ArticlePDF Available

Comparative evaluation of survivin, midkine, and CXCR4 promoters for transcriptional targeting of glioma gene therapy

Taylor & Francis
Cancer Biology & Therapy
Authors:
  • Quorum X Diagnostics Inc and AdCure Bio, Inc
  • Northwestern University

Abstract

Transcriptional targeting is a key strategy to enhance therapeutic efficacy of gene therapy applications. In the context of oncolytic virotherapy, transcriptional promoter elements are used from genes that are over expressed in a variety of malignant cancers. In the present study, we examined the feasibility of transcriptional targeting to glioma cells by comparing the activity of survivin, midkine, and CXCR4 tumor-specific promoters. To evaluate the expression level of several glioma related genes, we performed quantitative RT-PCR analyses on samples obtained from cell lines and patients. To determine specific level of gene expression mediated by selective promoter elements, we measured luciferase expression in glioma samples transduced with replication deficient adenoviral vectors. Finally, we incorporated the optimal promoters into a conditionally replicative adenoviral vector, CRAd-5/3, and examined the cytopathic effect in vitro. The survivin promoter demonstrated the highest level of mRNA expression in primary tumor samples and cell lines. Transcriptional targeting was confirmed by infection of glioma cells with an adenovirus expression vector containing a surviving-driven luciferase reporter gene. Of the tested promoters, minimal level of survivin activity was detected in normal human liver and brain. A novel vector, CRAd-survivin5/3, with E1a under the control of the survivin promoter, exhibited enhanced cytopathic effect in vitro. Our data demonstrate that the survivin promoter element is very active in glioma samples and has low activity in normal human brain and liver. A novel oncolytic virus, CRAd-survivin-5/3, was effective against a panel of glioma cell lines in vitro. Our results suggest that employing the survivin promoter element in the context of CRAd-5/3 may present a new opportunity for the development of glioma specific oncolytic vectors.
©2007 LANDES BIOSCIENCE. DO NOT DISTRIBUTE.
Research Paper
Comparative Evaluation of Survivin, Midkine and CXCR4 Promoters
for Transcriptional Targeting of Glioma Gene Therapy
[Cancer Biology & Therapy 6:5, 679-685, May 2007]; ©2007 Landes Bioscience
Ilya V. Ulasov1
Angel A. Rivera2
Adam M. Sonabend1
Lisa B. Rivera2
Ming Wang2
Zeng B. Zhu2
Maciej S. Lesniak1,*
1Division of Neurosurgery; The University of Chicago; Chicago, Illinois USA
2Division of Human Gene Therapy; Departments of Medicine, Pathology, Surgery;
The Gene Therapy Center; University of Alabama at Birmingham; Birmingham,
Alabama USA
*Correspondence to: Maciej S. Lesniak; The University of Chicago; Division of
Neurosurgery; 5841 S. Maryland Avenue; MC 3026; Chicago 60637 Illinois USA;
Tel.: 773.834.4757; Fax: 773.834.2608; Email: mlesniak@surgery.bsd.uchicago.edu
Original manuscript submitted: 11/24/06
Manuscript accepted: 02/03/07
Previously published online as a Cancer Biology & Therapy E-publication:
http://www.landesbioscience.com/journals/cbt/article/3957
KEY WORDS
adenovirus, CXCR4, glioma, midkine,
survivin, promoter
ACKNOWLEDGEMENTS
This study was supported by a grant from the
National Institute of Neurological Disorders
and Stroke (NINDS/NIH) 5K08NS046430
(M.L.) and the American College of Surgeons
(M.L.).
ABSTRACT
Objective: Transcriptional targeting is a key strategy to enhance therapeutic efficacy
of gene therapy applications. In the context of oncolytic virotherapy, transcriptional
promoter elements are used from genes that are over expressed in a variety of malignant
cancers. In the present study, we examined the feasibility of transcriptional targeting to
glioma cells by comparing the activity of survivin, midkine, and CXCR4 tumor‑specific
promoters.
Methods: To evaluate the expression level of several glioma related genes, we
performed quantitative RT‑PCR analyses on samples obtained from cell lines and patients.
To determine specific level of gene expression mediated by selective promoter elements,
we measured luciferase expression in glioma samples transduced with replication
deficient adenoviral vectors. Finally, we incorporated the optimal promoters into a con‑
ditionally replicative adenoviral vector, CRAd‑5/3, and examined the cytopathic effect
in vitro.
Results: The survivin promoter demonstrated the highest level of mRNA expression in
primary tumor samples and cell lines. Transcriptional targeting was confirmed by infec‑
tion of glioma cells with an adenovirus expression vector containing a survivin‑driven
luciferase reporter gene. Of the tested promoters, minimal level of survivin activity was
detected in normal human liver and brain. A novel vector, CRAd‑survivin5/3, with E1a
under the control of the survivin promoter, exhibited enhanced cytopathic effect in vitro.
Conclusions: Our data demonstrate that the survivin promoter element is very active
in glioma samples and has low activity in normal human brain and liver. A novel
oncolytic virus, CRAd‑survivin‑5/3, was effective against a panel of glioma cell lines
in vitro. Our results suggest that employing the survivin promoter element in the context
of CRAd‑5/3 may present a new opportunity for the development of glioma specific
oncolytic vectors.
INTRODUCTION
Glioblastoma multiforme (GBM) represents one of the most aggressive forms of
primary brain tumors. Even after clinically available treatments, including surgery,
radiotherapy, and chemotherapy, the median survival remains less than two years.1
Clearly, novel therapies for brain cancer are required in order to make an impact on this
devastating disease. Gene therapy and virotherapy constitute a novel therapeutic approach
for the treatment of glioblastoma. In gene therapy approaches, a therapeutic gene for
mutation compensation, immunopotentiation, or prodrug activation is transferred.2-7
In virotherapy, tumor cell killing is achieved by oncolysis; that is, virus replication
induced cell killing.8-10 Both of these therapeutic interventions allow for specific anti-
tumor effects via molecular targeting strategies that exploit tumor markers.
At present, one of the most promising gene delivery vehicles is the recombinant adeno-
viral (Ad) vector.9,11 Whereas adenoviral vectors are understood to exhibit superior levels
of in vivo gene transfer compared to some of the available alternative vector systems, their
present level of efficiency in clinical trials may nonetheless be suboptimal for cancer gene
therapy and virotherapy applications. Various approaches have been developed to enhance
the transcription selectivity of current vector systems for tumor cells, thereby limiting
ectopic expression in non-tumor cells and treatment-associated toxicities. Transcriptional
targeting strategies employ the use of tissue specific promoters (TSP) to restrict transgene
expression or viral replication to tumor cells. The ideal TSP for glioma would exhibit
the widest differential between ‘tumor on/brain and liver offexpression profiles, which
is fundamental for ablation of normal brain toxicity and potential liver toxicity from
ectopically localized adenovirus.
www.landesbioscience.com Cancer Biology & Therapy 679
©2007 LANDES BIOSCIENCE. DO NOT DISTRIBUTE.
Glioma Promoters for Transcriptional Targeting
Several candidate TSP genes have been found to be upregulated
in glial tumors, specifically: the chemokine C-X-C motif receptor 4
(CXCR4), the inhibitor and apoptosis protein (IAP) survivin, and the
growth factor midkine.9,12-14 Mutations in these genes play a major
role in cancer by producing anti-apoptotic activity or enhancing
survival or growth signals. To combine the benefits of adenoviral gene
therapy and specificity of these TSPs for glioma, we designed repli-
cation deficient adenoviral vectors that incorporate these promoters
upstream of the luciferase reporter gene. These viruses were tested in
vitro in glioma cell lines as well as in primary human glioma tissue.
The viruses were also tested in normal human brain and liver.
Although we have previously shown the utility of survivin in glioma
gene therapy,15 the three different promoters have not been system-
atically compared and explored in glioma. Our study thus provides
valuable new information for glioma adenoviral based gene therapy
and virotherapy with respect to the most efficient transcriptional
targeting strategy. Moreover, it provides a compelling argument for
further preclinical development of CRAd-survivin-5/3, a novel onco-
lytic virus that utilizes the survivin promoter and contains a chimeric
fiber protein encompassing adenovirus serotype 3 knob, which binds
to CD46 expressed on malignant glioma.16
MATERIALS AND METHODS
Cell lines and tissue specimens. Human glioma tumor cell
lines A172, U87MG, U373MG and U118MG were purchased
from American Type Culture Collection (ATCC, Manassas, VA).
Kings and No. 10 cells were purchased from the Japanese Tissue
Tumor Bank (Tokyo, Japan). A172, U87MG, U373MG and
U118MG cells were maintained in minimal essential medium
(MEM) (Mediatech, Herndon, VA) supplemented with 10% (v/v)
fetal bovine serum (FBS, HyClone, USA), 2 mM L-glutamine,
penicillin (100 IU/ml), and streptomycin (100 mg/ml). Kings
and No. 10 cells were maintained in RPMI medium (Gibco, Life
Technologies, USA) supplemented with 10% (v/v) FBS, 2 mM
L-glutamine, and penicillin/streptomycin. All cells were incubated at
37˚C in an atmosphere containing 5% CO2.
Human brain tumor specimens were obtained from patients
diagnosed with glioblastoma multiforme (WHO grade IV) under an
Institutional Review Board approved protocol. Primary samples were
reviewed by a pathologist to confirm that all tumors were GBMs.
A total of five GBM tumor specimens designated as T5, T13, T15,
T16 and T17 were used.
Human brain and liver samples (n = 3) were obtained from
patients undergoing a craniotomy or after liver transplantation.
All specimens obtained from patients were immediately used in
the experiments. To generate slice organ cultures, the tissue was
serially dissected into 0.5 mm-thick slices using the Krumdieck tissue
slicer17 (Alabama Research Development, Munford, AL). Next, the
tissue was cultured in 6-well plates in RPMI medium supplemented
with 10% FBS, 100 U/ml penicillin, 100 mg/ml streptomycin, and
5 mg/ml insulin. Cultures were maintained at 37˚C in a humidified
atmosphere of 95% air and 5% CO2 under continuous shaking.
Three tissue slices were examined per group.
Adenoviral vectors. The following recombinant adenoviral (Ad)
vectors were constructed in Division of Human Gene Therapy
(UAB) and used in this study: reAdGL3CMV, reAdGL3MK,
reAdGL3Survivin, and reAdGL3CXCR4. These vectors contain
the pGL3 plasmid Ad backbone and contain a luciferase expression
cassette under the human cytomegalovirus (CMV), midkine (MK),
survivin (SURV), or chemokine C-X-C motif receptor 4 (CXCR4)
promoter.18
The AdWT, CRAd-5/3 and CRAd-survivin-5/3 replication-
competent vectors were described before.19,20 CRAd-CXCR4-5/3
was created according to the scheme utilized for the production of
CRAd-survivin-5/3 vector, but instead of a shuttle plasmid carrying
the survivin promoter, we utilized a shuttle plasmid containing
CXCR4. All vectors were rescued in A549 cells. Viral particles were
purified on CsCl gradients by standard methods, and the concen-
tration of viral particles was determined by measuring absorbance
at 260 nm using a conversion factor of 1.1 x 1012 viral particles/
absorbance unit.
Isolation of RNA and real‑time quantitative RT‑PCR. Total
cellular RNA was extracted from cells with the RNeasy mini prep
kit (Qiagen, Ca, USA) and treated with DNAase I (Gibco, Life
Technologies, USA) for 30 min. RT-PCR products from midkine,
CXCR4, CMV and SURV were used for standard curve. RNA (1 mg)
isolated from samples was used in a one-step reverse transcription
PCR. The reaction was done using Gene Amp RNA PCR core kit
(Applied Biosystems, Inc., Foster City, CA). All primers and probe
set were design by the Primer Express software and synthesized by
Applied Biosystems, Inc. The primer and probe sequences used to
amplify and detect transcript were as follows (5'-3'): SURV sense
TGG AAG GCT GGG AGC CA; SURV antisense, GAA AGC
GCA ACC GGA CG; probe 6FAM-TGA CGA CCC CAT AGA
GGA ACA TAA AAA GCA T-TAMRA. Oligonucleotides for ampli-
fication CXCR4 gene were (5'-3'): CXCR4 sense, CTT CCC TTC
TGG GCA GTTGA; CXCR4 antisense, ACA TGG ACT GCC
TTG CAT AGG; and probe 6FAM-CCG TGG CAA ACT GGT
ACT TTG GGA ACT-TAMRA. The sequences to amplify midkine
were forward primer CAA TGC TCA GTG CCA GGA GA, reverse
primer TGG CTT TGG CCT TTG CTT T and probe 6FAM-CAT
CCG CGT CAC CAA GCC CTG-TAMRA. The primers to amplify
CMV gene were described before.18 A standard curve was generated
using serial 10-fold dilutions for quantification by reverse transcrip-
tion-PCR (RT-PCR). The PCR reaction included denaturation
(94˚C, 5 min) followed by 29 cycles, each consisting of denaturation
(94˚C, 1 min), annealing (60˚C, 1 min), and extension (72˚C,
2 min) with a final extension phase (10 min). The PCR reaction was
performed on a LightCycler system (Roche Molecular Biochemicals,
Model 2400). Reactions were incubated at 50˚C for 2 minutes,
60˚C for 30 minutes, 95˚C for 5 minutes, and then subjected to
40 cycles at the following conditions: 94˚C for 20 seconds and 60˚C
for 1 minute. Data was analyzed with LightCycler software. Each
data point was repeated two times. Quantitative values were obtained
from the threshold PCR cycle number (Ct), where the increase in
signal associated with an exponential growth of PCR product became
detectable. The relative mRNA levels in each sample were normalized
to GAPDH content.
To detect E1A expression, cells were cultured and infected as
previously described.21 At either 24 or 48 hours post infection, cell
culture media was removed, and the remaining cells were collected
by scraping into PBS. This solution was centrifuged, and supernatant
was removed. RNA obtained from the pelleted cells was stabilized
immediately using RNAlater reagent (Qiagen) and stored at -80˚C
until extraction. RNA was isolated using the standard protocol
provided by Qiagen for RNAeasy isolation. Contaminating DNA
was removed by DNase digestion (Qiagen). TaqMan primers and
probes were designed by Primer Express 1.0 software and synthesized
by Applied Biosystems. The sequences to amplify adenovirus genes
680 Cancer Biology & Therapy 2007; Vol. 6 Issue 5
©2007 LANDES BIOSCIENCE. DO NOT DISTRIBUTE.
Glioma Promoters for Transcriptional Targeting
were as follows: E1A forward, 5'-AACCAGTTGCCGTGAGAGT
TG-3'; E1A reverse, 5'-CTCGTTAAGCAAGTCCTCGATACA-3';
E1A probe, 5'-CACAGCCTGGCGACGCCCA-3'.21 All real-time
PCR reactions were performed under the same conditions as
described above.
Luciferase assays. For luciferase assay, 5 x 104 cells/well were
seeded onto 24-well plates 24 hours before infection. The next day,
cells were infected with adenovirus vectors at 100 vp/cell or mock
infected for 1 hour at 37˚C in fresh medium. After infection, the
cells were washed and fresh medium was added. At 48 hours after
infection, the medium was carefully removed and the cells were
harvested and luciferase activity was assayed in the cell lysates using a
standard luciferase expression protocol (Promega Wisconsin, USA).
Luciferase expression detected in glioma cells was normalized to
activity of human CMV promoter. To detect luciferase activity of
adenovectors in human samples, each group of three slices containing
a 250 mm piece of tissue was infected with 500 vp/cell by adding
virus in media containing 2% FBS. After 1 hour of virus infection,
the media was aspirated and media containing 10% FBS was added.
Forty-eight hours later, the tissues were harvested and luciferase
activity was assayed in the tissue homogenates using a standard
luciferase expression protocol presented by vendor (Promega). All
results are presented as ratio of luciferase activity per mg of total
protein. Data was recorded using a Modulus Luminometer (Turner
Biosystems, USA).
Detection of CRAd induced cytotoxicity. The in vitro cyto-
toxic effect of AdWT, CRAd-5/3, CRAd-CXCR4-5/3 and
CRAd-SURV-5/3 vectors was evaluated in a panel of glioma cell
lines. U373MG, U87MG, No.10 and U118MG cells were plate
in 24 well plate at a density of 50,000 cells per well. Twenty-four
hours later, the cells were infected for 1 hour with 300 mL of infec-
tion medium containing Ad vectors at doses of 1000, 100, 10, 1 and
0.1 vp per cell. Then the infection medium was replaced with the
appropriate complete medium. After 10 days, the cells were fixed
with 10% formaldehyde and stained with 1% crystal violet in 70%
ethanol solution.
Statistical analysis. All experimental determinations were
performed in triplicate. The CMV, CXCR4, SURV and MK mRNA
activity in tumors vs. normal tissue was compared using the Students
t test. In all analyses, p < 0.05 was considered statistically significant.
RESULTS
Survivin, CXCR4 and midkine mRNAs are overexpressed in
brain tumors compared to normal tissue. To investigate expression
level of the CXCR4, midkine, and survivin transcripts, we designed
specific real-time quantitative RT-PCR primers and probes recog-
nizing the expressed sequences of the human mRNAs. As shown in
Figure 1A, quantitative RT-PCR analysis of target transcripts in six
glioma cell lines of different morphological types revealed that all of
the glioma cell lines tested expressed detectable levels of CXCR4,
midkine, and survivin mRNAs. Among them, two glioma cell lines
(Kings and No. 10) demonstrated nearly the same pattern of expres-
sion characteristic of normal human brain tissue. In addition, four
of the six cell lines (A172, No. 10, Kings and U118MG) expressed
similar levels of midkine mRNA expression. In contrast to these
results, three glioma cell lines (U87MG, U118MG and U373MG)
showed increased expression of CXCR4 mRNA (26.97-fold to
63.21-fold) and in particular, survivin mRNA (158-fold to 672-fold)
as compared to normal human brain tissue (p < 0.05). The U87MG
and U373MG cell lines also showed significantly decreased expres-
sion of midkine mRNA (8.4 and 5.5-fold, p < 0.05) compared to
normal human brain tissue.
To detect expression of CXCR4, midkine, and survivin mRNAs
in primary tissues, we isolated total RNA from five GBM patients.
Quantitative RT-PCR revealed inter-individual variation in expres-
sion of all target mRNAs in these tissues (Fig. 1B). However, survivin
mRNA was significantly over-expressed in all primary gliomas tested
compared to matched normal human brain tissue (p < 0.05). Indeed,
the median difference in expression of survivin mRNA between
tumors and matched normal controls was 10,000-fold increased.
Four of five tumors samples analyzed also showed increased amounts
of CXCR4 mRNA (10–1000-fold, p < 0.05) compared to normal
human brain sample.
Level of luciferase expression driven by midkine, CXCR4 and
survivin promoters correlates with mRNA expression. The tran-
scriptional activities of CXCR4, survivin, and midkine promoters
driving luciferase expression in recombinant Ad vectors were evalu-
ated in the glioma cell lines (Fig. 2). In this experiment, the cell lines
were infected with the relevant adenoviral vectors of 100 vp/cell.
Figure 1. Results of survivin, CXCR4, and midkine mRNA expression in
glioma cell lines (A) and primary tumors (B). cDNAs were prepared from
total RNA extracted from each sample. The expression levels were deter‑
mined by quantitative RT‑PCR using equal amount of cDNA. Results of mRNA
expression were normalized to the GAPDH. All samples were determined in
duplicates twice (p < 0.05).
www.landesbioscience.com Cancer Biology & Therapy 681
©2007 LANDES BIOSCIENCE. DO NOT DISTRIBUTE.
Glioma Promoters for Transcriptional Targeting
AdCMVLuc was used to standardize for varying transduction
efficiencies between cell lines, and promoter activities were there-
fore plotted as percentage of cytomegalovirus (% CMV activity)
driven luciferase expression. In the glioma cell lines tested, each of
the candidate promoters demonstrated variable relative luciferase
activity between the different cell lines. The Ad vector containing
the survivin promoter, however, showed a higher relative luciferase
activity compared to the other candidate promoters in all six cell lines
tested. These results suggest that the survivin promoter is promising
for transcriptional targeting in glioma cell lines.
In primary slices (Fig. 2B), the survivin-driven vector demon-
strated the highest level of luciferase expression. These results support
the data obtained from mRNA expression in passaged cell lines.
Status of survivin, CXCR4 and midkine expression in normal
tissues. A key limitation for the use of a systemic adenoviral gene
therapy or virotherapy is the potential toxicity to non-target organs.
To investigate the specificity of expression of these candidate gene
promoters in glioma, we next compared the mean level of gene
expression in human liver, given the tropism of adenovirus for this
organ.22 Each of the candidate promoters was assessed for transgene
expression in fresh-cut human liver tissue and normal brain slices.
As shown in Figure 3A, transgene expression induced by CXCR4
promoter was similar in liver to that induced by the CMV promoter.
Of note, the survivin and the midkine promoters demonstrated the
lowest transgene expression (<1% of CMV activity).
To further evaluate the specificity of the candidate promoters
for potential use in localized gene therapy, normal human brain
(Fig. 3B) was infected with the different transcriptionally targeted
adenoviral vectors. In this experiment, tissue slices were transduced
with the Ad vectors and 48 hours later, transduction levels were
detected by luciferase expression. These results indicate that the
expression levels for all tested promoters were less than 2% relative
to the CMV promoter in normal brain tissues. These data underline
the previous results indicating that these promoters are a promising
selective tool for transcriptional targeting of glioma.
CRAds induce cytopathic effect in human glioma cells propor‑
tionate to the promoter activity detected by qRT‑PCR. Up to
this point, our results suggested that the survivin promoter may be
an optimal TSP for glioma therapy, with CXCR4 showing some
evidence of favorable activity. Based on these findings, we incorpo-
rated either the survivin or CXCR4 promoter into a novel oncolytic
vector, CRAd-5/3. This is a chimeric vector which contains the Ad5
backbone and Ad3 knob and binds to CD46 which is expressed on
malignant brain tumors.16 To investigate whether human survivin or
Figure 2. Transcriptional activity of promoter units detected by luciferase
assay. Human glioma cell lines (A) or tumor slices obtained from patient
specimen (B) were infected with replication deficient adenoviruses. Forty‑eight
hours later, luciferase detection was performed. Results are presented as
percent of CMV activity.
Figure 3. Ex vivo analysis of adenovirus replication in human liver slices (A)
and normal human brain (B). Tissue slices were incubated with 500 vp/slice
of each virus for 1 hr and then fresh media was added. Forty‑eight hours
later, luciferase expression was measured. Each point represents the mean
of triplicates from two independent experiments. Values for each vector are
as percent of CMV activity ± SD from three experiments.
682 Cancer Biology & Therapy 2007; Vol. 6 Issue 5
©2007 LANDES BIOSCIENCE. DO NOT DISTRIBUTE.
Glioma Promoters for Transcriptional Targeting
CXCR4 promoter can accelerate a cytopathic effect (CPE) in infected
tumor cells, CPE assays were done on U373MG, No. 10, U118MG,
U87MG human glioma cells. As shown in Figure 4, CRAd-SURV-5/3
infected U373MG and U87MG cells were completely lysed at
a 1000 vp/cell whereas No.10 and U118MG cells were killed at
100 vp/cell. In U373MG cells, killing activity was observed only for
AdWT and CRAd-CXCR4-5/3 vectors. Our results demonstrate
that incorporation of promoter elements into adenoviral genome
increases transcriptional control of E1A adenoviral region and
induces oncolysis.
Next, we infected U373MG, No.10, U118MG and U87MG
human glioma cells with AdWT, CRAd-5/3, CRAd-CXCR4-5/3
or CRAd-SURV-5/3 at 10 vp/cell. Forty-eight hours post-
infection, quantitative RT-PCR analysis demonstrated that the
CRAd-SURV-5/3 vector exhibited superior expression of E1A copy
numbers in U118MG (775.28 ± 12.57 fold), U87MG (454.3 ±
23.61 fold) and No.10 (557.8 ± 59.45) infected cells vs. AdWT
(p < 0.05, Fig. 5). This result indicates that combination of trans-
ductional approach (3 knob modification) and a transcriptional
modification (incorporation of survivin promoter) results in both
enhanced replication and enhanced cells lysis.
DISCUSSION
The therapeutic efficacy of virotherapy relies on a vector’s ability
to successfully target, transduce, and replicate in GBM. In order to
specifically replicate in tumor tissue, the adenovirus must posses a
TSP element in its genome which responds to the specific cellular
cues of tumor cells to mediate its replication. Unfortunately, the
number of available promoters with glioma specific activation
properties is very limited. While a number of TSP elements have
been tested in the context of GBM, there has been no comparative
analysis of these promoters in order to determine the optimal agent
for further preclinical and clinical development. This gap in knowl-
edge prompted us to compare three of the most widely utilized TSP
in glioma-CXCR4, midkine, and survivin.
CXCR4 is a chemokine which was originally described in the
context of providing migrational clues for leukocytes. However,
CXCR-4 has been found to be upregulated on malignant glioma.9,23-26
In a seminal article, Zhou et el., showed that CXCR4 on glioma
lines is a signaling receptor in that its agonist, stromal cell-derived
factor-1 (SDF-1; CXCL12), produced rapid phosphorylation of
mitogen-activated protein kinases.9 Furthermore, SDF-1 induced the
phosphorylation of Akt (protein kinase B), a kinase associated with
survival, and prevented the apoptosis of glioma cells. Most recently,
Ehtesham et al., have shown that invasive populations of glioma
cells overexpress CXCR4 at the message and protein levels, and that
this expression ranges from 25- to 89-fold higher than that found in
noninvasive tumor cells.25 Furthermore, neutralization of CXCR4
significantly impaired the in vitro invasive capacity of malignant glial
cells. Taken together, these findings underscore the importance of
CXCR4 as a potential therapeutic target for the treatment of invasive
glioblastoma.
The midkine promoter has been widely evaluated as a valuable tool
for cancer gene therapy as this regulatory sequence has been shown
selectively active in many malignancies including gliomas.27-34 The
activity of this promoter might be down-regulated by p53-dependent
pathways but further evidence is needed to elucidate the mechanism
of this phenomenon.32 In the case of glioma, midkine promoter
activity was shown to be two times higher in midkine-positive cells
than in midkine-negative primary normal brain cells.35 For this
reason, a midkine promoter-based CRAd (Ad-MK) was developed.27
This vector showed strong oncolytic activity in midkine-positive
glioma cells but did not exhibit cytotoxicity in midkine-negative
primary normal brain cells. When tested in an in vivo experiment,
Ad-MK eradicated midkine-positive glioma xenografts.
Finally, survivin is a novel member of the inhibitor-of-apoptosis
family which appears to be inappropriately expressed in human
cancers. The survivin promoter appears to be a promising tumor-
specific promoter in the context of human melanoma, breast cancer,
and glioma.27,36-38 Most recently, survivin has been found to be over-
expressed in up to 79% of astrocytic tumors.39-41 The expression of
this gene correlates with grade and is present in 90% of glioblastomas.
The activity of this promoter is enhanced by hypoxia,42 commonly
found in rapidly growing tumors like high grade gliomas. Survivin
seems to play an important role in the oncogenesis and progression
of these tumors.39,43,44 This is suggested by its expression pattern and
by the fact that patients with survivin-positive astrocytic tumors have
significantly shorter overall survival times compared with patients
who have survivin-negative tumors.39 The promoter has already
been proven effective for transgene expression in lung cancer.45 We
have recently shown that the incorporation of this promoter into the
adenoviral E1a region is responsible for enhanced viral replication
and an enhanced oncolytic effect in malignant glioma.15
The results of our comparative study indicate that of the tested
TSPs, the human survivin promoter is elevated in both passaged
glioma cells lines as well as primary tumor tissue. In fact, in three
of the six passaged glioma cells lines, we observed at least a 1,000
fold increase in survivin mRNA expression vs. normal human brain.
Similar results were observed in primary tissue, where the level of
survivin mRNA expression was 10,000 fold increased over normal
human brain. When utilized as a TSP, survivin increased transgene
Figure 4. Cell killing of human glioma cells by CRAd‑CXCR4‑5/3 and
CRAd‑SURV‑5/3 vectors. No.10, U373MG, U87MG or U118MG human
glioma cells were infected with AdWT, CRAd‑5/3, CRAd‑CXCR4‑5/3 or
CRAd‑SURV‑5/3 at indicated vp/cell. Noninfected cells served as control.
Ten days later, cells were stained with crystal violet.
www.landesbioscience.com Cancer Biology & Therapy 683
©2007 LANDES BIOSCIENCE. DO NOT DISTRIBUTE.
Glioma Promoters for Transcriptional Targeting
expression in all of the passaged (at least 10-fold) and primary glioma
(10–100 fold) tumor tissue. This finding was significant not only
with respect to control but also with respect to CXCR-4 and midkine
promoters. This study clearly establishes survivin as an optimal TSP
for glioma gene therapy.
Of note, we also examined the specificity of our promoters in
the context of human liver and normal brain. Adenoviral vectors
exhibit enhanced tropism for liver tissue, with hepatotoxicity
being a limiting side-effect of adenoviral based gene therapy.46 We
detected very low levels of survivin promoter activity in normal liver
(<1% of CMV activity) and human brain (<1% CMV activity).
Taken together, these studies establish the specificity of survivin
for malignant brain tumors and suggest a preclinical safety profile
which warrants further analysis and testing of these promoters in
in vivo experimental schemas. The improved efficacy of transgene
expression in malignant brain tumors along with improved safety
index in normal human liver and brain validate the use of survivin
in transcriptional targeting strategies to restrict transgene expression
and viral replication to tumor cells.
In conclusion, we have performed the first comparative study of
three promoters which are widely utilized in glioma gene therapy.
Our results suggest that the survivin promoter is an ideal TSP for
development in both gene therapy and virotherapy glioma clinical
trials. A combination strategy involving transcriptional and trans-
ductional modification of an adenovirus led us to the development
of a novel oncolytic vector, CRAd-SURV-5/3. When tested against a
panel of glioma cell lines, the virus preferentially killed glioma tumor
cells in vitro and provided a “proof-of-principle” with respect to the
use of TSP promoter elements in glioma gene therapy.
References
1. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K,
Brandes AA, Marosi C, Bogdahn U, Curschmann J, Janzer RC, Ludwin SK, Gorlia T,
Allgeier A, Lacombe D, Cairncross JG, Eisenhauer E, Mirimanoff RO. Radiotherapy
plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005;
352:987-96.
2. Lun X, Yang W, Alain T, Shi ZQ, Muzik H, Barrett JW, McFadden G, Bell J, Hamilton
MG, Senger DL, Forsyth PA. Myxoma virus is a novel oncolytic virus with significant
antitumor activity against experimental human gliomas. Cancer Res 2005; 65:9982-90.
3. Tai CK, Wang WJ, Chen TC, Kasahara N. Single-shot, multicycle suicide gene therapy by
replication-competent retrovirus vectors achieves long-term survival benefit in experimental
glioma. Mol Ther 2005; 12:842-51.
4. Chiu CC, Kang YL, Yang TH, Huang CH, Fang K. Ectopic expression of herpes simplex
virus-thymidine kinase gene in human non-small cell lung cancer cells conferred caspase-ac-
tivated apoptosis sensitized by ganciclovir. Int J Cancer 2002; 102:328-33.
5. Nafe C, Cao YJ, Quinones A, Dobberstein KU, Kramm CM, Rainov NG. Expression of
mutant non-cleavable Fas ligand on retrovirus packaging cells causes apoptosis of immuno-
competent cells and improves prodrug activation gene therapy in a malignant glioma model.
Life Sci 2003; 73:1847-60.
6. Cowen RL, Williams JC, Emery S, Blakey D, Darling JL, Lowenstein PR, Castro MG.
Adenovirus vector-mediated delivery of the prodrug-converting enzyme carboxypeptidase
G2 in a secreted or GPI-anchored form: High-level expression of this active conditional
cytotoxic enzyme at the plasma membrane. Cancer Gene Ther 2002; 9:897-907.
7. Manome Y, Wen PY, Dong Y, Tanaka T, Mitchell BS, Kufe DW, Fine HA. Viral vector
transduction of the human deoxycytidine kinase cDNA sensitizes glioma cells to the cyto-
toxic effects of cytosine arabinoside in vitro and in vivo. Nat Med 1996; 2:567-73.
8. Conrad C, Miller CR, Ji Y, Gomez-Manzano C, Bharara S, McMurray JS, Lang FF, Wong
F, Sawaya R, Yung WK, Fueyo J. Delta24-hyCD adenovirus suppresses glioma growth in
vivo by combining oncolysis and chemosensitization. Cancer Gene Ther 2005; 12:284-94.
9. Zhou Y, Larsen PH, Hao C, Yong VW. CXCR4 is a major chemokine receptor on glioma
cells and mediates their survival. J Biol Chem 2002; 277:49481-7.
10. Tyminski E, Leroy S, Terada K, Finkelstein DM, Hyatt JL, Danks MK, Potter PM, Saeki
Y, Chiocca EA. Brain tumor oncolysis with replication-conditional herpes simplex virus
type 1 expressing the prodrug-activating genes, CYP2B1 and secreted human intestinal
carboxylesterase, in combination with cyclophosphamide and irinotecan. Cancer Res 2005;
65:6850-7.
11. Douglas JT, Kim M, Sumerel LA, Carey DE, Curiel DT. Efficient oncolysis by a replicating
adenovirus (ad) in vivo is critically dependent on tumor expression of primary ad receptors.
Cancer Res 2001; 61:813-7.
Figure 5. Selective replication of CRAd‑CXCR4‑5/3 and CRAd‑SURV‑5/3 in vitro. U373MG, U118MG, U87MG and No. 10 glioma cells were infected
with AdWT, CRAd‑5/3, CRAd‑CXCR4‑5/3, or CRAd‑SURV‑5/3 at 10 vp/cell. After four hour adsorption, media was replaced and cell incubation continued
for next 24 or 48 hours. Replication activities were measured in triplicates by quantitative RT‑PCR with primers that recognize E1A viral region. All data are
presented as ratio between E1A copy number and human actin. (*p < 0.05).
684 Cancer Biology & Therapy 2007; Vol. 6 Issue 5
©2007 LANDES BIOSCIENCE. DO NOT DISTRIBUTE.
Glioma Promoters for Transcriptional Targeting
12. Rempel SA, Dudas S, Ge S, Gutierrez JA. Identification and localization of the cytokine
SDF1 and its receptor, CXC chemokine receptor 4, to regions of necrosis and angiogenesis
in human glioblastoma. Clin Cancer Res 2000; 6:102-11.
13. Oh JW, Drabik K, Kutsch O, Choi C, Tousson A, Benveniste EN. CXC chemokine receptor
4 expression and function in human astroglioma cells. J Immunol 2001; 166:2695-704.
14. Mishima K, Asai A, Kadomatsu K, Ino Y, Nomura K, Narita Y, Muramatsu T, Kirino T.
Increased expression of midkine during the progression of human astrocytomas. Neurosci
Lett 1997; 233:29-32.
15. Van Houdt WJ, Haviv YS, Lu B, Wang M, Rivera AA, Ulasov IV, Lamfers ML, Rein
D, Lesniak MS, Siegal GP, Dirven CM, Curiel DT, Zhu ZB. The human survivin pro-
moter: A novel transcriptional targeting strategy for treatment of glioma. J Neurosurg 2006;
104:583-92.
16. Ulasov IV, Tyler MA, Zheng S, Han Y, Lesniak MS. CD46 represents a target for adenoviral
gene therapy of malignant glioma. Hum Gene Ther 2006; 17:556-64.
17. Kirby TO, Rivera A, Rein D, Wang M, Ulasov I, Breidenbach M, Kataram M, Contreras JL,
Krumdieck C, Yamamoto M, Rots MG, Haisma HJ, Alvarez RD, Mahasreshti PJ, Curiel
DT. A novel ex vivo model system for evaluation of conditionally replicative adenoviruses
therapeutic efficacy and toxicity. Clin Cancer Res 2004; 10:8697-703.
18. Rein DT, Breidenbach M, Nettelbeck DM, Kawakami Y, Siegal GP, Huh WK, Wang M,
Hemminki A, Bauerschmitz GJ, Yamamoto M, Adachi Y, Takayama K, Dall P, Curiel DT.
Evaluation of tissue-specific promoters in carcinomas of the cervix uteri. J Gene Med 2004;
6:1281-9.
19. Ono HA, Davydova JG, Adachi Y, Takayama K, Barker SD, Reynolds PN, Krasnykh VN,
Kunisaki C, Shimada H, Curiel DT, Yamamoto M. Promoter-controlled infectivity-en-
hanced conditionally replicative adenoviral vectors for the treatment of gastric cancer. J
Gastroenterol 2005; 40:31-42.
20. Zhu ZB, Chen Y, Makhija SK, Lu B, Wang M, Rivera AA, Yamamoto M, Wang S, Siegal
GP, Curiel DT, McDonald JM. Survivin promoter-based conditionally replicative adenovi-
ruses target cholangiocarcinoma. Int J Oncol 2006; 29:1319-29.
21. Preuss MA, Lam JT, Wang M, Leath IIIrd CA, Kataram M, Mahasreshti PJ, Alvarez RD,
Curiel DT. Transcriptional blocks limit adenoviral replication in primary ovarian tumor.
Clin Cancer Res 2004; 10:3189-94.
22. Rots MG, Elferink MG, Gommans WM, Oosterhuis D, Schalk JA, Curiel DT, Olinga
P, Haisma HJ, Groothuis GM. An ex vivo human model system to evaluate specificity of
replicating and non-replicating gene therapy agents. J Gene Med 2006; 8:35-41.
23. Barbero S, Bajetto A, Bonavia R, Porcile C, Piccioli P, Pirani P, Ravetti JL, Zona G,
Spaziante R, Florio T, Schettini G. Expression of the chemokine receptor CXCR4 and its
ligand stromal cell-derived factor 1 in human brain tumors and their involvement in glial
proliferation in vitro. Ann N Y Acad Sci 2002; 973:60-9.
24. Barbero S, Bonavia R, Bajetto A, Porcile C, Pirani P, Ravetti JL, Zona GL, Spaziante R,
Florio T, Schettini G. Stromal cell-derived factor 1alpha stimulates human glioblastoma cell
growth through the activation of both extracellular signal-regulated kinases 1/2 and Akt.
Cancer Res 2003; 63:1969-74.
25. Ehtesham M, Winston JA, Kabos P, Thompson RC. CXCR4 expression mediates glioma
cell invasiveness. Oncogene 2006; 25:2801-6.
26. Woerner BM, Warrington NM, Kung AL, Perry A, Rubin JB. Widespread CXCR4 activa-
tion in astrocytomas revealed by phospho-CXCR4-specific antibodies. Cancer Res 2005;
65:11392-9.
27. Sadeghi H, Hitt MM. Transcriptionally targeted adenovirus vectors. Curr Gene Ther 2005;
5:411-27.
28. Yu L, Hamada K, Namba M, Kadomatsu K, Muramatsu T, Matsubara S, Tagawa M.
Midkine promoter-driven suicide gene expression and -mediated adenovirus replication pro-
duced cytotoxic effects to immortalised and tumour cells. Eur J Cancer 2004; 40:1787-94.
29. Yu L, Yamamoto N, Kadomatsu K, Muramatsu T, Matsubara S, Sakiyama S, Tagawa
M. Midkine promoter can mediate transcriptional activation of a fused suicide gene in a
broader range of human breast cancer compared with c-erbB-2 promoter. Oncology 2004;
66:143-9.
30. Miyauchi M, Shimada H, Kadomatsu K, Muramatsu T, Matsubara S, Ikematsu S, Takenaga
K, Asano T, Ochiai T, Sakiyama S, Tagawa M. Frequent expression of midkine gene in
esophageal cancer suggests a potential usage of its promoter for suicide gene therapy. Jpn J
Cancer Res 1999; 90:469-75.
31. Tomizawa M, Yu L, Wada A, Tamaoki T, Kadomatsu K, Muramatsu T, Matsubara S,
Watanabe K, Ebara M, Saisho H, Sakiyama S, Tagawa M. A promoter region of the midkine
gene that is frequently expressed in human hepatocellular carcinoma can activate a suicide
gene as effectively as the alpha-fetoprotein promoter. Br J Cancer 2003; 89:1086-90.
32. Yu L, Ugai S, J OW, Namba M, Kadomatsu K, Muramatsu T, Matsubara S, Sakiyama S,
Tagawa M. Cell growth- and P53-dependent transcriptional activity of the midkine pro-
moter confers suicide gene expression in tumor cells. Oncol Rep 2003; 10:1301-5.
33. Sakiyama S, Yu L, Tomizawa M, Shimada H, Kadomatsu K, Muramatsu T, Ikematsu S,
Nakagawara A, Tagawa M. Utilization of the promoter region of the midkine gene as a tool
to drive therapeutic genes in a tumor specific manner. Adv Enzyme Regul 2003; 43:57-66.
34. Adachi Y, Reynolds PN, Yamamoto M, Grizzle WE, Overturf K, Matsubara S, Muramatsu
T, Curiel DT. Midkine promoter-based adenoviral vector gene delivery for pediatric solid
tumors. Cancer Res 2000; 60:4305-10.
35. Kohno S, Nakagawa K, Hamada K, Harada H, Yamasaki K, Hashimoto K, Tagawa M,
Nagato S, Furukawa K, Ohnishi T. Midkine promoter-based conditionally replicative
adenovirus for malignant glioma therapy. Oncol Rep 2004; 12:73-8.
36. Zhu ZB, Makhija SK, Lu B, Wang M, Kaliberova L, Liu B, Rivera AA, Nettelbeck
DM, Mahasreshti PJ, Leath CA, Barker S, Yamaoto M, Li F, Alvarez RD, Curiel DT.
Transcriptional targeting of tumors with a novel tumor-specific survivin promoter. Cancer
Gene Ther 2004; 11:256-62.
37. Zhu ZB, Makhija SK, Lu B, Wang M, Rivera AA, Kim-Park S, Ulasov IV, Zhou F, Alvarez
RD, Siegal GP, Curiel DT. Incorporating the survivin promoter in an infectivity enhanced
CRAd-analysis of oncolysis and anti-tumor effects in vitro and in vivo. Int J Oncol 2005;
27:237-46.
38. Van Houdt W, Haviv YS, Lu B, Wang M, Rivera AA, Ulasov IV, Lamfers ML, Lesniak
MS, Siegal GP, Clemens MF, Curiel DT, Zhu ZB. The human survivin promoter is a novel
transcriptional targeting strategy for glioma. Journal of Neurosurgyer 2005.
39. Kajiwara Y, Yamasaki F, Hama S, Yahara K, Yoshioka H, Sugiyama K, Arita K, Kurisu K.
Expression of survivin in astrocytic tumors: Correlation with malignant grade and progno-
sis. Cancer 2003; 97:1077-83.
40. Yamada Y, Kuroiwa T, Nakagawa T, Kajimoto Y, Dohi T, Azuma H, Tsuji M, Kami K,
Miyatake S. Transcriptional expression of survivin and its splice variants in brain tumors in
humans. J Neurosurg 2003; 99:738-45.
41. Chakravarti A, Noll E, Black PM, Finkelstein DF, Finkelstein DM, Dyson NJ, Loeffler
JS. Quantitatively determined survivin expression levels are of prognostic value in human
gliomas. J Clin Oncol 2002; 20:1063-8.
42. Yang L, Cao Z, Li F, Post DE, Van Meir EG, Zhong H, Wood WC. Tumor-specific gene
expression using the survivin promoter is further increased by hypoxia. Gene Ther 2004;
11:1215-23.
43. Kleinschmidt-DeMasters BK, Heinz D, McCarthy PJ, Bobak JB, Lillehei KO, Shroyer
AL, Shroyer KR. Survivin in glioblastomas: Protein and messenger RNA expression and
comparison with telomerase levels. Arch Pathol Lab Med 2003; 127:826-33.
44. Das A, Tan WL, Teo J, Smith DR. Expression of survivin in primary glioblastomas. J Cancer
Res Clin Oncol 2002; 128:302-6.
45. Chen JS, Liu JC, Shen L, Rau KM, Kuo HP, Li YM, Shi D, Lee YC, Chang KJ, Hung MC.
Cancer-specific activation of the survivin promoter and its potential use in gene therapy.
Cancer Gene Ther 2004; 11:740-7.
46. Shayakhmetov DM, Gaggar A, Ni S, Li ZY, Lieber A. Adenovirus binding to blood factors
results in liver cell infection and hepatotoxicity. J Virol 2005; 79:7478-91.
www.landesbioscience.com Cancer Biology & Therapy 685
... Another targeting approach is by the insertion of tumor-specific promoters to drive specific viral replication in tumor cells and avoid toxicity to normal tissue [59]. Various promoter candidates have been applied to design tumor-specific promoter-driven OVs, including nestin, survivin, cyclooxygenase-2 (COX-2), C-X-C chemokine receptor type 4 (CXCR4), hypoxia inducible factor-1 (HIF-1) and telomerase [10,[60][61][62][63]. Considering the intertumoral heterogeneity in transcription profiles of GBM, it would be expected that the response to such OVs might vary between GBM subtypes. ...
... Various promoter candidates have been applied to design tumor-specific promoter-driven OVs, including nestin, survivin, cyclooxygenase-2 (COX-2), C-X-C chemokine receptor type 4 (CXCR4), hypoxia inducible factor-1 (HIF-1) and telomerase [10,[60][61][62][63]. Considering the intertumoral heterogeneity in transcription profiles of GBM, it would be expected that the response to such OVs might vary between GBM subtypes. One could hypothesize that GBM with proneural features might be more sensitive to viruses targeting cells expressing neuronal progenitor genes (e.g., nestin), whereas tumors of mesenchymal subtype may be more sensitive to viruses in which replication is driven by the inflammation-activated COX-2 or CXCR4 promoter [62][63][64]. ...
Article
Full-text available
Simple Summary Glioblastoma (GBM) is the most frequent and aggressive primary brain tumor. Despite multimodal treatment, the prognosis of GBM patients remains very poor. Oncolytic virotherapy is being evaluated as novel treatment for this patient group and clinical trials testing oncolytic viruses have shown impressive responses, albeit in a small subset of GBM patients. Obtaining insight into specific tumor- or patient-related characteristics of the responding patients, may in the future improve response rates. In this review we discuss factors related to oncolytic activity of the most widely applied oncolytic virus strains as well as potential biomarkers and future assays that may allow us to predict response to these agents. Such biomarkers and tools may in the future enable personalizing oncolytic virotherapy for GBM patients. Abstract Oncolytic virus (OV) treatment may offer a new treatment option for the aggressive brain tumor glioblastoma. Clinical trials testing oncolytic viruses in this patient group have shown promising results, with patients achieving impressive long-term clinical responses. However, the number of responders to each OV remains low. This is thought to arise from the large heterogeneity of these tumors, both in terms of molecular make-up and their immune-suppressive microenvironment, leading to variability in responses. An approach that may improve response rates is the personalized utilization of oncolytic viruses against Glioblastoma (GBM), based on specific tumor- or patient-related characteristics. In this review, we discuss potential biomarkers for response to different OVs as well as emerging ex vivo assays that in the future may enable selection of optimal OV for a specific patient and design of stratified clinical OV trials for GBM.
... The identification of suitable tumor-upregulated promoters is a prerequisite for this approach. Although several tumor-upregulated promoters have been utilized for human cancer gene therapy, none have been tested in canine tumors [40,41,43,[55][56][57]. Given that many canine tumors are excellent models of their human counterparts, evaluation of these promoters in canine cells would provide valuable comparative information. ...
... Among the tested genes, survivin showed the highest level of endogenous activity. Elevated survivin expression in human and murine melanoma, mammary cancer, and lymphoma have been previously reported [40,41,55,58]. We have observed similar expression levels in canine cell lines. ...
Preprint
Full-text available
Gene therapy is a promising treatment option for cancer. However, its utility may be limited due to expression in off-target cells. Cancer-specific promoters such as telomerase reverse transcriptase (TERT), survivin, and chemokine receptor 4 (CXCR4) have enhanced activity in a variety of human and murine cancers, however, little has been published regarding these promoters in dogs. Given the utility of canine cancer models, the activity of these promoters along with adenoviral E2F enhanced E1a promoter (EEE) was evaluated in a variety of canine tumors, both from the endogenous gene and from exoge-nously administered constructs. Endogenous expression levels were measured for cTERT, cSurvivin, and cCXCR4 and were low for all three, with some non-malignant and some tumor cell lines and tissues expressing the gene. Expression levels from exoge-nously supplied promoters were measured by both the number of cells expressing the construct and the intensity of expression in individual cells. Exogenously supplied promoters were active in more cells in all tumor lines than in normal cells, with the EEE promoter being most active, followed by cTERT. The intensity of expression varied more with cell type than with specific promoters. Ultimately, no single promoter was identified that would result in reliable expression, regardless of the tumor type. Thus, these findings imply that identification of a pan-cancer promoter may be difficult. In addition, this data raises the concern that endogenous expression analysis may not accurately predict exogenous promoter activity.
... The identification of suitable tumor-upregulated promoters is a prerequisite for this approach. Although several tumor-upregulated promoters have been utilized for human cancer gene therapy, none have been tested in canine tumors [40,41,43,[55][56][57]. Given that many canine tumors are excellent models of their human counterparts, evaluation of these promoters in canine cells would provide valuable comparative information. ...
... Among the tested genes, survivin showed the highest level of endogenous activity. Elevated survivin expression in human and murine melanoma, mammary cancer, and lymphoma have been previously reported [40,41,55,58]. We have observed similar expression levels in canine cell lines. ...
Article
Full-text available
Gene therapy is a promising treatment option for cancer. However, its utility may be limited due to expression in off-target cells. Cancer-specific promoters such as telomerase reverse transcriptase (TERT), survivin, and chemokine receptor 4 (CXCR4) have enhanced activity in a variety of human and murine cancers, however, little has been published regarding these promoters in dogs. Given the utility of canine cancer models, the activity of these promoters along with adenoviral E2F enhanced E1a promoter (EEE) was evaluated in a variety of canine tumors, both from the endogenous gene and from exogenously administered constructs. Endogenous expression levels were measured for cTERT, cSurvivin, and cCXCR4 and were low for all three, with some non-malignant and some tumor cell lines and tissues expressing the gene. Expression levels from exogenously supplied promoters were measured by both the number of cells expressing the construct and the intensity of expression in individual cells. Exogenously supplied promoters were active in more cells in all tumor lines than in normal cells, with the EEE promoter being most active, followed by cTERT. The intensity of expression varied more with cell type than with specific promoters. Ultimately, no single promoter was identified that would result in reliable expression, regardless of the tumor type. Thus, these findings imply that identification of a pan-cancer promoter may be difficult. In addition, this data raises the concern that endogenous expression analysis may not accurately predict exogenous promoter activity.
... [65][66][67] Preclinically, combination of different types of stem cells and CRAd have been studied as experimental treatments of glioma; mesenchymal stem cell MSC-Delta24-RGD, 68 and MSC-ICOVIR17, 36 MSC-CRAd-CXCR4-5/3, 69 neural stem cells (NSC)-CRAd-survivin-pk7 69-72 demonstrated antiglioma activity. CRAd-CXCR4-5/3 and CRAd survivin-pk7 are transcriptionally targeted CRAd using tumor-specific C-X-C chemokine receptor 4 (CXCR4) and survivin promotor, respectively, to control E1A expression and target the viruses to GBM. 73,74 CRAd-CXCR4-5/3 has a chimeric Ad 5/3 fiber (chimera of the shaft of human adenovirus type 5 and the knob of human adenovirus type 3) that targets CD46 or CD80/86 cellular receptors and exhibits increased transduction of malignant glioma compared with wild-type Ad5. 73,75 CRAd-Survivin-pk7 has fiber modification with pk7 encoding polylysine that was designed to increase transduction of malignant glioma by direct binding to heparan sulfate and polyanionic cellular receptors 74,76,77 Currently, a Phase I trial is ongoing in the US (NCT03072134) in which NSC loaded with CRAd-survivin-pk7 are injected into the resection cavity of newly diagnosed GBM. ...
... CRAd-CXCR4-5/3 and CRAd survivin-pk7 are transcriptionally targeted CRAd using tumor-specific C-X-C chemokine receptor 4 (CXCR4) and survivin promotor, respectively, to control E1A expression and target the viruses to GBM. 73,74 CRAd-CXCR4-5/3 has a chimeric Ad 5/3 fiber (chimera of the shaft of human adenovirus type 5 and the knob of human adenovirus type 3) that targets CD46 or CD80/86 cellular receptors and exhibits increased transduction of malignant glioma compared with wild-type Ad5. 73,75 CRAd-Survivin-pk7 has fiber modification with pk7 encoding polylysine that was designed to increase transduction of malignant glioma by direct binding to heparan sulfate and polyanionic cellular receptors 74,76,77 Currently, a Phase I trial is ongoing in the US (NCT03072134) in which NSC loaded with CRAd-survivin-pk7 are injected into the resection cavity of newly diagnosed GBM. ...
Article
Full-text available
Replication conditional oncolytic human adenovirus has long been considered a promising biological therapeutic to target high-grade gliomas (HGG), a group of essentially lethal primary brain cancer. The last decade has witnessed initiation and some completion of a number of Phase I and II clinical investigations of oncolytic adenovirus for HGG in the US and Europe. Results of these trials in patients are pivotal for not only federal approval but also filling an existing knowledge gap that primarily derives from the stark differences in permissivity to human adenovirus between humans and preclinical mouse models. DNX-2401 (Delta-24-RGD), the current mainstream oncolytic adenovirus with modifications in E1A and the fiber, has been shown to induce impressive objective response and long-term survival (>3 years) in a fraction of patients with recurrent HGG. Responders exhibited initial enlargement of the treated lesions for a few months post treatment, followed by shrinkage and near complete resolution. In accord with preclinical research, post-treatment specimens revealed virus-mediated alteration of the immune tumor microenvironment as evidenced by infiltration of CD8+ T cells and M1-polarized macrophages. These findings are encouraging and together with further information from ongoing studies have a potential to make oncolytic adenovirus a viable option for clinical management of HGG. This review deals with this timely topic; we will describe both preclinical and clinical development of oncolytic adenovirus therapy for HGG, summarize updated knowledge on clinical trials and discuss challenges that the field currently faces.
... Lastly, Ulasov and colleagues generated a glioma-specific recombinant AdV, called CRAd-S-pk7, by modifying the Ad5 fiber with pk7s and by regulating the expression of the E1A gene via the human survivin promoter [339]. Building on encouraging preclinical results [340,341], CRAd-S-pk7 virus loaded onto neural stem cells was administered during surgery in newly diagnosed GBM patients, along with chemo-radiotherapy [281]. ...
Article
Full-text available
Simple Summary Glioblastoma (GBM) poses a formidable challenge as a central nervous system tumor with extremely limited responsiveness to conventional treatments. While immunotherapeutic approaches have shown success in treating other solid tumors, their effectiveness against GBM is limited. Our review systematically addresses the intrinsic features of GBM that hinder the success of both standard therapies and immunotherapies. Furthermore, we comprehensively analyze all the immune-based approaches currently undergoing clinical evaluation for GBM, both as standalone treatments and in combination with standard therapy or other immunotherapies. Abstract Despite decades of research and the best up-to-date treatments, grade 4 Glioblastoma (GBM) remains uniformly fatal with a patient median overall survival of less than 2 years. Recent advances in immunotherapy have reignited interest in utilizing immunological approaches to fight cancer. However, current immunotherapies have so far not met the anticipated expectations, achieving modest results in their journey from bench to bedside for the treatment of GBM. Understanding the intrinsic features of GBM is of crucial importance for the development of effective antitumoral strategies to improve patient life expectancy and conditions. In this review, we provide a comprehensive overview of the distinctive characteristics of GBM that significantly influence current conventional therapies and immune-based approaches. Moreover, we present an overview of the immunotherapeutic strategies currently undergoing clinical evaluation for GBM treatment, with a specific emphasis on those advancing to phase 3 clinical studies. These encompass immune checkpoint inhibitors, adoptive T cell therapies, vaccination strategies (i.e., RNA-, DNA-, and peptide-based vaccines), and virus-based approaches. Finally, we explore novel innovative strategies and future prospects in the field of immunotherapy for GBM.
... An HSV engineered to express tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) allowed targeting of a broader tumor cell population by combining the effect of direct HSV mediated oncolysis and TRAIL mediated apoptosis induction, irrespective of differential susceptibility of tumor cells to either modes of cell death [153]. Lastly, OVs that are engineered to exploit specific gene mutations like mutated P53, Ras and Rb genes in tumors [152] or use tumor specific promoters to enhance their tumor-selectivity may result in a sub-optimal therapeutic efficacy due to the diversity in the level of expression of these specific mutated genes in tumor cells [154][155][156]. ...
Preprint
Glioblastoma is one of the most difficult tumor types to treat with conventional therapy options like tumor debulking, chemo and radiotherapy. Immunotherapeutic agents like oncolytic viruses, immune checkpoint inhibitors and chimeric antigen receptor T cells have revolutionized cancer therapy, but their success in glioblastoma remains limited and further optimization of immunotherapies is needed. Several oncolytic viruses have demonstrated ability to infect tumors and trigger anti-tumor immune responses in malignant glioma patients. Leading the pack, oncolytic herpesvirus, first in its class, awaits an approval for treating malignant glioma from MHLW, the federal authority of Japan. Nevertheless, some major hurdles like the blood brain barrier, immunosuppressive tumor microenvironment, and tumor heterogeneity can engender suboptimal efficacy in malignant glioma. In this review, we discuss the current status of malignant glioma therapies with a focus on oncolytic viruses in clinical trials. Furthermore, we discuss the obstacles faced by oncolytic viruses in malignant glioma patients and strategies that are being used to overcome these limitations to 1) optimize delivery of oncolytic viruses beyond the blood brain barrier; 2) trigger inflammatory immune responses in and around tumors; and 3) use of multimodal therapies in combination to tackle tumor heterogeneity, with an end goal of optimizing the therapeutic outcome of oncolytic virotherapy.
... An HSV engineered to express tumor-necrosis-factor-related apoptosis-inducing ligand (TRAIL) allowed the targeting of a broader tumor cell population by combining the effect of direct HSV-mediated oncolysis and TRAIL-mediated apoptosis induction, irrespective of the differential susceptibility of tumor cells to either modes of cell death [166]. Lastly, OVs that are engineered to exploit specific gene mutations like mutated P53, Ras, and Rb genes in tumors [165] or that use tumor-specific promoters to enhance their tumor-selectivity may result in a sub-optimal therapeutic efficacy because of the diversity in the level of expression of these specific mutated genes in tumor cells [167][168][169]. ...
Article
Full-text available
Glioblastoma is one of the most difficult tumor types to treat with conventional therapy options like tumor debulking and chemo- and radiotherapy. Immunotherapeutic agents like oncolytic viruses, immune checkpoint inhibitors, and chimeric antigen receptor T cells have revolutionized cancer therapy, but their success in glioblastoma remains limited and further optimization of immunotherapies is needed. Several oncolytic viruses have demonstrated the ability to infect tumors and trigger anti-tumor immune responses in malignant glioma patients. Leading the pack, oncolytic herpesvirus, first in its class, awaits an approval for treating malignant glioma from MHLW, the federal authority of Japan. Nevertheless, some major hurdles like the blood–brain barrier, the immunosuppressive tumor microenvironment, and tumor heterogeneity can engender suboptimal efficacy in malignant glioma. In this review, we discuss the current status of malignant glioma therapies with a focus on oncolytic viruses in clinical trials. Furthermore, we discuss the obstacles faced by oncolytic viruses in malignant glioma patients and strategies that are being used to overcome these limitations to (1) optimize delivery of oncolytic viruses beyond the blood–brain barrier; (2) trigger inflammatory immune responses in and around tumors; and (3) use multimodal therapies in combination to tackle tumor heterogeneity, with an end goal of optimizing the therapeutic outcome of oncolytic virotherapy.
Chapter
In the past decade, gene- and cell-based therapies have been translated into clinical application despite enormous costs for the establishment of an efficient gene therapeutic regimen. Technological advances were made with regards to smart vector design serving targeted, efficient, regulated, and sustained gene expression without interference of the host immune response. Great efforts have been made to serve safe delivery of vector particles to specific tissues and cells in vivo. Intriguing technologies for editing genes and correcting inherited mutations in vivo based on artificial zinc-finger nucleases and CRISPR/Cas9 technology have been developed. Stem- and T-cell technologies have moved the entire field of regenerative medicine and targeted engagement of immune cells to fight cancer a significant step forward. The developments in various aspects of molecular imaging (MI) technologies have enabled not only the possibility to follow the dynamics of disease-specific molecular alterations in vivo but also to visualize the location and dynamics of the expression of transduced genes or gene-modified stem- or T-cells on a quantitative level in vivo. In the past 20 years, various MI techniques for safe, repeated, and high-resolution in vivo imaging of gene expression have been successfully implemented in animals and humans. The MI technologies have been mostly applied in the field of gene therapy of highly dynamic diseases, such as cancer. In this chapter, the basis of gene expression imaging is described and the different steps of a gene therapy protocol from gene transduction to assessment of therapy response are illustrated. Linking MI and gene therapy will help to design smart, efficient, and safe gene therapy protocols for human application. The chapter was phrased during the SARS-CoV-2 pandemic and aims to summarize the most important developments of MI for gene- and cell-based therapies in dedication to all those worldwide who suffered and fought COVID disease.
Article
Although the multi‐tyrosine kinase inhibitor, sorafenib is useful in the treatment of several cancers, cholangiocarcinoma (CCA) is refractory to this drug. Among other mechanisms of chemoresistance impaired uptake via hOCT1 (gene SLC22A1) has been suggested. Here we have investigated the events accounting for this phenotypic characteristic and have evaluated the interest of selective gene therapy strategies to overcome this limitation. Gene expression and DNA methylation of SLC22A1 were analyzed using intrahepatic (iCCA) and extrahepatic (eCCA) biopsies (Copenhagen and Salamanca cohorts; n=132) and TCGA‐CHOL (n=36). Decreased hOCT1 mRNA correlated with hypermethylation status of the SLC22A1 promoter. Treatment of CCA cells with decitabine (demethylating agent) or butyrate (histone deacetylase inhibitor) restored hOCT1 expression and increased sorafenib uptake. MicroRNAs able to induce hOCT1 mRNA decay were analyzed in paired samples of TCGA‐CHOL (n=9) and Copenhagen (n=57) cohorts. Consistent upregulation in tumor tissue was found for miR‐141 and miR‐330. High proportion of aberrant hOCT1 mRNA splicing in CCA was also seen. Lentiviral‐mediated transduction of eCCA (EGI‐1 and TFK‐1) and iCCA (HuCCT1) cells with hOCT1 enhanced sorafenib uptake and cytotoxic effects. In chemically‐induced CCA in rats reduced rOct1 expression was accompanied by impaired sorafenib uptake. In xenograft models of eCCA cells implanted in mouse liver, poor response to sorafenib was observed. However, tumor growth was markedly reduced by co‐treatment with sorafenib and adenoviral vectors encoding hOCT1 under the control of the BIRC5 promoter, a gene highly upregulated in CCA. Conclusions:The reason for impaired hOCT1‐mediated sorafenib uptake by CCA is multifactorial. Gene therapy capable of selectively inducing hOCT1 in tumor cells can be considered a potentially useful chemosensitization strategy to improve the response of CCA to sorafenib. This article is protected by copyright. All rights reserved.
Article
Mesenchymal stromal cells (MSCs) are a type of adult stem cell that has been exploited for the treatment of a variety of diseases, including cancer. In particular, MSCs have been studied extensively for their ability to treat glioblastoma (GBM), the most common and deadly form of brain cancer in adults. MSCs are attractive therapeutics because they can be obtained relatively easily from patients, are capable of being expanded numerically in vitro, can be easily engineered and are inherently capable of homing to tumors, making them ideal vehicles for delivering biological antitumoral agents. Oncolytic viruses are promising biological therapeutic agents that have been used in the treatment of GBMs, and MSCs are currently being explored as a means of delivering these viruses. Here we review the role of MSCs in the treatment of GBMs, focusing on the intersection of MSCs and oncolytic viruses.
Article
Full-text available
Glioblastoma multiforme (GBM) tumors display extensive histomorphological heterogeneity, with great variability in the extent of invasiveness, angiogenesis, and necrosis. The identification of genes associated with these phenotypes should further the molecular characterization, permitting better definition of glioma subsets that may ultimately lead to better treatment strategies. Therefore, we performed a differential mRNA display analysis comparing six GBM-derived primary cell cultures from patients having tumors with varied histomorphological features. We identified stromal cell-derived factor 1 (SDF1) as a gene with varied expression. SDF1 (cytokine) and CXC chemokine receptor 4 (CXCR4) interactions are implicated in modulating cell migration. They are also implicated in modulating the immune response in AIDS patients by macrophage-mediated T-cell apoptosis. GBM patients also fail to mount an immune response, although their tumors are seemingly exposed to immune cells in regions of angiogenesis, where the blood-brain barrier is absent, or in areas of necrosis. To determine whether the expression and localization of SDF1 and CXCR4 are consistent with such a role in these brain tumors, immunohistochemical analyses of these proteins were performed on normal brain and astrocytomas (grades II-IV). In normal brain tissue, low levels of SDF1 (0.5+) were observed in astrocytic processes, in neurons, and in the occasional phagocytic cells around vessels. CXCR4 expression was negative in brain tissue but was observed in phagocytic cells within the vessel lumen. In tumors, SDF1 and CXCR4 expression was colocalized when both were expressed, and SDF1 and CXCR4 expression increased with increasing tumor grade (from 0.5+ to 6+). Additionally, CXCR4 was expressed in neovessel endothelial cells. The proteins were expressed in regions of angiogenesis and degenerative, necrotic, and microcystic changes. Those tumors displaying greater amounts of these features had greater staining intensity of the proteins. The expression of SDF1 and CXCR4 did not colocalize with the proliferation marker MIB-1. Thus, our data suggest that SDF1 and CXCR4 expressions: (a) increase with increasing grade; (b) colocalize to regions within these tumors where their interaction may contribute to angiogenesis and/or modulation of the immune response; and (c) may serve to characterize subsets of GBMs.
Article
Full-text available
Chemokines constitute a superfamily of proteins that function as chemoattractants and activators of leukocytes. Astrocytes, the major glial cell type in the CNS, are a source of chemokines within the diseased brain. Specifically, we have shown that primary human astrocytes and human astroglioma cell lines produce the CXC chemokines IFN-gamma-inducible protein-10 and IL-8 and the CC chemokines monocyte chemoattractant protein-1 and RANTES in response to stimuli such as TNF-alpha, IL-1beta, and IFN-gamma. In this study, we investigated chemokine receptor expression and function on human astroglioma cells. Enhancement of CXC chemokine receptor 4 (CXCR4) mRNA expression was observed upon treatment with the cytokines TNF-alpha and IL-1beta. The peak of CXCR4 expression in response to TNF-alpha and IL-1beta was 8 and 4 h, respectively. CXCR4 protein expression was also enhanced upon treatment with TNF-alpha and IL-1beta (2- to 3-fold). To study the functional relevance of CXCR4 expression, stable astroglioma transfectants expressing high levels of CXCR4 were generated. Stimulation of cells with the ligand for CXCR4, stromal cell-derived factor-1alpha (SDF-1alpha), resulted in an elevation in intracellular Ca(2+) concentration and activation of the mitogen-activated protein kinase cascade, specifically, extracellular signal-regulated kinase 2 (ERK2) mitogen-activated protein kinase. Of most interest, SDF-1alpha treatment induced expression of the chemokines monocyte chemoattractant protein-1, IL-8, and IFN-gamma-inducible protein-10. SDF-1alpha-induced chemokine expression was abrogated upon inclusion of U0126, a pharmacological inhibitor of ERK1/2, indicating that the ERK signaling cascade is involved in this response. Collectively, these data suggest that CXCR4-mediated signaling pathways in astroglioma cells may be another mechanism for these cells to express chemokines involved in angiogenesis and inflammation.
Article
Survivin, one of the apoptosis inhibitor proteins, has been detected in most cancers in humans. In addition, two splice variants (survivin-2B and survivin-deltaEx3) have been identified. The authors investigated the transcription levels of survivin messenger (m)RNA and its splice variants in nine tumor cell lines, including gliomas, and in 25 brain tumor samples, by performing quantitative reverse transcription-polymerase chain reaction. The correlation between transcript expression levels and pathological findings were also analyzed. Transcription levels were measured using primer pairs specific for survivin and either of its splice variants and were normalized to the glyceraldehyde 6-phosphate dehydrogenase. Among the tumor cell lines tested, glioblastoma cell lines showed the highest levels of survivin expression. Among brain tumor samples studied, survivin was preferentially expressed in malignant brain tumors and gliomas. The relative expression level of survivin-deltaEx3/survivin was significantly higher in malignant than in benign brain tumor samples. Expression patterns were dominant for survivin-deltaEx3 in malignant brain tumors and dominant for survivin-2B in benign ones. A significant linear correlation between survivin mRNA expression and MIB-1 labeling index was demonstrated in all brain tumor samples. The authors' results indicate that quantifying the levels of survivin and its splice variants is useful for the prediction of the cell biological malignancy of gliomas, independent of their pathological features.
Article
Cytosine arabinoside (ara-C) is a cytidine analog that incorporates into replicating DNA and induces lethal DNA strand breaks. Although ara-C is a potent antitumor agent for hematologic malignancies, it has only minimal activity against most solid tumors. The rate-limiting step in intracellular ara-C activation is phosphorylation of the prodrug by deoxycytidine kinase (dCK). The present results demonstrate that both retroviral and adenoviral vector-mediated transduction of the dCK cDNA results in marked sensitization of glioma cells lines to the cytotoxic effects of ara-C in vitro. We also demonstrate that ara-C treatment of established intradermal and intracerebral gliomas transduced with dCK results in significant antitumor effects in vivo. These data suggest that viral vector transduction of the dCK gene followed by treatment with ara-C represents a new chemosensitization strategy for cancer gene therapy.
Article
Midkine (MK), a member of a new family of neurotrophic and angiogenic growth factors whose expression is developmentally regulated, is produced in fetal astrocytes. Malignant astrocytomas, one of the most neovascularized tumors, are derived from astrocytes. There has been no investigation of the expression of MK in human astrocytic tumors. To determine if increased levels of MK expression correlate with the progression of human astrocytomas, we examined surgical specimens of astrocytic tumors of various grades using Northern and Western blotting. MK mRNA and protein expression levels were higher in high-grade astrocytomas (anaplastic astrocytomas and glioblastomas) than in low-grade astrocytomas. As shown by in situ hybridization, MK mRNA expression was intense in a majority of glioblastoma cells but was weak in a small number of low-grade astrocytoma cells. These findings suggest that MK expression correlates with the malignant progression of astrocytomas. The aberrant MK expression in high-grade astrocytomas may underlie their rapid growth and well-vascularized features.
Article
We have examined the expression of midkine (MK), a neurotrophic factor with heparin-binding activity, in human esophageal cancer cells. Seven esophageal cell lines tested expressed the transcript and 8 out of 14 human esophageal tumor specimens were positively stained with anti-MK antibody, while surrounding normal esophageal tissues in these specimens were not stained. The 5'-flanking, 2.3 kb genomic region of the MK gene was shown to drive the transcription of a reporter gene in the esophageal cell lines in a cis acting manner. Forced expression in esophageal cancer cells of herpes simplex virus-thymidine kinase gene mediated by the flanking region of the MK gene conferred sensitivity to a prodrug, ganciclovir. The 5'-upstream region of the MK gene thus possesses putative promoter activity which can be used for suicide gene-based gene therapy for esophageal cancer.
Article
It is important to develop a system to express therapeutic genes in tumor cells with sufficient selectivity for cancer gene therapy. Midkine (MK) is a newly identified heparin-binding growth factor that is transiently expressed in the early stages of retinoic acid-induced differentiation of embryonal carcinoma cells. It has been reported that many human malignant tumors express high levels of MK mRNA or protein. However, no MK expression is detected in human or mouse liver. These interesting features of MK led us to examine the MK promoter as a candidate for tumor-specific gene expression. We thus developed new recombinant adenoviral (Ad) vectors containing either luciferase reporter gene (AdMKLuc) or herpes simplex thymidine kinase gene (AdMKTK) under the control of the human MK promoter. AdMKLuc achieved relatively high activity in Wilms' tumor (G-401) and neuroblastoma (SK-N-SH) cell lines. In addition, AdMKTK induced marked cell death in response to ganciclovir (GCV) in these same lines. Conversely, very low activity of the MK promoter was observed in mouse liver in vivo compared with the cytomegalovirus promoter. Importantly, AdMKTK + GCV did not induce liver toxicity, whereas substantial toxicity was seen with AdCMVTK + GCV treatment. On the basis of these findings, we conclude that the MK promoter is a candidate tumor-specific promoter for Wilms' tumor or neuroblastoma.
Article
Replicating adenoviruses (Ads) are designed to replicate in and destroy cancer cells, generating viral progeny that spread within the tumor. To address the importance of the primary cellular receptor for Ads, the coxsackievirus and Ad receptor (CAR), in permitting intratumoral spread of a replicating Ad, we have used a pair of tumor cell lines differing only in the expression of a primary receptor for Ad5. This novel system thus allowed the first direct evaluation of the relationship between the efficacy of a replicating Ad and the primary receptor levels of the host cell without the confounding influence of other variable cellular factors. We demonstrate that the absence of the primary cellular receptor on the tumor cells restricts the oncolytic potency of a replicating Ad both in vitro and in vivo. Based on these findings, it is apparent that the potential therapeutic advantages afforded by viral replication would be negated by poor intratumoral spread of the viral progeny due to the failure to infect neighboring tumor cells. Because a number of studies have reported that primary cancer cells express only low levels of CAR, our results suggest that strategies to redirect Ads to achieve CAR-independent infection will be necessary to realize the full potential of replicating Ads in the clinical setting.
Article
Survivin is a novel antiapoptotic gene that has been recently cloned and characterized. Its expression has been found to be of prognostic significance in several tumor types. This is the first study on the prognostic significance of survivin expression in human gliomas. We used quantitative Western blot analysis with densitometry to determine survivin protein expression levels in 92 glioma cases for which frozen tissue was available for analysis. Survivin positivity and expression levels were correlated with histopathologic features of the tumors, apoptosis (as measured by cleaved, or activated, caspase 3 levels), and clinical outcome. Survivin expression has clear prognostic value in human gliomas. Patients with detectable survivin expression had significantly shorter overall survival times (P <.0001) compared with those without detectable expression when all glioma patients were considered. Although glioblastoma multiforme (GBM) patients had significantly higher rates of survivin positivity and higher levels of survivin expression (P <.0001) than their non-GBM counterparts, the prognostic value of survivin expression seemed to be independent of histology alone. Survivin-positive GBM patients had significantly shorter overall survival times compared with survivin-negative GBM patients (P <.0001). Likewise, survivin-positive non-GBM patients had shorter survival times compared with survivin-negative non-GBM patients (P =.029). Furthermore, increasing levels of survivin expression significantly correlated with reduced survival times when all glioma patients were considered, and markedly so for GBM patients (P <.0001). Increasing survivin levels significantly correlated with reduced expression of cleaved caspase 3, indicating its association with antiapoptotic activity. Survivin positivity and protein expression levels, as determined quantitatively, are of significant prognostic value in human gliomas and seem to be associated with reduced apoptotic capacity of these tumors.
Article
Studies in several tumour types have suggested that the inappropriate expression of the novel inhibitor of apoptosis protein survivin may play a key role in tumourigenesis. This study presents the first immunohistochemical examination of survivin expression in glioblastomas. The cohort consisted of 39 ethnic Chinese patients diagnosed with primary glioblastoma multiforme. Samples were archival paraffin-embedded blocks. Concomitant with examination for survivin expression, samples were also examined for over-expression of the p53 protein as well as for evidence of apoptotic cells via the terminal deoxynucleotide transferease (TdT) mediated nick end labelling (TUNEL) technique. Results showed that survivin was expressed in nearly 80% (31/39) of samples. Over-expression of moderate or high levels of survivin was correlated with the absence of apoptotic cells ( P=0.03). Expression of survivin and p53 was found to be significantly related ( P=0.037), and 70% (27/39) of tumours showed co-ordinate expression of p53 and survivin. Given that p53 over-expression in primary glioblastomas is predominantly detected in the absence of mutations of the gene, and that both survivin and p53 are regulated at the level of the protein by the same ubiquitin-proteosome degradation pathway, these results suggest that primary glioblastomas may occur as a result of a failure of appropriate protein degradation regulation.