa1
(not shown). The most frequent clonal changes
observed consisted of three unbalanced translocations
involving six chromosomes (#4, 6, 12, 13, 22 and Y).
Figure 4 Sections of an orthotopic graft of PCa1-met human prostate cancer tissue and its lung metastases. Tumor cells have grown
around a mouse host prostatic duct (a, H&E, arrow), staining positively with anti-human mitochondria antibody, while the host’s
prostatic ductal epithelial cells are negative (b). Two metastatic foci are present in the lung parenchyma (c, H&E, arrows). The tumor cells
stain positively with anti-human mitochondria (d), anti-Ki-67 (e) and anti-AR (f) antibodies.
A new orthotopic prostate cancer model
Y Wang et al
1399
Laboratory Investigation (2005) 85, 13921404
Discussion
Therapy-resistant metastasis is the leading cause of
human prostate cancer death.
1
Currently, there are
no effective therapies for metastatic prostate cancer,
and this deficiency is in part a reflection of a lack of
understanding of the mechanisms involved in the
progression of the disease.
2
Research in this area has
been hampered by a lack of clinically relevant,
experimental models.
3
For example, certain models
are based on injection of highly anaplastic human
prostate cancer cells into the bloodstream or into
target organs (eg, tibia) of immuno-deficient mice and
produce ‘experimental metastatic foci’.
18
Such foci are
not a consequence of the normal metastatic behavior
of cancer cells (intravasation, migration, extravasa-
tion). Instead, they rather reflect the ability of the
injected cells to proliferate in certain distant locations.
In contrast, models based on implantation of prostate
cancer tissue, as distinct from cells, into prostates of
experimental animals appear clinically more appro-
priate, since an implantation site is used that is more
representative of the original tumor environment.
19
There is accumulating evidence that metastatic
behavior of a neoplasm is governed not only by
intrinsic tumor factors, but also to a large extent by
multiple interactions between cancer cells and their
microenvironment promoting metastasis of discrete
tumor subpopulations.
4
Tumor-associated macro-
phages, for example, can induce matrix breakdown,
tumor cell motility and angiogenesis.
20,21
In esta-
blishing a model that represents a patient’s tumor, it
Table 1 Incidence of prostate cancer metastases in host
tissues (%)
Weeks after
grafting
(# of mice)
Lymph
nodes
Lung Liver Spleen Kidney Bone
2 (10) 80 30 0 0 0 0
3 (10) 100 100 30 30 10 0
4 (10) 100 100 80 90 40 0
6 (10) 100 100 90 90 100 0
8 (7) 100 100 100 100 100 43
Figure 5 Sections of liver metastases from orthotopically engrafted PCa1-met human prostate cancer tissue. Tumor cells are present in
the liver parenchyma (a, H&E, arrow), staining positively with anti-human mitochondria (b), anti-Ki-67 (c) and anti-AR (d) antibodies.
A new orthotopic prostate cancer model
Y Wang et al
1400
Laboratory Investigation (2005) 85, 13921404
is therefore essential that the grafted specimen
contains all of the crucial components of the original
tumor and that these are retained in the grafts. In
this regard, it is notable that the initial subrenal
capsule xenografts of prostate cancer tissue indeed
contained human stroma which, however, was
gradually replaced by mouse stroma.
22
Primary prostatic neoplasms are thought to con-
sist of a variety of carcinoma subpopulations
showing differences in invasive and metastatic
properties.
7
The present study shows that prostate
cancer tissue from a patient can be readily grown in
prostates of NOD-SCID mice following its establish-
ment as subrenal capsule xenografts. It appears that
the high tumor take rate achievable with the
subrenal capsule grafting technique, for both low
and higher grade cancers, minimizes the chance of
losing subpopulations of the original prostatectomy
specimen.
13
Potential loss of such subpopulations
appears much greater when other grafting sites
with much lower engraftment rates are used
(eg the subcutaneous compartment). The PCa1 line,
developed in this study, represents an actively
growing human prostatic adenocarcinoma line with
poorlydifferentiated features and general lack of
glandular differentiation (Figure 2). PCa1-met, the
metastatic subline derived from lymph node metas-
tases of the PCa1 line, resembles its parental line
with regard to poorly differentiated features, as well
as its human origin and expression of AR and PSA
(Figures 3 and 4). The metastases in the mice
receiving orthotopic transplantation of PCa1-met
tissue were located in lymph nodes, lung, liver,
kidney, spleen and bone (Figures 3–7), which are
common metastatic sites in prostate cancer patients.
The finding in mouse bone of orthotopically grafted
cancer cells of prostatic origin and exhibiting
osteolytic action is particularly interesting, since
this phenomenon is not observed with other
currently available metastatic prostate cancer
models.
23
It appears therefore that a subline such
as the PCa1-met, produced by subrenal capsule
and subsequent orthotopic xenografting, is highly
suitable for use in NOD-SCID mice as a model for
Figure 6 Sections of spleen metastases from orthotopically engrafted PCa1-met human prostate cancer tissue. Foci of tumor cells are
present in splenic parenchyma (a, H&E, arrows), staining positively with anti-human mitochondria (b), anti-Ki-67 (c) and anti-AR (d)
antibodies.
A new orthotopic prostate cancer model
Y Wang et al
1401
Laboratory Investigation (2005) 85, 13921404
Figure 7 Sections of bone marrow containing metastases from orthotopically engrafted PCa1-met human prostate cancer tissue. Low-
power photomicrograph (produced by IHC staining using anti-human mitochondria) showing replacement of marrow elements by
metastatic carcinoma (a, arrows) with thinning of boney trabeculae resulting in osteolytic appearance. Higher power photomicrographs
showing sheets of tumor cells and a single thinned trabecula (arrows) (b, H&E staining; c, staining with anti-human mitochondria
antibodies). (d) RT-PCR products for PSA (human) and b2-microglobulin (mouse; control) using total RNA harvested from tissues.
(e) Western blot of human AR from various tissues.
Figure 8 Spectral karyotype composite of the PCa1-met human prostate cancer line. DAPI-banded preparation of metaphase
chromosomes from a cell following short-term culture (a), hybridized to SKY paints (b) and after pseudocolor classification (c). Resultant
karyotype table is presented in (d). A SKY profile of PCa1-met: 45, X,Y,der(6)t(6;13)(q24;q22), der(12)t(Y;12)(q11;p11),
der(22)t(4;22)(?;p11).
A new orthotopic prostate cancer model
Y Wang et al
1402
Laboratory Investigation (2005) 85, 13921404
metastatic prostate cancer. It may be noted that the
PCa1-met line was found to be androgen-indepen-
dent, growing readily in mice following androgen
ablation (unpublished observation).
SKY analyses of three widely used prostate can-
cer cell lines (LNCaP, DU-145, PC-3) have demon-
strated aneuploid karyotypes with many chromo-
somal alterations including complex chromosomal
rearrangements and a high degree of karyotypic
instability. No common chromosomal rearrangement
or translocation breakpoint was identified.
24,25
In
contrast to these highly anaplastic cell lines, early-
stage prostatic tumors are typically diploid with
some propensity to progress to aneuploidy and
karyotypic complexity in advanced stages.
26
The
karyotype of PCa1-met is comparable to the latter,
with surprisingly few chromosomal alterations
produced by as few as six breakpoints involving
chromosomes 4, 6, 12, 13, 22 and Y. Common
involvement of chromosome 8, typically a loss of 8p
with gain of 8q, reported previously,
27
was not seen
in the present study using SKY. However, smaller
but significant chromosomal alterations, such as
small interstitial deletions, insertions/duplications
and inversions cannot be revealed by whole-chro-
mosome painting and would require higher-resolu-
tion techniques such as locus-specific-FISH, Mband
FISH or array CGH. Loss of the Y chromosome is
frequently associated with aging. However, it has
also been suggested that loss of certain Y chromo-
some specific genes may play a role in the
pathogenesis of prostate cancer.
28
The finding of
simple and identical karyotypes in PCa1 (data not
shown) and PCa1-met, its metastatic subline, sug-
gests that significant chromosome and karyotypic
instability are not characteristics of these tumor
lines, in contrast to commonly used prostate cancer
cell lines such as DU-145.
29
It is also likely that
conservation of such few chromosomal changes is
of biological significance, by offering selective
advantage to the tumor cells and maintenance of
malignant and/or metastatic properties.
In conclusion, the orthotopic prostate cancer
metastasis model developed in this study should
be useful for investigations of mechanisms under-
lying prostate cancer metastasis and therapeutic
applications. The methodology used to develop this
model may also be suitable for developing ortho-
topic metastatic cancer models of other types of
human cancer. Thus, the first step of the methodo-
logy, that is, subrenal capsule grafting, has been
successful for a variety of primary human cancer
tissues, including cancers of the ovary,
30
kidney,
22
lung, pancreas and lymphoid tissue (unpublished
observations).
Acknowledgements
We thank Rebecca Wu and Lily Wei for excellent
technical assistance. Paula Marrano is thanked for
technical expertise and help in SKY analyses. This
study was supported in part by grants awarded
to YZW by NCI Canada (#014053), the US Army
Department of Defense, USAMRMC W81XWH-04-1-
0290, the Prostate Cancer Foundation of Canada
and to GRC by the National Cancer Institute
(# CA89520). J-C Cutz was a CIHR fellow in
Molecular Oncologic Pathology.
References
1 Fidler IJ. Critical determinants of metastasis. Semin
Cancer Biol 2002;12:89–96.
2 Guseva NV, Taghiyev AF, Rokhlin OW, et al. Death
receptor-induced cell death in prostate cancer. J Cell
Biochem 2004;91:70–99.
3 Klein KA, Reiter RE, Redula J, et al. Progression of
metastatic human prostate cancer to androgen inde-
pendence in immunodeficient SCID mice. Nat Med
1997;3:402–408.
4 Fidler IJ. The pathogenesis of cancer metastasis: the
‘seed and soil’ hypothesis revisited. Nat Rev Cancer
2003;3:453–458.
5 Almholt K, Johnsen M. Stromal cell involvement in
cancer. Recent Results Cancer Res 2003;162:31–42.
6 Voskoglou-Nomikos T, Pater JL, Seymour L. Clinical
predictive value of the in vitro cell line, human
xenograft, and mouse allograft preclinical cancer
models. Clin Cancer Res 2003;9:4227–4239.
7 Van Weerden WM, de Ridder CM, Verdaasdonk CL,
et al. Development of seven new human prostate
tumor xenograft models and their histopathological
characterization. Am J Pathol 1996;149:1055–1062.
8 Lubaroff DM, Cohen MB, Schultz LD, et al. Survival
of human prostate carcinoma, benign hyperplastic
prostate tissues, and IL-2-activated lymphocytes in
scid mice. Prostate 1995;27:32–41.
9 Pretlow TG, Wolman SR, Micale MA, et al. Xenografts
of primary human prostatic carcinoma. J Natl Cancer
Inst 1993;85:394–398.
10 Wainstein MA, He F, Robinson D, et al. CWR22:
androgen-dependent xenograft model derived from a
primary human prostatic carcinoma. Cancer Res 1994;
54:6049–6052.
11 Pretlow TG, Delmoro CM, Dilley GG, et al. Transplant-
ation of human prostatic carcinoma into nude mice in
matrigel. Cancer Res 1991;51:3814–3817.
12 Stephenson RA, Dinney CP, Gohji K, et al. Metastatic
model for human prostate cancer using orthotopic
implantation in nude mice. J Natl Cancer Inst 1992;
84:951–957.
13 Wang YZ, Revelo MP, Sudilovsky D, et al. Develop-
ment and characterization of efficient xenograft
models for benign and malignant human prostate
tissue. Prostate 2005;64:149–159.
14 Wang YZ, Hayward SW, Donjacour AA, et al. Sex
hormone-induced carcinogenesis in Rb-deficient pro-
state tissue. Cancer Res 2000;60:6008–6017.
15 Wang Y Z, Sudilovsky D, Zhang B, et al. A human
prostatic epithelial model of hormonal carcinogenesis.
Cancer Res 2001;61:6064–6072.
16 Bayani J, Zielenska M, Marrano P, et al. Molecular
cytogenetic analysis of medulloblastomas and supraten-
torial primitive neuroectodermal tumors by using con-
ventional banding, comparative genomic hybridization,
and spectral karyotyping. J Neurosurg 2000;93:437–448.
A new orthotopic prostate cancer model
Y Wang et al
1403
Laboratory Investigation (2005) 85, 13921404
17 Schrock E, du Manoir S, Veldman T, et al. Multicolor
spectral karyotyping of human chromosomes. Science
1996;273:494–497.
18 Pfitzenmaier J, Quinn JE, Odman AM, et al. Character-
ization of C4-2 prostate cancer bone metastases and
their response to castration. J Bone Miner Res 2003;
18:1882–1888.
19 Killion JJ, Radinsky R, Fidler IJ. Orthotopic models are
necessary to predict therapy of transplantable tumors
in mice. Cancer Metastasis Rev 1998;17:279–284.
20 Radisky DC, Bissell MJ. Cancer. Respect thy neighbor.
Science 2004;303:775–777.
21 Pollard JW. Tumour-educated macrophages promote
tumour progression and metastasis. Nat Rev Cancer
2004;4:71–78.
22 Liou LS, Zhou M, Wang Y, et al. A living human tumor
bank using an orthotopic xenograft mouse model:
feasibility and implications of angiogenesis and pro-
liferation. J Urol 2004;171:208 (abstract).
23 Nemeth JA, Harb JF, Barroso U, et al. Severe combined
immunodeficient-hu model of human prostate cancer
metastasis to human bone. Cancer Res 1999;59:
1987–1993.
24 Pan Y, Kytola S, Farnebo F, et al. Characterization of
chromosomal abnormalities in prostate cancer cell
lines by spectral karyotyping. Cytogenet Cell Genet
1999;87:225–232.
25 Beheshti B, Karaskova J, Park PC, et al. Identification
of a high frequency of chromosomal rearrangements in
the centromeric regions of prostate cancer cell lines by
sequential giemsa banding and spectral karyotyping.
Mol Diagn 2000;5:23–32.
26 Ozen M, Pathak S. Genetic alterations in human
prostate cancer: a review of current literature. Anti-
cancer Res 2000;20:1905–1912.
27 Cher ML, MacGrogan D, Bookstein R, et al. Comparative
genomic hybridization, allelic imbalance, and fluores-
cence in situ hybridization on chromosome 8 in prostate
cancer. Genes Chromosomes Cancer 1994;11:153–162.
28 Perinchery G, Sasaki M, Angan A, et al. Deletion
of Y-chromosome specific genes in human prostate
cancer. J Urol 2000;163:1339–1342.
29 Beheshti B, Park PC, Sweet JM, et al. Evidence of
chromosomal instability in prostate cancer determined
by spectral karyotyping (SKY) and interphase fish
analysis. Neoplasia 2001;3:62–69.
30 Lee CH, Xue H, Sutcliffe M, et al. Establishment of
subrenal capsule xenografts of primary human ovarian
tumors in SCID mice: potential models. Gynecol Oncol
2005;96:48–55.
A new orthotopic prostate cancer model
Y Wang et al
1404
Laboratory Investigation (2005) 85, 13921404
... The PC3 cell line was identified in 1979 from a patient with bone metastatic prostate cancer [42]. It is highly metastatic, hormone resistant and PSA negative [43][44][45][46]. In addition, the PC3 cell line has stronger characteristics of neuroendocrine or small cell carcinoma. ...
... PDXs are known to retain the morphology and have the same immunohistochemical profile of the original human prostate cancer donor [79,80,83]. As is the issue with cell line derived animal models, in order to create a spontaneous bone metastatic prostate cancer PDX model, repeated inoculation of extracted cancer cells over several generations is mandatory [43]. In a study by Wang and colleagues [43], patient tumor samples were transplanted into the subrenal capsule of SCID mice for cell culture, and then reinjected into the prostate of mice until the mice developed lymph node metastasis. ...
... As is the issue with cell line derived animal models, in order to create a spontaneous bone metastatic prostate cancer PDX model, repeated inoculation of extracted cancer cells over several generations is mandatory [43]. In a study by Wang and colleagues [43], patient tumor samples were transplanted into the subrenal capsule of SCID mice for cell culture, and then reinjected into the prostate of mice until the mice developed lymph node metastasis. Cancer cells retrieved from the metastatic lymph nodes thereafter showed to spontaneously develop metastatsis to various organs including the bone when inoculated into the prostate of mice [43]. ...
Article
Full-text available
Metastatic disease is a main cause of mortality in prostate cancer and remains to be incurable despite emerging new treatment agents. Development of novel treatment agents are confined within the boundaries of our knowledge of bone metastatic prostate cancer. Exploration into the underlying mechanism of metastatic tumorigenesis and treatment resistance will further expose novel targets for novel treatment agents. Up to date, many of these researches have been conducted with animal models which have served as classical tools that play a pivotal role in understanding the fundamental nature of cancer. The ability to reproduce the natural course of prostate cancer would be of profound value. However, currently available models do not reproduce the entire process of tumorigenesis to bone metastasis and are limited to reproducing small portions of the entire process. Therefore, knowledge of available models and understanding the strengths and weaknesses for each model is key to achieve research objectives. In this article, we take an overview of cell line injection animal models and patient derived xenograft models that have been applied to the research of human prostate cancer bone metastasis.
... Tumor tissue was fixed with formalin and embedded in paraffin. Tissues were sectioned, probed, and stained for IHC analyses as previously described [19,20]. The following antibodies and conjugates were used: rabbit anti-human B7-H3 (Clone: EPR20115; AbCam, Cambridge, UK; 1:1000), mouse anti-human Ki67 antibody (Clone: MIB-1; Dako, Jena, Germany; 1:25), rabbit anti-human androgen receptor (AR) antibody (Clone: ER179(2); AbCam; 1:100), rabbit anti-human prostate-specific antigen (PSA) antibody (Clone: EP1588Y; AbCam; 1:200), biotinylated goat anti-rabbit antibody (Vector Laboratories, Burlingame, CA, USA; 1:200), and biotinylated goat anti-mouse antibody (Vector Laboratories; 1:200). ...
Article
Full-text available
Androgen deprivation therapy (ADT) is the standard care for advanced prostate cancer (PCa) patients. Unfortunately, although tumors respond well initially, they enter dormancy and eventually progress to fatal/incurable castration-resistant prostate cancer (CRPC). B7-H3 is a promising new target for PCa immunotherapy. CD276 (B7-H3) gene has a presumptive androgen receptor (AR) binding site, suggesting potential AR regulation. However, the relationship between B7-H3 and AR is controversial. Meanwhile, the expression pattern of B7-H3 following ADT and during CRPC progression is largely unknown, but critically important for identifying patients and determining the optimal timing of B7-H3 targeting immunotherapy. In this study, we performed a longitudinal study using our unique PCa patient-derived xenograft (PDX) models and assessed B7-H3 expression during post-ADT disease progression. We further validated our findings at the clinical level in PCa patient samples. We found that B7-H3 expression was negatively regulated by AR during the early phase of ADT treatment, but positively associated with PCa proliferation during the remainder of disease progression. Our findings suggest its use as a biomarker for diagnosis, prognosis, and ADT treatment response, and the potential of combining ADT and B7-H3 targeting immunotherapy for hormone-naïve PCa treatment to prevent fatal CRPC relapse.
... PDXs are established by grafting human tumours into immunedeficient mice 12 . In experienced laboratories, 33-94% of prostate cancer tissue samples survive for the first generation after grafting as primary PDXs [13][14][15][16] . Difficulty arises in establishing grafts that grow in size and can be transplanted into new host mice to continue their propagation. ...
Article
Patient-derived xenografts (PDXs) are generated by engrafting human tumours into mice. Serially transplantable PDXs are used to study tumour biology and test therapeutics, linking the laboratory to the clinic. Although few prostate cancer PDXs are available in large repositories, over 330 prostate cancer PDXs have been established, spanning broad clinical stages, genotypes and phenotypes. Nevertheless, more PDXs are needed to reflect patient diversity, and to study new treatments and emerging mechanisms of resistance. We can maximize the use of PDXs by exchanging models and datasets, and by depositing PDXs into biorepositories, but we must address the impediments to accessing PDXs, such as institutional, ethical and legal agreements. Through collaboration, researchers will gain greater access to PDXs representing diverse features of prostate cancer. This Perspective covers existing patient-derived xenografts (PDXs) of prostate cancer, and their features and uses in basic and preclinical research. The authors also discuss the need for additional PDXs, and how collaboration in prostate cancer PDX research can be improved.
... This model mimics the metastatic process more precisely (129). Reports of orthotopic models for human PCa have been published (130)(131)(132). A modification of the orthotopic model for PCa using a cell injection in one of the anterior lobes of the NOD/sCID mouse prostate has been developed by our laboratory (133,134). ...
Chapter
Prostate cancer (PCa) incidence has increased during the last decades, becoming one of the leading causes of death by cancer in men worldwide. During an extended period of prostate cancer, malignant cells are androgen-sensitive being testosterone the main responsible for tumor growth. Accordingly, treatments blocking production and action of testosterone are mostly used. However, during disease progression, PCa cells become androgen insensitive producing a castration-resistant stage with a worse prognosis. Overcoming castration-resistant prostate cancer (CRPC) has become a great challenge in the management of this disease. In the search for molecular pathways leading to therapy resistance, the epithelial–mesenchymal transition (EMT), and particularly the transcription factors zinc finger E-box-binding homeobox 1 (Zeb1) and zinc finger protein SNAI1 (Snail), master genes of the EMT, have shown to have pivotal roles. Also, the discovery that cancer stem cells (CSCs) can be generated de novo from their non-CSCs counterpart has led to the question whereas these EMT transcription factors could be implicated in this dynamic conversion between non-CSC and CSC. In this review, we analyze evidence supporting the idea that Zeb1 and Snail induce cell malignancy and cancer stem cell phenotype in prostate cells, increasing androgen synthesis capacity and therapy resistance.KeywordsProstate cancerepithelial-mesenchymal transitionZeb1SNAILCancer stem cellsAndrogens
... This model mimics, more accurately, the metastatic process. Lastly, a few reports on the orthotopic models for human PCa have been published [114,115]. We have developed a modification of an orthotopic model for PCa using a cell injection in one of the anterior lobes of the NOD/SCID mouse prostate [90]. ...
Article
Full-text available
Prostate cancer (PCa) is a leading cause of cancer death in men, worldwide. Mortality is highly related to metastasis and hormone resistance, but the molecular underlying mechanisms are poorly understood. We have studied the presence and role of cancer stem cells (CSCs) and the Epithelial–Mesenchymal transition (EMT) in PCa, using both in vitro and in vivo models, thereby providing evidence that the stemness–mesenchymal axis seems to be a critical process related to relapse, metastasis and resistance. These are complex and related processes that involve a cooperative action of different cancer cell subpopulations, in which CSCs and mesenchymal cancer cells (MCCs) would be responsible for invading, colonizing pre-metastatic niches, initiating metastasis and an evading treatments response. Manipulating the stemness–EMT axis genes on the androgen receptor (AR) may shed some light on the effect of this axis on metastasis and castration resistance in PCa. It is suggested that the EMT gene SNAI2/Slug up regulates the stemness gene Sox2, and vice versa, inducing AR expression, promoting metastasis and castration resistance. This approach will provide new sight about the role of the stemness–mesenchymal axis in the metastasis and resistance mechanisms in PCa and their potential control, contributing to develop new therapeutic strategies for patients with metastatic and castration-resistant PCa.
Chapter
Patient-derived xenografts (PDXs) represent a critical advancement in preclinical cancer research, wherein human tumor samples are implanted into animal models for evaluation of therapeutic responses. PDXs have emerged as indispensable tools in translational cancer research, facilitating investigation into tumor microenvironments and personalized medicine. This chapter elucidates the historical evolution of PDXs, from early attempts in the eighteenth century to contemporary immunocompromised host models that enhance engraftment success.
Article
Androgen deprivation therapy has a central role in the treatment of advanced prostate cancer, often causing initial tumour remission before increasing independence from signal transduction mechanisms of the androgen receptor and then eventual disease progression. Novel treatment approaches are urgently needed, but only a fraction of promising drug candidates from the laboratory will eventually reach clinical approval, highlighting the demand for critical assessment of current preclinical models. Such models include standard, genetically modified and patient-derived cell lines, spheroid and organoid culture models, scaffold and hydrogel cultures, tissue slices, tumour xenograft models, patient-derived xenograft and circulating tumour cell eXplant models as well as transgenic and knockout mouse models. These models need to account for inter-patient and intra-patient heterogeneity, the acquisition of primary or secondary resistance, the interaction of tumour cells with their microenvironment, which make crucial contributions to tumour progression and resistance, as well as the effects of the 3D tissue network on drug penetration, bioavailability and efficacy.
Article
Metastatic disease is a main cause of mortality in prostate cancer and remains to be uncurable despite emerging new treatment agents. Development of novel treatment agents are confined within the boundaries of our knowledge of bone metastatic prostate cancer. Exploration into the underlying mechanism of metastatic tumorigenesis and treatment resistance will further expose novel targets for novel treatment agents. Up to date, many of these researches have been conducted with animal models which have served as classical tools that play a pivotal role in understanding the fundamental nature of cancer. The ability to reproduce the natural course of prostate cancer would be of profound value. However, currently available models cannot reproduce the entire process of tumorigenesis to bone metastasis and are limited to reproducing small portions of the entire process. Therefore, knowledge of available models and understanding the strengths and weaknesses for each model is key to achieve research objectives. In this article, we take an overview of cell line injection animal models and patient-derived xenograft models that have been applied to the research of human prostate cancer bone metastasis.
Chapter
Researchers' understanding of the precise biological mechanisms responsible for bone metastases formation is incomplete, hampering the development of successful treatment and prevention strategies. In order to address this problem, researchers have turned increasingly to the use of animal models, although these generally represent only particular stages of human disease rather than the gradual transition from benign disease through hyperplasia and subsequent aggressive and advanced metastatic disease. Bone metastases represent a particular challenge, as these are often preceded or accompanied by visceral metastasis that have profound effects on morbidity and survival. The majority of in vivo studies of bone metastases use rodents, mainly mice, as these are widely available, have a short life span, and a relatively large number of animals can be easily investigated. Only a few specialist facilities have the capacity to carry out research using larger animals (primarily dogs), and as a result only a limited amount of work is carried out using such models. However, rodents do not naturally develop bone metastases, suggesting that they do not provide the optimal setting to study this aspect of human cancer, and key molecules and mechanisms that drive human disease may potentially be missed in murine model systems. Despite their clear limitations, in vivo models of bone metastases have provided valuable insights into the molecules and pathways that may play a role in the development of human disease, as well as the effects of a range of therapies.
Article
Full-text available
Prostate cancer mortality results from metastasis and is often coupled with progression from androgen-dependent to androgen-independent growth. Unfortunately, no effective treatment for metastatic prostate cancer increasing patient survival is available. The absence of effective therapies reflects in part a lack of knowledge about the molecular mechanisms involved in the development and progression of this disease. Apoptosis, or programmed cell death, is a cell suicide mechanism that enables multicellular organisms to regulate cell number in tissues. Inhibition of apoptosis appears to be a critical pathophysiological factor contributing to the development and progression of prostate cancer. Understanding the mechanism(s) of apoptosis inhibition may be the basis for developing more effective therapeutic approaches. Our understanding of apoptosis in prostate cancer is relatively limited when compared to other malignancies, in particular, hematopoietic tumors. Thus, a clear need for a better understanding of apoptosis in this malignancy remains. In this review we have focused on what is known about apoptosis in prostate cancer and, more specifically, the receptor/ligand-mediated pathways of apoptosis as potential therapeutic targets.
Article
Full-text available
The way in which cytogenetic aberrations develop in prostate cancer (Cap) is poorly understood. Spectral karyotype (SKY) analysis of Cap cell lines has shown that they have unstable karyotypes and also have features associated with chromosomal instability (CIN). To accurately determine the incidence of de novo structural and numerical aberrations in vitro in Cap, we performed SKY analysis of three independent clones derived from one representative cell line, DU145. The frequent generation of new chromosomal rearrangements and a wide variation in the number of structural aberrations within two to five passages suggested that this cell line exhibited some of the features associated with a CIN phenotype. To study numerical cell-to-cell variation, chromosome 8 aneusomy was assessed in the LNCaP, DU145, and PC-3 cell lines and a patient cohort of 15 Cap primary tumors by interphase fluorescence in situ hybridization (FISH). This analysis showed that a high frequency of numerical alteration affecting chromosome 8 was present in both in vitro and in Cap tissues. In comparison to normal controls, the patient cohort had a statistically significant (P
Article
Full-text available
The role of estrogen (ER) and progesterone receptors (PR) in breast cancer is well established. Identification of the second human estrogen receptor, the estrogen receptor β (ERβ), prompted us to evaluate its role in breast cancer. We studied the expression of ERβ by immunohistochemistry and mRNA in situ hybridization in 92 primary breast cancers and studied its association with ERα, PR, and various other clinicopathological factors. Sixty percent of tumors were defined as ERβ-positive (nuclear staining in >20% of the cancer cells). Normal ductal epithelium and 5 of 7 intraductal cancers were also found to express ERβ. Three-fourths of the ERα- and PR-positive tumors were positive for ERβ, whereas ERα and PR were positive in 87% and 67. of ERβ-positive tumors, respectively. ERβ was associated with negative axillary node status (P < 0.0001), low grade (P = 0.0003), low S-phase fraction (P = 0.0003), and premenopausal status (P = 0.04). In conclusion, the coexpression of ERβ with ERα and PR as well as its association with the other indicators of low biological aggressiveness of breast cancer suggest that ERβ-positive tumors are likely to respond to hormonal therapy. The independent predictive value of ERβ remains to be established.
Article
Background: Currently, prostate cancer (CaP) cytogenetics is not well defined, largely because of technical difficulties in obtaining primary tumor metaphases. Methods and Results: We examined three CaP cell lines (LNCaP, DU145, PC-3) using sequential Giemsa banding and spectral karyotyping (SKY) to search for a common structural aberration or translocation breakpoint. No consistent rearrangement common to all three cell lines was detected. A clustering of centromeric translocation breakpoints was detected in chromosomes 4, 5, 6, 8, 11, 12, 14, and 15 in DU145 and PC-3. Both these lines were found to have karyotypes with a greater level of complexity than LNCaP. Conclusion: The large number of structural aberrations present in DU145 and PC-3 implicate an underlying chromosomal instability and subsequent accumulation of cytogenetic alterations that confer a selective growth advantage. The high frequency of centromeric rearrangements in these lines indicates a potential role for mitotic irregularities associated with the centromere in CaP tumorigenesis.
Article
CONTEXT: Despite decades of accumulated observational evidence, the balance of risks and benefits for hormone use in healthy postmenopausal women remains uncertain. OBJECTIVE: To assess the major health benefits and risks of the most commonly used combined hormone preparation in the United States. DESIGN: Estrogen plus progestin component of the Women's Health Initiative, a randomized controlled primary prevention trial (planned duration, 8.5 years) in which 16608 postmenopausal women aged 50-79 years with an intact uterus at baseline were recruited by 40 US clinical centers in 1993-1998. INTERVENTIONS: Participants received conjugated equine estrogens, 0.625 mg/d, plus medroxyprogesterone acetate, 2.5 mg/d, in 1 tablet (n = 8506) or placebo (n = 8102). MAIN OUTCOMES MEASURES: The primary outcome was coronary heart disease (CHD) (nonfatal myocardial infarction and CHD death), with invasive breast cancer as the primary adverse outcome. A global index summarizing the balance of risks and benefits included the 2 primary outcomes plus stroke, pulmonary embolism (PE), endometrial cancer, colorectal cancer, hip fracture, and death due to other causes. RESULTS: On May 31, 2002, after a mean of 5.2 years of follow-up, the data and safety monitoring board recommended stopping the trial of estrogen plus progestin vs placebo because the test statistic for invasive breast cancer exceeded the stopping boundary for this adverse effect and the global index statistic supported risks exceeding benefits. This report includes data on the major clinical outcomes through April 30, 2002. Estimated hazard ratios (HRs) (nominal 95% confidence intervals [CIs]) were as follows: CHD, 1.29 (1.02-1.63) with 286 cases; breast cancer, 1.26 (1.00-1.59) with 290 cases; stroke, 1.41 (1.07-1.85) with 212 cases; PE, 2.13 (1.39-3.25) with 101 cases; colorectal cancer, 0.63 (0.43-0.92) with 112 cases; endometrial cancer, 0.83 (0.47-1.47) with 47 cases; hip fracture, 0.66 (0.45-0.98) with 106 cases; and death due to other causes, 0.92 (0.74-1.14) with 331 cases. Corresponding HRs (nominal 95% CIs) for composite outcomes were 1.22 (1.09-1.36) for total cardiovascular disease (arterial and venous disease), 1.03 (0.90-1.17) for total cancer, 0.76 (0.69-0.85) for combined fractures, 0.98 (0.82-1.18) for total mortality, and 1.15 (1.03-1.28) for the global index. Absolute excess risks per 10 000 person-years attributable to estrogen plus progestin were 7 more CHD events, 8 more strokes, 8 more PEs, and 8 more invasive breast cancers, while absolute risk reductions per 10 000 person-years were 6 fewer colorectal cancers and 5 fewer hip fractures. The absolute excess risk of events included in the global index was 19 per 10 000 person-years. CONCLUSIONS: Overall health risks exceeded benefits from use of combined estrogen plus progestin for an average 5.2-year follow-up among healthy postmenopausal US women. All-cause mortality was not affected during the trial. The risk-benefit profile found in this trial is not consistent with the requirements for a viable intervention for primary prevention of chronic diseases, and the results indicate that this regimen should not be initiated or continued for primary prevention of CHD.
Article
The transforming growth factor (TGF)-βs are potent growth inhibitors of normal epithelial cells. In established tumor cell systems, however, the preponderant experimental evidence suggests that TGF-βs can foster tumor-host interactions that indirectly support the viability and/or progression of cancer cells. The timing of this 'TGF-β switch' during the progressive transformation of epithelial cells is not clear. More recent evidence also suggests that autocrine TGF-β signaling is operative in some tumor cells, and can also contribute to tumor invasiveness and metastases independent of an effect on nontumor cells. The dissociation of antiproliferative and matrix associated effects of autocrine TGF-β signaling at a transcriptional level provides for a mechanism(s) by which cancer cells can selectively use this signaling pathway for tumor progression. Data in support of the cellular and molecular mechanisms by which TGF-β signaling can accelerate the natural history of tumors will be reviewed in this section.
Article
We have carried out a quantitative analysis of ER-α and ER-β mRNA expression in normal (n = 11) and breast cancer (n = 112) tissues using a real-time (Taq-Man) PCR assay. Expression of ER-β mRNA variants has also been studied by triple-primer PCR assay. ER-α mRNA levels in normal breast tissues were significantly (p < 0.01) lower than those in ER-positive breast cancers but not significantly different from those in ER-negative breast cancers. However, ER-β mRNA levels in normal breast tissues were significantly (p < 0.01) higher than those in ER-positive and ER-negative breast cancers. Proportions of ER-β1 and ER-β2 mRNA expression among total ER-β mRNA expression were significantly higher and those of ER-β5 and ER-β5` mRNA were significantly lower in normal breast tissues than in ER-positive and ER-negative breast cancers. ER-β mRNA levels and proportions of ER-β mRNA variants did not show any significant correlation with age, tumor size, lymph node status and histological grade. Our results demonstrate that ER-α mRNA is up-regulated and ER-β mRNA is down-regulated during carcinogenesis of breast cancers. Changes in proportions of ER-β mRNA variants are also implicated in this process. Int. J. Cancer 88:733–736, 2000.