ArticlePDF Available

Local Immune Stimulation by Intravesical Instillation of Baculovirus to Enable Bladder Cancer Therapy

Authors:

Abstract and Figures

Intravesical instillation of Bacillus Calmette-Guérin is currently used as adjuvant therapy for superficial, non-muscle invasive bladder cancer (NMIBC). However, nearly 40% of patients with NMIBC will fail Bacillus Calmette-Guérin therapy. In an attempt to investigate the feasibility of using insect baculovirus-based vectors for bladder cancer therapy, we observed that intravesical instillation of baculoviruses without transgene up-regulated a set of Th1-type of cytokines and increased the survival rate of mice bearing established orthotopic bladder tumors. When baculoviral vectors were used to co-deliver the mouse CD40 ligand and IL-15 genes through intravesical instillation, the immunogene therapy triggered significantly increased bladder infiltrations of inflammatory monocytes, CD4+, CD8+ and γδ T lymphocytes. All treated animals survived beyond 12 months whereas control animals died around 2 months after tumor inoculation. We conclude that direct intravesical instillation of baculoviral gene transfer vectors holds the potential to be a novel therapeutic modality for NMIBC.
In vivo baculoviral transduction after intravesical instillation of baculoviral vectors in mice. (A) Bioluminescence images of luciferase reporter gene expression in immunodeficient BALB/c nude mice. Images of representative animals transduced with 3 different baculoviral vectors (10 7 viral particles per mouse) are shown. Heat map represents the transgene expression area and color represents the intensity. The schematic structures of baculoviral vector expression cassettes are shown on the left. Abbreviations: CMV: the human cytomegalovirus immediate-early gene promoter and enhancer; WPRE: the woodchuck hepatitis virus posttranscriptional regulatory element; RU5: R segment and part of the U5 sequence of the long terminal repeat from the human T-cell leukemia virus type 1. (B) Time course analysis of luciferase reporter gene expression in BALB/c nude mice. In vivo gene expression levels are quantified by measuring bioluminescence signals. The data represent the mean + s.d., n = 5 per group. (C) Time course analysis of luciferase reporter gene expression in C57BL/6 mice. Mice were transduced with the baculoviral vector BV-RU5-Luc-WPRE at a dose of 10 8 or 10 7 viral particles per mouse. The data represent the mean + s.d., n = 5 per group. (D) MB49 tumor growth in the bladder of C57BL/6 mice. MB49 mouse bladder tumor cells (10 5 per mouse) were implanted into poly-Llysine pre-treated bladder and allowed to establish tumors for one week. A bright field image of an H&E stained section showing mouse bladder architecture and a tumor inside the bladder (arrow). (E) Immunostaining to demonstrate baculoviral transduction in bladder tumors. Tumor-bearing bladders were collected 24 hours after transduction with BV-RU5-Luc-WPRE for immunostaining with antibodies against the luciferase protein. A fluorescence image is shown. (F) Flow cytometric analysis to demonstrate transduction of MB49 tumor cells with BV-RU5-eGFP-WPRE. Phase contrast and fluorescence images of transduced cells are shown in the bottom panels.
… 
Baculovirus as an immunostimulatory agent for bladder cancer therapy. (A) Baculovirusmediated cytokine/chemokine expression in mice treated with PBS or BacPAK6, a baculoviral vector without mammalian gene expression cassette. Top: Bladders were harvested 48 hours after the respective instillation, homogenized and extracts were used to probe the cytokine/chemokine arrays. After densitometric analysis, the top 5 up-regulated cytokines/chemokines are labeled on the blots. The pictures shown are representative. Bottom: Densitometric analysis of the cytokine/chemokine arrays. The RayBio ® Analysis Tool was used to relate average signal intensity to cytokine/chemokine expression level. The average signal intensity of 3 mice per group and a > 3-fold change as a baseline was used for analysis. (B) Therapeutic effects of BacPAK6 as demonstrated by bladder weight measurement. Mice were inoculated with MB49 tumor cells intravesically and 7 days later treated with PBS or BacPAK6. Survived mice were sacrificed on day 21 post-tumor inoculation and bladders were collected for weight measurement. The data represent mean + s.d., n = 6 or 10 per group. (C) BacPAK6 transduction prolongs survival of bladder tumor-bearing mice. Survival curves till day 95 are shown. n = 10 in the PBS group and n = 20 in the BacPAK6 group. The statistical analysis was performed using the log rank test. (D) Induction of protective immunity in mice survived in BacPAK6 group in Fig. 2C. These mice were rechallenged by intravesical tumor inoculation with MB49 cells (n = 10). Age-matched naïve mice were used as controls and inoculated with the same number of MB49 cells (n = 10). Survival curves till day 180 post-tumor inoculation are shown.
… 
Content may be subject to copyright.
1
Scientific RepoRts | 6:27455 | DOI: 10.1038/srep27455
www.nature.com/scientificreports
Local Immune Stimulation
by Intravesical Instillation of
Baculovirus to Enable Bladder
Cancer Therapy
Wei Xia Ang1,2, Ying Zhao2, Timothy Kwang3, Chunxiao Wu2,3, Can Chen3, Han Chong Toh 4,
Ratha Mahendran5, Kesavan Esuvaranathan5 & Shu Wang1,2
Intravesical instillation of Bacillus Calmette-Guérin is currently used as adjuvant therapy for supercial,
non-muscle invasive bladder cancer (NMIBC). However, nearly 40% of patients with NMIBC will
fail Bacillus Calmette-Guérin therapy. In an attempt to investigate the feasibility of using insect
baculovirus-based vectors for bladder cancer therapy, we observed that intravesical instillation of
baculoviruses without transgene up-regulated a set of Th1-type of cytokines and increased the survival
rate of mice bearing established orthotopic bladder tumors. When baculoviral vectors were used to
co-deliver the mouse CD40 ligand and IL-15 genes through intravesical instillation, the immunogene
therapy triggered signicantly increased bladder inltrations of inammatory monocytes, CD4+, CD8+
and γδ T lymphocytes. All treated animals survived beyond 12 months whereas control animals died
around 2 months after tumor inoculation. We conclude that direct intravesical instillation of baculoviral
gene transfer vectors holds the potential to be a novel therapeutic modality for NMIBC.
Adjuvant therapy is oen given following transurethral resection of supercial, non-muscle invasive bladder
cancer (NMIBC) to control tumor recurrence and progression1,2. Intravesical immunotherapy with Bacillus
Calmette-Guérin (BCG) is currently recommended for patients with intermediate- or high-risk of bladder can-
cer recurrence. BCG is produced from attenuated live bovine tuberculosis bacterium and can activate innate
immune responses mediated by cytokines such as interleukin-2 (IL-2), IL-12, IL-18 and interferon-gamma
(INF-γ ). Unfortunately, up to 40% of patients with NMIBC will fail BCG immunotherapy within the rst year
due to BCG refractory, resistance, relapsing, or intolerance1–3. Currently, there is still no gold standard for salvage
therapy aer BCG failure, highlighting the urgent need to develop new adjuvant therapies to improve treatment
outcomes for patients with NMIBC.
e bladder is a hollow organ, allowing nonsurgical intravesical drug administration through a urethral cathe-
ter and evaluation of treatment ecacy by the means of endoscopy. Taking advantage of the fact that intravesically
delivered therapeutics act locally with limited systemic exposure and that supercial bladder cancer is easily
accessible, viruses, either replication-competent viruses or replication-decient recombinant viral vectors, have
been tested for bladder cancer therapy in tumor models3,4. ese viral therapeutics include vaccinia virus5–8,
adenovirus7,9–15, canarypox virus7, reovirus16, retrovirus17–20, lentivirus21, and vesicular stomatitis virus22. At least
6 clinical trials have been reported on the use of vaccinia virus and adenoviral vectors to treat bladder can-
cer4,6,11,12,14,15. However, the hurdles to translating the current success of these viral therapeutics to a broad clinical
application appear high, which include the lack of gene delivery vectors with active immunostimulatory function
and inecient expression of a therapeutic gene in bladder tumor cells and in the organ.
Recombinant vectors derived from the insect baculovirus Autographa californica multiple nucleopolyhedro-
virus (AcMNPV) hold the ability to enter mammalian cells without replicating or causing toxicity to the trans-
duced cell. Since baculoviral vectors with a mammalian expression cassette could be highly eective in mediating
1Department of Biological Sciences, National University of Singapore, 117543 Singapore. 2Institute of Bioengineering
and Nanotechnology, 138669 Singapore. 3Tessa Therapeutics, 239351 Singapore. 4Division of Medical Oncology,
National Cancer Centre, 169610 Singapore. 5Department of Surgery, Yong Loo Lin School of Medicine, National
University of Singapore, 119074 Singapore. Correspondence and requests for materials should be addressed to S.W.
(email: dbsws@nus.edu.sg)
Received: 24 August 2015
Accepted: 19 May 2016
Published: 08 June 2016
OPEN
www.nature.com/scientificreports/
2
Scientific RepoRts | 6:27455 | DOI: 10.1038/srep27455
transient expression and usually do not integrate into the genome of the transduced cells23, they are ideally suited
for applications requiring short-term, high level transgene expression and pose much less risk of insertional
mutagenesis. From an application point of view, baculoviral vectors are easy to manipulate, able to carry large
(at least 38 kbp) and multiple DNA inserts, and can be readily produced in serum-free cell culture medium and
puried at high titers in Biosafety Level 1 laboratories. us, recombinant baculoviruses have been suggested as a
novel type of vectors for cancer gene therapy23–27.
In preclinical animal studies, baculoviral vectors have been shown to possess a strong adjuvant activity in
inducing humoral and cellular immune responses against co-administered antigens, possibly through promoting
maturation of dendritic cells (DCs) and producing pro-inammatory cytokines, chemokines and type I IFNs28,29
and activating natural killer (NK) cell-dependent antitumor immunity30,31. e “adjuvant” eect of baculoviral
vectors is attributed to the high frequency of CpG motifs in the viral genome, which is similar to that in bacterial
DNA and signicantly higher than that of mammalian and adenovirus DNA32,33. However, a critical obstacle
toward in vivo baculoviral transduction is the inactivation of systemically delivered baculoviral vectors as a con-
sequence of virus recognition by serum complement proteins, a major component of the innate immune system34.
Hence, in immunocompetent animals systemically delivered baculoviral vectors failed to transduce target cells.
Bladder cancer therapy can be performed with intravesical catheterization through the urethra and easily
avoids many barriers to systemic virus administration, thereby presenting a unique opportunity to explore in vivo
applications of baculoviral vectors. Herein we report that intravesical treatment with baculoviruses alone is able
to prolong survival of mice with established orthotopic bladder cancer. We further demonstrate that baculoviral
vectors are eective in delivering therapeutic genes into normal and malignant urothelial cells and can be used for
immunogene therapy for bladder cancer in a mouse model.
Materials and Methods
Baculoviral vectors. Recombinant baculoviral vectors, CMV-Luc, CMV-Luc-WPRE and CMV-RU5-
Luc-WPRE, were constructed using BAC-to-BAC baculovirus expression system (Invitrogen, Carlsbad, CA).
BV-CD40 ligand (CD40L) virus and BV-IL15 virus which contain the mouse CD40L gene and mouse IL15 gene
respectively were produced by homologous recombination aer co-transfection of Sf9 insect cells with pBacPAK6
transfer vector containing the expression cassette and linearized AcMNPV viral DNA (Clontech, Mountain View,
CA). BacPAK6, the parental virus with the lacZ gene driven by viral polyhedrin promoter, was obtained from
Clontech.
Mouse tumor model. Adult female C57BL/6 mice were used to generate orthotopic bladder tumors on the
luminal surface of the bladder by intravesical instillation of syngeneic MB49 cells with a 24-gauge catheter. Aer
a 30-minute PLL pre-treatment, 100 μ l of MB49 cells in PBS was instilled and retained in the bladder for 1 hour
by leaving the catheter in situ and clamped. A dose of 1 × 105 cancer cells per animal was used in all experiments.
ereaer, the catheter was removed and the bladder was evacuated by spontaneous voiding. MB49 cells used
for tumor inoculation were pre-labeled with a lipophilic, near-infrared uorescent dye DiR (20 ng/ml) (Caliper
Life Sciences) overnight. One week aer intravesical instillation of MB49 cells, mice were imaged with IVIS100
in vivo imaging system using 710 nm excitation and 760 nm emission lter set to examine tumor implantation.
Animals successfully implanted with DiR-labeled MB49 cells and with similar tumor burden were kept for exper-
iments designed to evaluate therapeutic ecacy. In the current study, the uorescent signal of the DiR-labelled
MB49 cells persisted without substantial change for at least 7 days. e signal began to decay thereaer, although
remaining weakly detectable even on day 14. Hematuria, an indicator for the tumour formation in the bladder,
was also monitored in some experiments to further conrm the tumor implantation and growth.
To test therapeutic eects of baculoviral vectors, mice bearing bladder tumors were randomized to control
or treatment groups, re-anaesthetized, and re-catheterized. Baculoviruses (1 × 108 pfu in 100 μ l), PBS (100 μ l), or
BCG (1.35 mg = 2.3 × 107 colony forming units in 100 μ l, Aventis Pasteur, Canada) were intravesically instilled
and retained in the bladders for 2 hours. In some experiments, mice were given up to three weekly intravesical
instillations. Animals were observed for up to 12 months for signs and symptoms of bladder cancer (hematuria
and weight loss) and viability status.
e animal study protocol was reviewed and approved by Institutional Animal Care and Use Committee
(IACUC), the Biological Resource Centre, the Agency for Science, Technology and Research (A*STAR), Singapore
(Permit Number: BRC IACUC 110612). e methods were carried out according to the guidelines for the Care
and Use of Animals for Scientic Purposes issued by the National Advisory Committee for Laboratory Animal
Research, Singapore.
Cytokine/chemokine expression. Mice were euthanized 48 hours aer intravesical instillation of PBS or
BacPAK6. Bladders were harvested and weighed. Bladder homogenates were obtained by adding 1 ml of a tis-
sue lysis buer (Fermentas, Maryland, USA) with a protease inhibitor cocktail (Merck, Darmstadt, Germany)
to 50 mg of tissue sample and homogenizing by sonication. Bladder homogenates were then centrifuged at
16,000 × g for 30 min at 4 °C and the supernatants collected. Protein concentrations of the supernatants were
determined by the Biorad protein assay method (Biorad, California, USA). An aliquot of the supernatant con-
taining 80 μ g of total protein concentration was loaded onto the Mouse Cytokine Array C3 (Raybiotech, Norcross,
GA) to measure expression levels of cytokines and chemokines. RayBio® Analysis Tool (Raybiotech) was used to
correlate the average signal intensities to relative expression levels of cytokines.
Analysis of inltrated immune cells. For ow cytometry analysis of inltrated immune cells, the col-
lected bladder cells were pre-incubated with Fc block (CD16/CD32, Clone 2.4G2, BD), washed, and incubated
with appropriate uorescent-conjugated antibodies. For immunohistochemistry analysis, the tissue sections were
www.nature.com/scientificreports/
3
Scientific RepoRts | 6:27455 | DOI: 10.1038/srep27455
incubated with 0.3% H2O2 for 10 min to block the endogenous peroxidase activity. e tissue sections were then
incubated in 5% BSA for 1 h to block non-specic binding sites before incubation with appropriate primary anti-
bodies and HRP-conjugated secondary antibodies. Staining was developed by 3,3 -diaminodbenzidine substrate
and the nuclei were counterstained by hematoxylin.
Statistical analysis. For survival rates, statistical analysis was performed using the log-rank test. For others,
statistical signicance was assessed by Students t-test; p < 0.05 was considered statistically signicant.
Supplemental methods. Details of methods for baculoviral vector construction, cell culturing, in vitro &
in vivo baculoviral transduction, ow cytometry analysis, histological analysis, and immunostaining are provided
in the Supplementary Materials and Methods.
Results
Baculoviral vectors eectively transduce the mouse bladder and bladder tumors after intra-
vesical instillation. We rst assessed whether baculovirus could transduce the bladder in immunodecient
BALB/c nude mice. For that, we constructed three dierent recombinant baculoviral vectors containing a rey
luciferase gene and compared their in vivo transduction eciency aer intravesical instillation into the bladder
at a dose of 107 viral particles per mouse. e bladders were pre-treated with PLL before viral vector instillation
as we found that this pre-treatment signicantly enhanced the uptake of baculoviral vectors (Fig. S1A). All three
vectors were able to transduce the mouse bladder as evidenced by using the IVIS living animal imaging system
(Fig.1A). One of the baculoviral vectors, BV-RU5-Luc-WPRE, that contains two viral transcriptional regulatory
elements WPRE and RU5, provided the highest transgene expression level in the bladder. While decreasing over
time, the expression levels provided by the three vectors remained signicantly higher than a background level
for at least 35 days (Fig.1B).
We next tested baculoviral transduction in the bladder of immunocompetent C57BL/6 mice aer intravesical
instillation of BV-RU5-Luc-WPRE. e in vivo transduction eciency was dosage-dependent with the luciferase
expression level at day 1 in C57BL/6 mice treated with 108 viral particles per mouse being approximately 5-fold
greater than that provided by treatment with 107 viral particles per mouse (Fig.1C). Although the initial expres-
sion level provided by 108 viral particles of BV-RU5-Luc-WPRE in C57BL/6 mice was similar to that observed in
immunodecient nude mice treated with 107 viral particles, the level dropped quickly and the detectable trans-
gene expression lasted for approximately 2 weeks only. e dierence in transgene expression between immuno-
decient nude mice and immunocompetent mice indicates a strong immune response to baculoviral transduction
that might eliminate the viral vectors in the transduced organ. Aer intravesical instillation with 108 viral par-
ticles in C57BL/6 mice we observed no behavioral abnormalities and hepato- and nephro-toxicities (Fig. S2).
Hence, this dose was used for all following animal experiments.
We then tested whether BV-RU5-Luc-WPRE, following intravesical instillation, could transduce bladder tum-
ors in an orthotopic tumor model generated by implantation of syngeneic MB49 bladder cancer cells into the
bladders of C57BL/6 mice (Fig.1D). Immunohistological staining with an antibody against the luciferase protein
conrmed that the viral vectors transduced the tumors eciently, penetrating deep into the tumor mass and
distributing extensively throughout the whole tumor bed (Fig.1E). However, in tumor-free areas, as well as in the
normal bladder, baculovirus-mediated transgene expression was conned to the supercial bladder epithelium,
suggesting a restricted regional transgene delivery by intravesically instilled baculoviral vectors in the normal
bladder (Fig.1E; Fig. S1B). Baculoviral transduction of MB49 mouse bladder cancer cells was further conrmed
using BV-RU5-eGFP-WPRE (Fig.1F). is viral vector could also transduce several of human bladder cancer cell
lines, including T24, HBC1, HTB2 and HTB5 (Fig. S3).
Baculoviral transduction alone is capable of retarding bladder tumor growth. As baculoviral
transduction in the bladder possibly stimulates local immune responses, we investigated expression of cytokines
and chemokines in the organ upon intravesical instillation of baculovirus in C57BL/6 mice. We used BacPAK6,
a baculoviral vector without a mammalian gene expression cassette, for this purpose to avoid possible inter-
ference by transgene expression. Using an antibody array method, we detected the up-regulation (> 3-fold
increase in expression) of 61% of the cytokines and chemokines in a murine array (38 out of 62) in the bladder
that received intravesical instillation of BacPAK6 two days before as compared to the expression levels in the
bladder that received PBS instillation (Fig.2A). e top 5 up-regulated proteins were granulocyte-macrophage
colony-stimulating factor (GM-CSF, 154-fold increase), interleukin-6 (IL-6, 101-fold increase), interleukin-1 beta
(IL1-beta, 48-fold increase), granulocyte colony-stimulating factor (G-CSF, 42-fold increase), and leptin receptor
(Leptin R, 33-fold increase). A comparison between the normal mouse bladder receiving no treatment and the
bladder receiving PBS instillation showed no signicant dierence in the expression levels of the cytokines and
chemokines (data not shown).
e above antibody array results led us to hypothesize that the up-regulation of cytokines and chemokines
upon baculoviral transduction would aect bladder tumor growth. To test this, C57BL/6 mice were inoculated
with MB49 tumor cells and one week later randomly distributed into two groups (n = 10 per group): one group
received single intravesical instillation of BacPAK6 and another group received PBS as an instillation control.
Bladders were harvested on day 21 post-tumor inoculation for weight measurement. Indeed, reduced bladder
weight as compared with the PBS group, an indication of retarded tumor growth, was observed in the BacPAK6
group (Fig.2B). We further observed signicantly prolonged survival of the bladder tumor-bearing mice aer
BacPAK6 instillation in the bladder. While all animals in the PBS control group died within 70 days, 50% of the
animals in the treatment group were still alive on day 95 post-tumor inoculation (Fig.2C, p < 0.01 in log-rank
test). Furthermore, no therapeutic eects were observed aer baculovirus instillation in immunodecient nude
www.nature.com/scientificreports/
4
Scientific RepoRts | 6:27455 | DOI: 10.1038/srep27455
mice (data not shown). ese ndings indicate that BacPAK6-triggered immune responses are responsible for
the observed antitumor eects.
To further test the impact of baculovirus-triggered immune responses on tumor growth, BacPAK6-treated
mice that survived from the above animal experiment in Fig.2C were re-challenged with MB49 tumor cells intra-
vesically. All mice survived for at least 180 days aer the 2nd tumor challenge (Fig.2D). As a control, age-matched
naïve mice were challenged with the same number of MB49 cells and all animals died by day 60. us, baculovirus
instillation in the bladder is able not just to trigger local immune responses, but also promote systemic, adaptive
anti-tumor immunity against MB49 tumor cells.
Baculoviral transduction-mediated immunogene therapy for bladder cancer in mice. To eval-
uate the potential of using baculovirus as a gene therapy vector, two recombinant baculoviral vectors (BV-CD40L
and BV-IL15) armed with either the mouse CD40L or the mouse IL-15 genes were constructed by replacing the
luciferase gene in BV-RU5-Luc-WPRE. Baculovirus-mediated CD40L and IL-15 expressions were conrmed
by in vitro transduction in MB49 cells, followed by Western blot analysis 48 hours post transduction (Fig. S4).
rough subcutaneous injection of original MB49 cells, in vivo selection/enrichment, and primary tumor cell
culturing, we collected faster growing MB49 tumor cells to evaluate the therapeutic eects of BV-CD40L and
BV-IL15 (see Supplementary Information). In the orthotopic model generated with aggressive MB49 cells,
Figure 1. In vivo baculoviral transduction aer intravesical instillation of baculoviral vectors in mice.
(A) Bioluminescence images of luciferase reporter gene expression in immunodecient BALB/c nude mice.
Images of representative animals transduced with 3 dierent baculoviral vectors (107 viral particles per mouse)
are shown. Heat map represents the transgene expression area and color represents the intensity. e schematic
structures of baculoviral vector expression cassettes are shown on the le. Abbreviations: CMV: the human
cytomegalovirus immediate-early gene promoter and enhancer; WPRE: the woodchuck hepatitis virus post-
transcriptional regulatory element; RU5: R segment and part of the U5 sequence of the long terminal repeat
from the human T-cell leukemia virus type 1. (B) Time course analysis of luciferase reporter gene expression
in BALB/c nude mice. In vivo gene expression levels are quantied by measuring bioluminescence signals. e
data represent the mean + s.d., n = 5 per group. (C) Time course analysis of luciferase reporter gene expression
in C57BL/6 mice. Mice were transduced with the baculoviral vector BV-RU5-Luc-WPRE at a dose of 108 or
107 viral particles per mouse. e data represent the mean + s.d., n = 5 per group. (D) MB49 tumor growth in
the bladder of C57BL/6 mice. MB49 mouse bladder tumor cells (105 per mouse) were implanted into poly-L-
lysine pre-treated bladder and allowed to establish tumors for one week. A bright eld image of an H&E stained
section showing mouse bladder architecture and a tumor inside the bladder (arrow). (E) Immunostaining to
demonstrate baculoviral transduction in bladder tumors. Tumor-bearing bladders were collected 24 hours
aer transduction with BV-RU5-Luc-WPRE for immunostaining with antibodies against the luciferase protein.
A uorescence image is shown. (F) Flow cytometric analysis to demonstrate transduction of MB49 tumor
cells with BV-RU5-eGFP-WPRE. Phase contrast and uorescence images of transduced cells are shown in the
bottom panels.
www.nature.com/scientificreports/
5
Scientific RepoRts | 6:27455 | DOI: 10.1038/srep27455
hematuria, an early sign of tumor growth, was observed in most of the mice as early as one week aer tumor cell
inoculation. We treated these tumor-bearing mice with one, two or three repeated instillations of viral vectors
or control reagents on days 7, 14 and 21 post-tumor implantation respectively. Since BCG treatment is the gold
standard for immunotherapy of bladder cancer, it was included for comparison with baculoviral vectors.
In the rst set of the gene therapy experiments, animals were sacriced on day 35 post-tumor inoculation
and bladders were harvested for weight measurement (Fig.3A). erapeutic eects were obvious aer one instil-
lation of viral vectors or BCG and became increasingly pronounced with increase in the number of instilla-
tions. Aer giving two or three instillations of both BV-CD40L and BV-IL15, the weight of the bladders from the
tumor-inoculated mice was almost same as that in the normal mice group, indicating that multiple instillations
of therapeutic gene-expressing baculoviral vectors may promote complete tumor regression. is was supported
by a long-term survival study in which all MB49 cell-inoculated mice that were administered three instillations
of both BV-CD40L and BV-IL15 survived for at least 12 months. Figure3B depicts the survival rates of dierent
groups at day 125 post-tumor inoculation, with 100% survival in the group treated with both BV-CD40L and
BV-IL15, followed by 90% in the mice receiving BV-CD40L or BV-IL15, 75% in the BacPAK6 group, and 60%
in BCG-treated mice. e results of statistical analysis of survival rates are shown in Table S1. Complete tumor
regression aer three instillations of both BV-CD40L and BV-IL15 was conrmed by histological examination
(Fig.3C). In the mice sacriced on day 35 post-tumor inoculation bladder tumors were almost undetectable and
distinct normal transitional epithelium structure of the bladder mucosa was seen. Taken together, our results
Figure 2. Baculovirus as an immunostimulatory agent for bladder cancer therapy. (A) Baculovirus-
mediated cytokine/chemokine expression in mice treated with PBS or BacPAK6, a baculoviral vector without
mammalian gene expression cassette. Top: Bladders were harvested 48 hours aer the respective instillation,
homogenized and extracts were used to probe the cytokine/chemokine arrays. Aer densitometric analysis,
the top 5 up-regulated cytokines/chemokines are labeled on the blots. e pictures shown are representative.
Bottom: Densitometric analysis of the cytokine/chemokine arrays. e RayBio® Analysis Tool was used to relate
average signal intensity to cytokine/chemokine expression level. e average signal intensity of 3 mice per group
and a > 3-fold change as a baseline was used for analysis. (B) erapeutic eects of BacPAK6 as demonstrated
by bladder weight measurement. Mice were inoculated with MB49 tumor cells intravesically and 7 days later
treated with PBS or BacPAK6. Survived mice were sacriced on day 21 post-tumor inoculation and bladders
were collected for weight measurement. e data represent mean + s.d., n = 6 or 10 per group. (C) BacPAK6
transduction prolongs survival of bladder tumor-bearing mice. Survival curves till day 95 are shown. n = 10
in the PBS group and n = 20 in the BacPAK6 group. e statistical analysis was performed using the log rank
test. (D) Induction of protective immunity in mice survived in BacPAK6 group in Fig. 2C. ese mice were re-
challenged by intravesical tumor inoculation with MB49 cells (n = 10). Age-matched naïve mice were used as
controls and inoculated with the same number of MB49 cells (n = 10). Survival curves till day 180 post-tumor
inoculation are shown.
www.nature.com/scientificreports/
6
Scientific RepoRts | 6:27455 | DOI: 10.1038/srep27455
conrmed that three instillations of both BV-CD40L and BV-IL15 could eectively suppress bladder cancer
growth in vivo.
Baculoviral transduction in the bladder is associated with robust infiltration of immune
cells. To elucidate the cellular mechanisms underlying the immune response activated by baculoviral trans-
duction, we used ow cytometric analysis to characterize immune cell inltrates in the bladder aer repeated
instillations. Sixteen-hours aer the third instillation as illustrated in Fig.3A, C57BL/6 mouse bladders were
harvested, digested and stained for analysis. Neutrophils were dened as live CD45.2+Ly-6G+ cells and inam-
matory monocytes as live CD45.2+Ly-6GLy-6ChighCD11b+ cells. Results were expressed as % of a lymphocyte
subset among live leukocytes (Fig.4). We observed signicantly elevated inltration of inammatory monocytes
in the bladder aer repeated intravesical instillations of BCG, BacPAK6, or baculoviral gene therapy vectors
as compared to PBS instillation. e percentage of inammatory monocytes among live leukocytes increased
from 0.2% in the PBS group to approximately 20 to 30% aer treatment with BCG, BacPAK6, and baculoviral
gene therapy vectors, with the highest inltration rate (34.3%) aer co-instillations of BV-CD40L and BV-IL15.
Neutrophil inltration increased modestly, from 10% in the PBS group to 12 to 18% aer treatment with BCG,
BacPAK6, or baculoviral gene therapy vectors. us, inammatory monocytes could be one type of eector cells
for eliminating bladder tumors by baculoviral transduction in our mouse model.
T lymphocytes were gated as live CD45.2+CD3ε
+NK1.1 cells, and then as γ δ T-cell receptor (TCR) positive
or negative cells. From the γ δ TCR negative population CD4 and CD8α expression were assessed. As illustrated
in Fig.5, both BCG and BacPAK6 increased the accumulation of T lymphocytes in the bladder, with a slightly
higher number of CD8+ T lymphocytes aer BacPAK6 instillation. Using BV-CD40L and BV-IL15 for repeated
instillations, total T cell populations increased by 2-fold and 1.5-fold respectively as compared with BacPAK6.
Co-instillations of BV-CD40L and BV-IL15 increased the accumulation of CD4+ T cells and CD8+ T cells by
approximately 4-fold. Furthermore, co-instillations with BV-CD40L and BV-IL15 generated a profound stimu-
latory eect on the accumulation of γ δ T cells, providing an approximately 10-fold increase in γ δ T cell inltra-
tion in the bladder. We further analyzed local T lymphocyte subsets based on the expression of T-cell memory
Figure 3. Baculovirus as a gene transfer vector for bladder cancer therapy. (A) Bladder weight measurement.
Le: e animal experiment protocol used. Mice were inoculated with MB49 tumor cells intravesically and
subsequently treated three times with baculoviral vectors (days 7, 14, and 21). Right: Mice were treated with
baculoviral vectors expressing CD40L, IL-15, or both. Control animals were treated with PBS, BCG, or empty
baculoviral vector BacPAK6. Mice were sacriced on day 35 post-tumor inoculation and bladders were
collected for weight measurement. e data represent mean + s.d., n = 5 per group. (B) Survival curves to
demonstrate anti-tumor eects of baculoviral gene therapy aer 3 instillations in the mouse bladder. e tumor
inoculation and virus/BCG instillations were performed as shown in (A). Survival curves till day 125 are shown.
e statistical analysis was performed using the log rank test. n = 20 for PBS, BacPAK6, and BCG groups and
10 for BV-CD40L, BV-IL-15 and BV-CD40L+ BV-IL-15 groups. (C) H&E staining shows MB49 bladder tumor
development. Bladders were collected on day 35 post-tumor inoculation.
www.nature.com/scientificreports/
7
Scientific RepoRts | 6:27455 | DOI: 10.1038/srep27455
markers CD44 and CD62L (Fig.6A). Upon repeated intravesical instillations with baculoviral vectors or BCG, the
naïve T cells dened as CD44lowCD62Lhigh were markedly decreased, accompanied by increased percentages of
CD44highCD62Llow eector T cells and CD44highCD62Lhigh memory T cells in both CD4+ and CD8+ com-
partments (Fig.6B). ese changes were most obvious aer co-instillations with BV-CD40L and BV-IL15. e
percentages of eector and memory T cells, which play a pivotal role in the development of immune responses,
increased from 12% in the PBS group to 49% in the CD40L+ IL15 group in the CD4+ T cell population and 11%
in the PBS group to 44% in the CD40L+ IL15 group in the in the CD8+ T cell population. Lymphocytic inltra-
tions aer intravesical instillation of BacPAK6 and BV-CD40L/BV-IL15 were also conrmed by immunostaining
(Fig. S5). Such pronounced lymphocyte accumulation triggered by baculoviral transduction might play impor-
tant roles in the above observed bladder tumor regression.
Discussion
Bladder cancer is the most common form of malignancy in the urinary tract. At initial diagnosis the majority
of bladder cancer are NMIBC. e recurrence rate of NMIBC aer transurethral resection could be as high as
70%, necessitating adjuvant therapy to control recurrence and progression1,2. e current study demonstrated for
the rst time that baculoviral transduction is a possible new approach for adjuvant treatment of bladder cancer.
Baculoviruses can be used not only for immune stimulation, but also for therapeutic gene delivery, providing
a new combinatorial approach that harnesses the power of immunotherapy and gene therapy in a single viral
ve ctor.
Given their intrinsic potent immunostimulatory property and ecient cell transduction capacity, baculovi-
ral vectors oer an unprecedented advantage over many other viral vectors in immunotherapy against bladder
cancer. We observed that intravesical instillation of empty baculoviral vectors, without the use of any therapeutic
genes, was eective in prolonging the life of mice with established orthotopic bladder cancer. is nding is
consistent with previous studies that found that aer being injected into the animal body, baculovirus can elicit
protective innate immune responses30,35–38. is property has been exploited to protect animals from lethal chal-
lenges with encephalomyocarditis virus35 and inuenza virus37, suppress liver cirrhosis induced by dimethylni-
rosamine38, and inhibit tumor growth30.
Presumably, the initial step aer baculovirus instillation in the bladder should be the binding of the virus to
the urothelium, aer which the virus can enter both normal and malignant cells, resulting in urothelial activation
Figure 4. Inltration of innate immune cells into the bladder aer intravesical instillation with PBS, BCG,
or baculoviral vectors. ree bladder samples per group were collected 16 hours aer the third instillation for
ow cytometric analysis. Neutrophils were dened as live CD45.2+Ly-6G+ cells; inammatory monocytes were
dened as live CD45.2+Ly-6GLy-6ChighCD11b+ cells. Representative FACS plots of two repeated experiments
are shown.
www.nature.com/scientificreports/
8
Scientific RepoRts | 6:27455 | DOI: 10.1038/srep27455
and subsequent inammatory responses within the bladder. Virus antigens can be presented on the surfaces
of transduced urothelial and antigen-presenting cells in the context of MHC class II to stimulate CD4+ T cells.
Intracellular viral antigens will predominantly induce 1 immune response, resulting in secretion of 1-type
cytokines. Indeed, we have observed the induction of an array of cytokines by intravesical instillation of bacu-
lovirus. ese baculovirus-induced cytokines include 1-type of cytokines such as IFN-gamma, IL-2, TNF-α ,
and IL12 as well as 2-type of cytokines IL-13 and IL-10. Although the specic role of each of these cytokines in
orchestrating baculovirus-induced anti-tumor immunity is not clear at this point, a high-level expression of 1
cytokines has been observed to be associated with BCG responders and eective BCG therapy of bladder cancer
depends largely on proper induction of the 1 immune pathway. Baculovirus DNA contains abundant CpG
motifs32,33, and CpG stimulates host production of IL-12 and drives 1 immune responses, providing potential
therapeutic value in treating cancer.
Previous studies have further demonstrated that baculovirus potentiates adaptive immune responses by trans-
ducing professional antigen-presenting cells and inducing INF-α and INF-β
28,29,31,39. e initiation of an adaptive
immune response is critically dependent on the activation, functional maturation and migration of dendritic
cells. Baculovirus can transduce dendritic cells, activate these cells via the interaction with intracellular toll-like
receptor 9, and promote their maturation28,29,31,32,37,39. ese immune stimulatory eects can possibly be har-
nessed to promote systemic anti-tumor immunity. is hypothesis is supported by the current study using a
syngeneic model system that provides intact immune functions and allows the study of therapeutic vaccines
against bladder cancer. With this mouse model, we demonstrated that tumor-bearing animals that were cured by
baculovirus instillation survived aer the 2nd tumor challenge.
To test the idea of using wild-type baculovirus as an adjunct in the design of antitumor therapies, Takaku
et al. have examined the eects of intravenously injected baculoviruses that do not express foreign genes on
anti-tumor immunity in immunocompetent mice with B16 mouse melanoma and demonstrated the activation of
NK cell-dependent antitumor immunity by baculovirus30. Further studies from the same laboratory indicate that
baculovirus-induced antitumor action possibly also involves acquired immunity by enhancing tumor-specic
cytotoxic T lymphocyte (CTL) responses and tumor-specic antibody production31,39. ese ndings highlight
Figure 5. Inltration of T cells into the bladder aer intravesical instillation with PBS, BCG or baculoviral
vectors. ree bladder samples per group were collected 16 hours aer the third instillation for ow cytometric
analysis. T cells were gated as live CD45.2+CD3ε
+NK1.1 cells. e gated T cells were further gated as γ δ TCR
positive or negative cells, and the latter population was assessed for CD4 or CD8a expression. Representative
FACS plots of two repeated experiments are shown. Bottom: Average numbers of T cell subpopulations are
shown.
www.nature.com/scientificreports/
9
Scientific RepoRts | 6:27455 | DOI: 10.1038/srep27455
that the intrinsic immunostimulatory property of baculovirus can possibly be favorably exploited for cancer
immunotherapy. However, since baculoviruses are highly sensitive to the inactivation by serum complement pro-
teins34, systemically delivered baculoviral vectors usually do not transduce target cells. In the above studies from
Takaku’s lab, high titers of baculoviruses were used to overcome the ability of the complement to neutralize the
viruses, yet there was no demonstration of baculovirus-mediated transgene expression30,31,39.
As shown in the current study, the ecacy of baculovirus-mediated bladder cancer therapy was further
improved by including therapeutic genes into baculoviral vectors. In this regard, we showed the strong anti-tumor
eects of baculoviral vector-mediated CD40L or IL-15 expression in an animal model with aggressively growing
bladder cancer. Local transduction, either in normal or malignant bladder cells, that suciently activates only
targeted cytokines would be an attractive strategy to improve the eciency of immunotherapy and to diminish
possible side eects associated with systemic exposure. Using an intravesical liposomal gene delivery approach in
a mouse bladder cancer model, IL-15 gene therapy has been demonstrated to be a new promising approach for
bladder cancer treatment40. Adenoviral vectors encoding IL-15 were also found to be able to abolish tumorigenic-
ity of murine bladder tumor MB4941. Following preclinical testing in experimental bladder cancer animal models,
a Phase I/IIa clinical trial using adenoviral vector expressing CD40L for immunogene therapy of bladder cancer
has been performed and demonstrated a boosted immune activation14.
CD40L, also called CD154, is a potent 1 immune stimulator. CD40L is mainly expressed on activated CD4+
T cells and interacts with CD40 expressed on a wide range of antigen-presenting cells (APCs) and malignant
cells42. CD40/CD40L ligation may activate APCs, stimulating their maturation to present antigens to T cells and
ensuring the generation of antigen-specic CTLs. CD40L stimulation can also activate secretion of cytokines,
such as IL-12 and IFN-γ , leading to a 1 response, and abrogate the suppressive eect of T regulatory cells.
Moreover, CD40 expression has been found on various tumor cells, and CD40/CD40L ligation can inhibit pro-
liferation and induce apoptosis directly in tumor cells by activation of NF-κ B, AP-1, CD95, or caspase-depended
Figure 6. T lymphocyte subset analysis upon repeated intravesical instillations with PBS, baculoviral
vectors or BCG. ree mouse bladder samples per group were collected 16 hours aer the third instillation
and cut into small pieces for enzyme digestion. e collected bladder cells were stained with CD4, CD8, CD62L
and CD44 antibodies conjugated with uorescence, and the results were analyzed by ow cytometry. (A)
CD4+ (le) and CD8+ (right) cells were gated for T lymphocyte subset analysis. Naïve T cells were dened as
CD44lowCD62Lhigh, memory T cells as CD44highCD62Lhigh and eector T cells as CD44highCD62Llow.
FACS plots shown are representative of two independent experiments. (B) Average numbers of CD4+ (le) and
CD8+ (right) T lymphocyte subsets.
www.nature.com/scientificreports/
10
Scientific RepoRts | 6:27455 | DOI: 10.1038/srep27455
pathways43. Acting both as an activator of immune cells and cancer cell death inducer, CD40L is currently under
intensive investigation for its potent anti-tumor eects.
IL-15 is a 15-kDa cytokine member in the IL-2 family. While IL-15 has pleiotropic immune-enhancing
activities, it plays a pivotal role in the generation and maintenance of memory CD8+ T cells and NK cells44. A
rst-in-human Phase I clinical trial that involved recombinant IL-15 in patients with refractory metastatic malig-
nant melanoma and metastatic renal cell cancer has just completed45. e study revealed that NK cells and γ δ T
lymphocytes in blood were most dramatically aected, followed by CD8+ memory cells, by IL-15 administration.
Since IL-15 activates anti-tumor eectors such as NK cells, γ δ T lymphocytes, and memory phenotype CD8+ T
cells and exerts more long-lasting antitumor eects, it might be especially suitable for immunotherapy of NMIBC.
When viewed from the interaction perspective, IL-15 can induce CD40 expression on conventional DCs
and interaction between CD40 on conventional DCs and CD40L on plasmacytoid DCs leads to IL-12 produc-
tion by conventional DCs, which is essential for CpG-induced immune activation46. On the other hand, stim-
ulation of monocytes or DCs with the ligation of CD40 with CD40L or an agonistic anti-CD40 monoclonal
antibody can coordinately induce IL-15 and IL-15Ra expression. IL-15Ra on the cell surfaces of monocytes
or DCs can then present IL-15 in trans to cells such as CD8+ memory T cells and NK cells that express IL-2/
IL-15Rb47. Also, IL-15 can cooperate with CD40L to increase growth of normal and follicular lymphoma
B-cells48. Although the mechanism of their interaction is still unclear, the baculoviral transduction-mediated
co-expression of IL-15 and CD40L, as demonstrated in the current study, exhibits potent therapeutic ecacy
for bladder cancer.
e bladder is a conned compartment in which an immunotherapy agent can be given in a high concentra-
tion to eectively recruit immune cells and activate them locally, thus serving as an ideal target organ for immu-
notherapy. Normally, the bladder can be regarded as an organ that is not inltrated by large numbers of immune
cells3,49. is study shows that following intravesical baculoviral transduction, the bladder is inltrated by mono-
nuclear leukocytes, including inammatory monocytes, CD4+, CD8+, and γ δ T lymphocytes. We focused mainly
on immune cell inltration aer repeated instillations since this is a clinically relevant scheme. is inltration
pattern of immune cells aer repeated instillations of baculoviral vectors is in general similar to the one observed
following intravesical BCG instillation3,49. However, instillation with baculoviral vectors expressing CD40L
and IL-15 signicantly increased the inux of CD3+ T lymphocytes with higher absolute numbers of CD4+,
CD8+, and γ δ T lymphocytes. is increase is much more pronounced when looking at γ δ T lymphocytes, the
non-MHC-restricted cytotoxic cells that can initiate cytotoxicity aer infection, even though BCG is known to
be able to induce the proliferation of γ δ T lymphocytes50. Also noteworthy, intravesical instillation of baculovirus
alone was able to increase the number of γ δ T lymphocytes in the bladder. While the antitumor activity of bacu-
loviral transduction in the current bladder cancer model possibly depends on the interplay of dierent immune
cells, the correlation between increased numbers of γ δ T lymphocytes in the bladder and anti-cancer ecacies
achieved with dierent therapeutics used for bladder instillation indicates that this type of cytotoxic eector cells
play an important role in treatment success.
In conclusion, we have demonstrated in a syngeneic orthotopic animal model of bladder cancer that insect
baculovirus can be used as a new agent for bladder cancer therapy with such functions as stimulating local innate
immune reactions, promoting systemic anti-tumor immune responses, and delivering therapeutic genes. When
tested as an adjuvant therapy for urothelial malignancies, the success of the approach will benet bladder cancer
patients who fail conventional BCG immunotherapy or who are intolerant of BCG treatment. When used as a
single therapy, this intravesical immunotherapy approach may improve bladder preservation and the quality of
life for patients, thus delivering better health outcomes. Hence, intravesical instillation of recombinant baculoviral
vectors holds potential to develop into an advanced therapy for NMIBC.
References
1. Chiong, E. & Esuvaranathan, . New therapies for non-muscle-invasive bladder cancer. World J. Urol. 28, 71–78 (2010).
2. Patel, S. G., Cohen, A., Weiner, A. B. & Steinberg, G. D. Intravesical therapy for bladder cancer. Expert Opin. Pharmacother. 16,
889–901 (2015).
3. Fuge, O., Vasdev, N., Allchorne, P. & Green, J. S. Immunotherapy for bladder cancer. es. ep. Urol. 7, 65–79 (2015).
4. Bure, J. Virus therapy for bladder cancer. Cytoine Growth Factor ev. 21, 99–102 (2010).
5. Lee, S. S., Eisenlohr, L. C., McCue, P. A., Mastrangelo, M. J. & Lattime, E. C. Intravesical gene therapy: in vivo gene transfer using
recombinant vaccinia virus vectors. Cancer es. 54, 3325–3328 (1994).
6. Gomella, L. G. et al. Phase i study of intravesical vaccinia virus as a vector for gene therapy of bladder cancer. J. Urol. 166, 1291–1295
(2001).
7. Siemens, D. ., Crist, S., Austin, J. C., Tartaglia, J. & atli, T. L. Comparison of viral vectors: gene transfer eciency and tissue
specicity in a bladder cancer model. J. Urol. 170, 979–984 (2003).
8. Fodor, I. et al. Vaccinia virus mediated p53 gene therapy for bladder cancer in an orthotopic murine model. J. Urol. 173, 604–609
(2005).
9. Morris, B. D. et al. Adenoviral-mediated gene transfer to bladder in vivo. J. Urol. 152, 506–509 (1994).
10. Sutton, M. A. et al. Adenovirus-mediated suicide gene therapy for experimental bladder cancer. Urology 49, 173–180 (1997).
11. uball, J. et al. Successful adenovirus-mediated wild-type p53 gene transfer in patients with bladder cancer by intravesical vector
instillation. J. Clin. Oncol. O. J. Am. Soc. Clin. Oncol. 20, 957–965 (2002).
12. Pagliaro, L. C. et al. epeated intravesical instillations of an adenoviral vector in patients with locally advanced bladder cancer: a
phase I study of p53 gene therapy. J. Clin. Oncol. O. J. Am. Soc. Clin. Oncol. 21, 2247–2253 (2003).
13. Losog, A. S. I., Fransson, M. E. & Totterman, T. T. H. AdCD40L gene therapy counteracts T regulatory cells and cures aggressive
tumors in an orthotopic bladder cancer model. Clin. Cancer es. O. J. Am. Assoc. Cancer es. 11, 8816–8821 (2005).
14. Malmström, P.-U. et al. AdCD40L immunogene therapy for bladder carcinoma–the rst phase I/IIa trial. Clin. Cancer es. O. J. Am.
Assoc. Cancer es. 16, 3279–3287 (2010).
15. Bure, J. M. et al. A rst in human phase 1 study of CG0070, a GM-CSF expressing oncolytic adenovirus, for the treatment of
nonmuscle invasive bladder cancer. J. Urol. 188, 2391–2397 (2012).
www.nature.com/scientificreports/
11
Scientific RepoRts | 6:27455 | DOI: 10.1038/srep27455
16. Hanel, E. G. et al. A novel intravesical therapy for supercial bladder cancer in an orthotopic model: oncolytic reovirus therapy.
J. Urol. 172, 2018–2022 (2004).
17. Shiau, A. L., Lin, P. ., Chang, M. Y. & Wu, C. L. etrovirus-mediated transfer of prothymosin gene inhibits tumor growth and
prolongs survival in murine bladder cancer. Gene er. 8, 1609–1617 (2001).
18. imura, T. et al. Antitumor immunity against bladder cancer induced by ex vivo expression of CD40 ligand gene using retrovirus
ve cto r. Cancer Gene er. 10, 833–839 (2003).
19. Dumey, N. et al. In vivo retroviral mediated gene transfer into bladder urothelium results in preferential transduction of tumoral
cells. Eur. Urol. 47, 257–263 (2005).
20. iuchi, E. et al. Highly ecient gene delivery for bladder cancers by intravesically administered replication-competent retroviral
vectors. Clin. Cancer es. O. J. Am. Assoc. Cancer es. 13, 4511–4518 (2007).
21. iuchi, E. et al. Inhibition of orthotopic human bladder tumor growth by lentiviral gene transfer of endostatin. Clin. Cancer es.
O. J. Am. Assoc. Cancer es. 10, 1835–1842 (2004).
22. Hadaschi, B. A. et al. Oncolytic vesicular stomatitis viruses are potent agents for intravesical treatment of high-ris bladder cancer.
Cancer es. 68, 4506–4510 (2008).
23. Airenne, . J. et al. Baculovirus: an Insect-derived Vector for Diverse Gene Transfer Applications. Mol. er. 21, 739–749 (2013).
24. Hofmann, C. et al. Ecient gene transfer into human hepatocytes by baculovirus vectors. Proc. Natl. Acad. Sci. USA 92, 10099
(1995).
25. ost, T. A., Condreay, J. P. & Jarvis, D. L. Baculovirus as versatile vectors for protein expression in insect and mammalian cells. Nat.
Biotechnol. 23, 567–575 (2005).
26. Hu, Y.-C. Baculovirus: a promising vector for gene therapy? Curr. Gene er. 10, 167 (2010).
27. Wang, S. & Balasundaram, G. Potential cancer gene therapy by baculoviral transduction. Curr. Gene er. 10, 214–225 (2010).
28. Hervas-Stubbs, S., ueda, P., Lopez, L. & Leclerc, C. Insect Baculoviruses Strongly Potentiate Adaptive Immune esponses by
Inducing Type I IFN. J. Immunol. 178, 2361–2369 (2007).
29. Strauss, . et al. Baculovirus-based Vaccination Vectors Allow for Ecient Induction of Immune esponses Against Plasmodium
falciparum Circumsporozoite Protein. Mol. er. 15, 193–202 (2007).
30. itajima, M. et al. Induction of Natural iller Cell-dependent Antitumor Immunity by the Autographa californica Multiple Nuclear
Polyhedrosis Virus. Mol. er. 16, 261–268 (2007).
31. Suzui, T., Oo Chang, M., itajima, M. & Taau, H. Induction of antitumor immunity against mouse carcinoma by baculovirus-
infected dendritic cells. Cell. Mol. Immunol. 7, 440–446 (2010).
32. Abe, T. et al. Involvement of the Toll-Lie eceptor 9 Signaling Pathway in the Induction of Innate Immunity by Baculovirus. J. Virol.
79, 2847–2858 (2005).
33. Abe, T. et al. Baculovirus Induces Type I Interferon Production through Toll-Lie eceptor-Dependent and -Independent Pathways
in a Cell-Type-Specic Manner. J. Virol. 83, 7629–7640 (2009).
34. Hofmann, C., Hüser, A., Lehnert, W. & Strauss, M. Protection of baculovirus-vectors against complement-mediated inactivation by
recombinant soluble complement receptor type 1. Biol. Chem. 380, 393–395 (1999).
35. Gronowsi, A. M., Hilbert, D. M., Sheehan, . C. F., Garotta, G. & Schreiber, . D. Baculovirus Stimulates Antiviral Eects in
Mammalian Cells. J. Virol. 73, 9944–9951 (1999).
36. Bec, N. B., Sidhu, J. S. & Omiecinsi, C. J. Baculovirus vectors repress phenobarbital-mediated gene induction and stimulate
cytoine expression in primary cultures of rat hepatocytes. Gene er. 7, 1274–1283 (2000).
37. Abe, T. et al. Baculovirus Induces an Innate Immune esponse and Confers Protection from Lethal Inuenza Virus Infection in
Mice. J. Immunol. 171, 1133–1139 (2003).
38. Nishibe, Y. et al. Baculovirus-mediated interferon alleviates dimethylnitrosamine-induced liver cirrhosis symptoms in a murine
model. Gene er. 15, 990–997 (2008).
39. itajima, M. & Taau, H. Induction of antitumor acquired immunity by baculovirus Autographa californica multiple nuclear
polyhedrosis virus infection in mice. Clin. Vaccine Immunol. CVI 15, 376–378 (2008).
40. Matsumoto, . et al. Intravesical interleuin-15 gene therapy in an orthotopic bladder cancer model. Hum. Gene er. 22, 1423–1432
(2011).
41. Ninalga, C., Losog, A., levenfeldt, M., Essand, M. & Tötterman, T. H. CpG oligonucleotide therapy cures subcutaneous and
orthotopic tumors and evoes protective immunity in murine bladder cancer. J. Immunother. Hagerstown Md 1997 28, 20–27 (2005).
42. Hassan, S. B., Sørensen, J. F., Olsen, B. N. & Pedersen, A. E. Anti-CD40-mediated cancer immunotherapy: an update of recent and
ongoing clinical trials. Immunopharmacol. Immunotoxicol. 36, 96–104 (2014).
43. ornilu, A., emona, H. & Dymica-Piearsa, V. Multifunctional CD40L: pro- and anti-neoplastic activity. Tumour Biol. 35,
9447–9457 (2014).
44. Steel, J. C., Waldmann, T. A. & Morris, J. C. Interleuin-15 biology and its therapeutic implications in cancer. Trends Pharmacol. Sci.
33, 35–41 (2012).
45. Conlon, . C. et al. edistribution, Hyperproliferation, Activation of Natural iller Cells and CD8 T Cells, and Cytoine Production
During First-in-Human Clinical Trial of ecombinant Human Interleuin-15 in Patients With Cancer. J. Clin. Oncol. 33, 74–82
(2015).
46. uwajima, S. et al. Interleuin 15–dependent crosstal between conventional and plasmacytoid dendritic cells is essential for CpG-
induced immune activation. Nat. Immunol. 7, 740–746 (2006).
47. Waldmann, T. A. e Biology of IL-15: Implications for Cancer erapy and the Treatment of Autoimmune Disorders. J. Investig.
Dermatol. Symp. Proc. 16, S28–S30 (2013).
48. Epron, G. et al. Monocytes and T cells cooperate to favor normal and follicular lymphoma B-cell growth: role of IL-15 and CD40L
signaling. Leuemia 26, 139–148 (2012).
49. Biot, C. et al. Preexisting BCG-Specic T Cells Improve Intravesical Immunotherapy for Bladder Cancer. Sci. Transl. Med. 4,
137ra72–137ra72 (2012).
50. Lee, J., Choi, ., Olin, M. ., Cho, S.-N. & Molitor, T. W. γ δ T Cells in Immunity Induced by Mycobacterium bovis Bacillus Calmette-
Guérin Vaccination. Infect. Immun . 72, 1504–1511 (2004).
Acknowledgements
is work was supported by the Singapore Ministry of Healths National Medical Research Council (NMRC/
CIRG/1367/2013; NMRC/CIRG/1406/2014) and Institute of Bioengineering and Nanotechnology (Biomedical
Research Council, Agency for Science, Technology and Research, Singapore).
Author Contributions
W.X.A, Y.Z., T.K., C.W. and C.C. collection and assembly of data, data analysis and interpretation, and manuscript
writing; H.C.T., R.M. and K.E. conception, design, and manuscript preparation. S.W. conception, design,
manuscript preparation, and grant support. All authors reviewed the manuscript.
www.nature.com/scientificreports/
12
Scientific RepoRts | 6:27455 | DOI: 10.1038/srep27455
Additional Information
Supplementary information accompanies this paper at http://www.nature.com/srep
Competing nancial interests: e authors declare no competing nancial interests.
How to cite this article: Ang, W. X. et al. Local Immune Stimulation by Intravesical Instillation of Baculovirus
to Enable Bladder Cancer erapy. Sci. Rep. 6, 27455; doi: 10.1038/srep27455 (2016).
is work is licensed under a Creative Commons Attribution 4.0 International License. e images
or other third party material in this article are included in the article’s Creative Commons license,
unless indicated otherwise in the credit line; if the material is not included under the Creative Commons license,
users will need to obtain permission from the license holder to reproduce the material. To view a copy of this
license, visit http://creativecommons.org/licenses/by/4.0/

Supplementary resource (1)

... Since then, the viral genome has been modified and manipulated to improve transduction efficiency and simplify its production. Consequently, several vector systems have been developed, including BacMam, Bac-to-Bac, MultiBac, and derivatives of these AcMNPV transfer vectors [92][93][94][95]. ...
Article
Full-text available
Baculoviruses are enveloped, insect-specific viruses with large double-stranded DNA genomes. Among all the baculovirus species, Autographa californica multiple nucleopolyhedrovirus (AcMNPV) is the most studied. Due to its characteristics regarding biosafety, narrow host range and the availability of different platforms for modifying its genome, AcMNPV has become a powerful biotechnological tool. In this review, we will address the most widespread technological applications of baculoviruses. We will begin by summarizing their natural cycle both in larvae and in cell culture and how it can be exploited. Secondly, we will explore the different baculovirus-based protein expression systems (BEVS) and their multiple applications in the pharmaceutical and biotechnological industry. We will focus particularly on the production of vaccines, many of which are either currently commercialized or in advanced stages of development (e.g., Novavax, COVID-19 vaccine). In addition, recombinant baculoviruses can be used as efficient gene transduction and protein expression vectors in vertebrate cells (e.g., BacMam). Finally, we will extensively describe various gene therapy strategies based on baculoviruses applied to the treatment of different diseases. The main objective of this work is to provide an extensive up-to-date summary of the different biotechnological applications of baculoviruses, emphasizing the genetic modification strategies used in each field.
... The BV genome is approximately 134 kb, can harbor large transgenes, and is comparatively easy to engineer [132,133]. There is no pre-existing anti-BV immunity in humans, proposing them as a potentially effective therapy option, but there are no clinical trials employing them yet [134]. The BV can avoid some of the problems carried by mammalian viral vectors, while also providing another option for cancer gene therapy. ...
Article
Full-text available
Introduction: : High-grade gliomas (HGG) are the most common malignant primary brain tumors in adults, with a median survival of ~18 months. The standard of care (SOC) is maximal safe surgical resection, and radiation therapy with concurrent and adjuvant temozolomide. This protocol remains unchanged since 2005, even though HGG median survival has marginally improved. Areas covered: Gene therapy was developed as a promising approach to treat HGG. Here we review completed and ongoing clinical trials employing viral and non-viral vectors for adult and pediatric HGG, as well as the key supporting preclinical data. Expert opinion: These therapies have proven safe, and pre- and post-treatment tissue analyses demonstrated tumor cell lysis, increased immune cell infiltration, and increased systemic immune function. Although viral therapy in clinical trials has not yet significantly extended survival of HGG, promising strategies are being tested. Oncolytic HSV vectors have shown promising results both for adult and pediatric HGG. A recently published study demonstrated that HG47Δ improved survival in recurrent HGG. Likewise, PVSRIPO has shown survival improvement compared to historical controls. It is likely that further analysis of these trials will stimulate the development of new administration protocols, and new therapeutic combinations which will improve HGG prognosis.
... BCG can prolong the progressionfree interval after initial tumor resection in patients with NMIBC. It exhibits a nonspecific immune response to reduce tumor burden by increasing the number of natural killer (NK) cells and CD8 + T cells and by cytokine secretion, including that of interleukin (IL)-2, IL-12, IL-18, and interferon-gamma (INF-g) (5,6). However, there are no biomarkers for predicting the response to BCG. ...
Article
Full-text available
Bladder cancer (BCa) is one of the most common malignant tumors that cause death. Approximately 75%–85% of BCa develop into non-muscle-invasive bladder cancer (NMIBC). Bacillus Calmette-Guérin (BCG) is the gold standard for avoiding cystectomy in the treatment of NMIBC. Unfortunately, up to 30% of patients do not respond to BCG treatment, and up to 70% of BCG responders relapse. The United States Food and Drug Administration (FDA) approved valrubicin (1998) and pembrolizumab (2020) for the treatment of BCG-unresponsive (BCGu) NMBIC. However, the complete remission rate for valrubicin and pembrolizumab was only 16% and 40.6%, respectively. ALT-803 (N-803) is an IL-15 superagonist and reduces tumor burden by promoting the proliferation and activation of NK cells and CD8⁺ T cells. The FDA received (23 May 2022) and accepted to review (28 July 2022) the marketing submission of ALT-803 plus BCG for the treatment of BCGu NMIBC. However, the FDA previously rejected the application for oportuzumab monatox (OM) due to a lack of data comparing it with pembrolizumab on August 20, 2021. Interestingly, the clinical efficacy and safety of ALT-803 were higher than that of pembrolizumab and OM, suggesting that ALT-803 may be approved by FDA. This review aims to further knowledge of the preclinical and clinical evidence of ALT-803 in the treatment of NMIBC and discuss its translational potential.
... Superficial bladder tumors were established using previously published protocols. 44 Briefly, after anesthetizing mice with isoflurane, mouse urethra was catheterized and instilled with 100 mL of poly(l-lysine) (PLL) (0.01 mg/mL) for 15 min. After emptying the bladder, 25,000 MB49 cells in 100 mL of HBSS were instilled into mouse bladders for 30 min. ...
Article
Full-text available
Interferon alpha (IFNα) gene therapy is emerging as a new treatment option for patients with non-muscle invasive bladder cancer (NMIBC). Adenoviral vectors expressing IFNα have shown clinical efficacy treating Bacillus Calmette Guerin (BCG) unresponsive bladder cancer (BLCA). However, transient transgene expression and adenoviral immunogenicity may limit therapeutic activity. Lentiviral vectors can achieve stable transgene expression and are less immunogenic. In this study, we evaluated lentiviral vectors expressing murine IFNα (LV-IFNα) and demonstrate IFNα expression by transduced murine BLCA cell lines, bladder urothelium and within the urine following intravesical instillation. Murine BLCA cell lines (MB49 and UPPL1541) were sensitive to IFN-mediated cell death after LV-IFNα, whereas BBN975 was inherently resistant. Upregulation of IL-6 predicted sensitivity to IFN-mediated cell death mediated by caspase signaling, which when inhibited abrogated IFN-mediated cell killing. Intravesical therapy with LV-IFNα/Syn3 in a syngeneic BLCA model significantly improved survival, and molecular analysis of treated tumors revealed upregulation of apoptotic and immune cell-mediated death pathways. In particular, biomarker discovery analysis identified three clinically actionable targets, PD-L1, EGFR, and ALDHA1A, in murine tumors treated with LV-IFNα/Syn3. Our findings warrant the comparison of adenoviral and LV-IFNα, and the study of novel combination strategies with IFNα gene therapy for the BLCA treatment.
... This could be relief by reducing the instillation duration. 33,36,37 p53 gene therapy experiences several failed clinical trials and it is still far from 100% efficacy. There are barriers that blocked gene therapy efficacy. ...
Article
Full-text available
Chemotherapy and radiotherapy are gold standard treatment for bladder cancer (BC) for over 50 years. The efficacy on early stage BC patients is virtuous. However, patients with aggressive cancer growth benefited less from the therapy. Aberrant p53 was found in more than 50% of high-grade BC patients. Therefore, targeting p53 in a subset of high-grade BC patients expressing aberrant p53 is a promising strategy. In this paper, p53 role in BC carcinogenesis is discussed. Followed by p53-targeting strategies in current BC treatment. Besides, p53-targeting strategies that have been implemented in other types of cancer and their potential to be adapted in BC will be deliberated. Although targeting p53 is promising, none of the strategies studied were successfully implemented in healthcare settings. Restoration of p53 as the guardian of the genome is an exciting area for translational research. It has potential to replace the genotoxic chemotherapy and radiotherapy, thus, eliminating the notorious painful sideeffects on a subset of high-grade BC patients. Searches were performed on PubMed and Google Scholar web using the keywords “bladder cancer” or “urothelial cancer” or “urothelial cell carcinoma” and “p53”. Only full papers of research articles and review papers were included for analysis. Papers were categorized as either p53 function, current treatment using p53 and future potential treatment using p53 for details analysis.
... In view of our results, further investigation is warranted to elucidate the complete immune response (other immune subsets of cells or local and systemic immune biomarkers) induced by M. brumae compared to BCG to completely understand the role of mycobacteria in modulating the tumor microenvironment. Different strategies for superficial bladder treatment are currently being proposed: diverse bacteria 30-32 , modified virus 33,34 , chemotherapeutic agents 35 , molecules that target specific immune receptors 36 , peptide vaccines 37 , plant derived compounds 38 , etc. Mycobacteria can have a role to synergy to these treatments, as has been already demonstrated 35,37,38 , enabling to reduce the toxicity and resistance to a single treatment. Further understanding of the immune reaction associated with mycobacteria treatment could be critical for identifying the benefits of new immunotherapeutic possibilities. ...
Article
Full-text available
Abstract The standard treatment for high-risk non-muscle invasive bladder cancer (BC) is the intravesical administration of live Mycobacterium bovis BCG. Previous studies suggest improving this therapy by implementing non-pathogenic mycobacteria, such as Mycobacterium brumae, and/or different vehicles for mycobacteria delivery, such as an olive oil (OO)-in-water emulsion. While it has been established that BCG treatment activates the immune system, the immune effects of altering the mycobacterium and/or the preparation remain unknown. In an orthotopic murine BC model, local immune responses were assessed by measuring immune cells into the bladder and macromolecules in the urine by flow cytometry and multiplexing, respectively. Systemic immune responses were analyzed by quantifying sera anti-mycobacteria antibody levels and recall responses of ex vivo splenocytes cultured with mycobacteria antigens. In both BCG- and M. brumae-treated mice, T, NK, and NKT cell infiltration in the bladder was significantly increased. Notably, T cell infiltration was enhanced in OO-in-water emulsified mycobacteria-treated mice, and urine IL-6 and KC concentrations were elevated. Furthermore, mycobacteria treatment augmented IgG antibody production and splenocyte proliferation, especially in mice receiving OO-in-water emulsified mycobacteria. Our data demonstrate that intravesical mycobacterial treatment triggers local and systemic immune responses, which are most significant when OO-in-water emulsified mycobacteria are used.
... Recently, the intravesical instillation of transgene devoid baculovirus is found to elicit local immune stimulation by upregulating a set of Th-1-type cytokines in orthotopic bladder tumours in mice (Ang et al. 2016). However, the application of such strategy for non-muscle invasive bladder cancer (NMIBC) in humans is awaited. ...
Chapter
Full-text available
Different strategies are being worked out for engineering the original baculovirus expression vector (BEV) system to produce cost-effective clinical biologics at commercial scale. To date, thousands of highly variable molecules in the form of heterologous proteins, virus-like particles, surface display proteins/antigen carriers, heterologous viral vectors and gene delivery vehicles have been produced using this system. These products are being used in vaccine production, tissue engineering, stem cell transduction, viral vector production, gene therapy, cancer treatment and development of biosensors. Recombinant proteins that are expressed and post-translationally modified using this system are also suitable for functional, crystallographic studies, microarray and drug discovery-based applications. Till now, four BEV-based commercial products (Cervarix®, Provenge®, Glybera® and Flublok®) have been approved for humans, and myriad of others are in different stages of preclinical or clinical trials. Five products (Porcilis® Pesti, BAYOVAC CSF E2®, Circumvent® PCV, Ingelvac CircoFLEX® and Porcilis® PCV) got approval for veterinary use, and many more are in the pipeline. In the present chapter, we have emphasized on both approved and other baculovirus-based products produced in insect cells or larvae that are important from clinical perspective and are being developed as preventive, diagnostic or therapeutic agents. Further, the potential of recombinant adeno-associated virus (rAAV) as gene delivery vector has been described. This system, due to its relatively extended gene expression, lack of pathogenicity and the ability to transduce a wide variety of cells, gained extensive popularity just after the approval of first AAV-based gene therapy drug alipogene tiparvovec (Glybera®). Numerous products based on AAV which are presently in different clinical trials have also been highlighted.
Article
Full-text available
Baculoviruses are insect pathogens widely used as biotechnological tools in different fields of life sciences and technologies. The particular biology of these entities (biosafety viruses 1; large circular double-stranded DNA genomes, infective per se; generally of narrow host range on insect larvae; many of the latter being pests in agriculture) and the availability of molecular-biology procedures (e.g., genetic engineering to edit their genomes) and cellular resources (availability of cell lines that grow under in vitro culture conditions) have enabled the application of baculoviruses as active ingredients in pest control, as systems for the expression of recombinant proteins (Baculovirus Expression Vector Systems—BEVS) and as viral vectors for gene delivery in mammals or to display antigenic proteins (Baculoviruses applied on mammals—BacMam). Accordingly, BEVS and BacMam technologies have been introduced in academia because of their availability as commercial systems and ease of use and have also reached the human pharmaceutical industry, as incomparable tools in the development of biological products such as diagnostic kits, vaccines, protein therapies, and—though still in the conceptual stage involving animal models—gene therapies. Among all the baculovirus species, the Autographa californica multiple nucleopolyhedrovirus has been the most highly exploited in the above utilities for the human-biotechnology field. This review highlights the main achievements (in their different stages of development) of the use of BEVS and BacMam technologies for the generation of products for infectious and noninfectious human diseases. Key points • Baculoviruses can assist as biotechnological tools in human health problems. • Vaccines and diagnosis reagents produced in the baculovirus platform are described. • The use of recombinant baculovirus for gene therapy–based treatment is reviewed.
Chapter
The discovery ofthe genome-editing tool CRISPR-Cas9 is revolutionizing the world ofgene therapy and will extend the gene therapy product pipeline. While applying gene therapy products, the main difficulty is an efficient and effective transfer of the nucleic acids carrying the relevant information to their target destination, the nucleus ofthe cells. Baculoviruses have shown to be very suitable transport vehicles for this task due to, inter alia, their ability to transduce mammalian/human cells without being pathogenic. This property allows the usage of baculovirus-transduced cells as cell therapy products, thus, combining the advantages of gene and cell therapy. To make such pharmaceuticals available for patients, a successful production and purification is necessary. In this chapter, we describe the generation of a pseudotyped baculovirus vector, followed by downstream processing using depth and tangential-flow filtration. This vector is used subsequently to transduce human mesenchymal stem cells. The production of the cells and the subsequent transduction process are illustrated.
Chapter
Accumulating evidence indicates that immune cells in the microenvironment play a significant role in tumor progression. Understanding the dynamic interplay between tumor cells and the host immune system is therefore of critical importance. While some immunotherapeutic strategies aim to specifically overcome hyporesponsiveness to tumor-associated antigens, others seek to nonspecifically activate the immune system; efficacy and toxicity are overriding, and often conflicting, concerns. This chapter, while occasionally providing brief historical perspectives, describes ongoing efforts in the area, including strategies for anticancer vaccination and adoptive cell transfer, the use of cytokines and development of oncolytic viruses, and the administration of checkpoint inhibitors. While significant successes have been achieved, the field is also characterized by notable failure, an indication that much remains unknown about factors that trigger and promote tumorigenesis, about current limitations in the effective delivery of therapeutic moieties, and about the workings of the immune system.
Article
Full-text available
It is nearly 40 years since Bacillus Calmette-Guérin (BCG) was first used as an immunotherapy to treat superficial bladder cancer. Despite its limitations, to date it has not been surpassed by any other treatment. As a better understanding of its mechanism of action and the clinical response to it have evolved, some of the questions around optimal dosing and treatment protocols have been answered. However, its potential for toxicity and failure to produce the desired clinical effect in a significant cohort of patients presents an ongoing challenge to clinicians and researchers alike. This review summarizes the evidence behind the established mechanism of action of BCG in bladder cancer, highlighting the extensive array of immune molecules that have been implicated in its action. The clinical aspects of BCG are discussed, including its role in reducing recurrence and progression, the optimal treatment regime, toxicity and, in light of new evidence, whether or not there is a superior BCG strain. The problems of toxicity and non-responders to BCG have led to development of new techniques aimed at addressing these pitfalls. The progress made in the laboratory has led to the identification of novel targets for the development of new immunotherapies. This includes the potential augmentation of BCG with various immune factors through to techniques avoiding the use of BCG altogether; for example, using interferon-activated mononuclear cells, BCG cell wall, or BCG cell wall skeleton. The potential role of gene, virus, or photodynamic therapy as an alternative to BCG is also reviewed. Recent interest in the immune check point system has led to the development of monoclonal antibodies against proteins involved in this pathway. Early findings suggest benefit in metastatic disease, although the role in superficial bladder cancer remains unclear.
Article
Full-text available
Interleukin-15 (IL-15) has significant potential in cancer immunotherapy as an activator of antitumor CD8 T and natural killer (NK) cells. The primary objectives of this trial were to determine safety, adverse event profile, dose-limiting toxicity, and maximum-tolerated dose of recombinant human IL-15 (rhIL-15) administered as a daily intravenous bolus infusion for 12 consecutive days in patients with metastatic malignancy. We performed a first in-human trial of Escherichia coli-produced rhIL-15. Bolus infusions of 3.0, 1.0, and 0.3 μg/kg per day of IL-15 were administered for 12 consecutive days to patients with metastatic malignant melanoma or metastatic renal cell cancer. Flow cytometry of peripheral blood lymphocytes revealed dramatic efflux of NK and memory CD8 T cells from the circulating blood within minutes of IL-15 administration, followed by influx and hyperproliferation yielding 10-fold expansions of NK cells that ultimately returned to baseline. Up to 50-fold increases of serum levels of multiple inflammatory cytokines were observed. Dose-limiting toxicities observed in patients receiving 3.0 and 1.0 μg/kg per day were grade 3 hypotension, thrombocytopenia, and elevations of ALT and AST, resulting in 0.3 μg/kg per day being determined the maximum-tolerated dose. Indications of activity included clearance of lung lesions in two patients. IL-15 could be safely administered to patients with metastatic malignancy. IL-15 administration markedly altered homeostasis of lymphocyte subsets in blood, with NK cells and γδ cells most dramatically affected, followed by CD8 memory T cells. To reduce toxicity and increase efficacy, alternative dosing strategies have been initiated, including continuous intravenous infusions and subcutaneous IL-15 administration. © 2014 by American Society of Clinical Oncology.
Article
Full-text available
The CD40 ligand is a type I transmembrane protein that belongs to a tumor necrosis factor (TNF) superfamily. It is present not only on the surface of activated CD4+ T cells, B cells, blood platelets, monocytes, and natural killer (NK) cells but also on cancer cells. The receptor for ligand is constitutively expressed on cells, TNF family protein: CD40. The role of the CD40/CD40L pathway in the induction of body immunity, in inflammation, or in hemostasis has been well documented, whereas its involvement in neoplastic disease is still under investigation. CD40L ligand may potentiate apoptosis of tumor cells by activation of nuclear factor-κB (NF-κB), AP-1, CD95, or caspase-depended pathways and stimulate host immunity to defend against cancer. Although CD40L has a major contribution to anti-cancer activity, many reports point at its ambivalent nature. CD40L enhance release of strongly pro-angiogenic factor, vascular endothelial growth factor (VEGF), and activator of coagulation, TF, the level of which is correlated with tumor metastasis. CD40L involvement in the inhibition of tumor progression has led to the emergence of not only therapy using recombinant forms of the ligand and vaccines in the treatment of cancer but also therapy consisting of inhibiting platelets-main source of CD40L. This article is a review of studies on the ambivalent role of CD40L in neoplastic diseases.
Article
Full-text available
Abstract The costimulatory molecule CD40 is a member of the tumor necrosis factor (TNF) receptor superfamily and is expressed on various antigen presenting cells (APCs) as well as some tumor cells. The binding to the natural ligand CD40L, which is expressed on T helper cells, leads to APC activation and thus enhancement of immune responses. Treatment with anti-CD40 monoclonal antibodies has been exploited in several cancer immunotherapy studies in mice and led to the development of anti-CD40 antibodies for clinical use. Here, Dacetuzumab and Lucatumumab are in the most advanced stage and are being tested as treatment for malignancies such as chronic lymphatic leukemia (CLL), Multiple Myeloma (MM), and non-Hodgkin's lymphoma (NHL). The promising results from these early clinical trials have encouraged clinical drug development in order to investigate the effect of CD40 mAbs in combination with other cancer immunotherapies, in particular interleukin (IL)-2. An in-depth analysis of this immunotherapy is provided elsewhere. In the present review, we provide an update of the most recent clinical trials with anti-CD40 antibodies. We present and discuss recent and ongoing clinical trials in this field, including clinical studies which combine anti-CD40 treatment with other cancer-treatments, such as Rituximab and Tremelimumab.
Article
Full-text available
Insect-derived baculoviruses have emerged as versatile and safe workhorses of biotechnology. Baculovirus expression vectors (BEVs) have been applied widely for crop and forest protection, as well as safe tools for recombinant protein production in insect cells. However, BEVs ability to efficiently transduce noninsect cells is still relatively poorly recognized despite the fact that efficient baculovirus-mediated in vitro and ex vivo gene delivery into dormant and dividing vertebrate cells of diverse origin has been described convincingly by many authors. Preliminary proof of therapeutic potential has also been established in preclinical studies. This review summarizes the advantages and current status of baculovirus-mediated gene delivery. Stem cell transduction, preclinical animal studies, tissue engineering, vaccination, cancer gene therapy, viral vector production, and drug discovery are covered.Molecular Therapy (2013); doi:10.1038/mt.2012.286.
Article
This study was designed to examine the potential for gene therapy in bladder in vivo using adenoviral vectors. Gene transfer to rat bladders was accomplished via direct intravesical instillation using a replication-defective adenoviral vector containing a marker gene encoding for Escherichia coli β-galactosidase (β-gal). Successful gene transfer was confirmed by analyzing bladder samples for DNA and RNA using polymerase chain reaction (PCR) with primers specific for β-gal and adeno sequences, detecting β-gal in full-thickness bladder wall using specific histochemical staining (X-gal) and documenting recombinant protein production. Bladder architecture was preserved, without evidence of distant spread of virus as assessed by PCR. Gene expression was evident for at least 7 days. In summary, bladder cells can be genetically altered using replication-deficient adenoviral vectors via simple intravesical instillation of vector. Introduction of exogenous genetic material is a potentially powerful therapeutic modality for immunomodulation of bladder neoplasms.
Article
PURPOSE: To study safety, feasibility, and biologic activity of adenovirus-mediated p53 gene transfer in patients with bladder cancer. PATIENTS AND METHODS: Twelve patients with histologically confirmed bladder cancer scheduled for cystectomy were treated on day 1 with a single intratumoral injection of SCH 58500 (rAd/p53) at cystoscopy at one dose level (7.5 × 10 ¹¹ particles) or a single intravesical instillation of SCH 58500 with a transduction-enhancing agent (Big CHAP) at three dose levels (7.5 × 10 ¹¹ to 7.5 × 10 ¹³ particles). Cystectomies were performed in 11 patients on day 3, and transgene expression, vector distribution, and biologic markers of transgene activity were assessed by molecular and immunohistochemical methods in tumors and normal bladder samples. RESULTS: Specific transgene expression was detected in tissues from seven of eight assessable patients treated with intravesical instillation of SCH 58500 but in none of three assessable patients treated with intratumoral injection of SCH 58500. Induction of RNA and protein expression of the p53 target gene p21/WAF1 was demonstrated in samples from patients treated with SCH 58500 instillation at higher dose levels. Distribution studies after intravesical instillation of SCH 58500 revealed both high transduction efficacy and vector penetration throughout the whole urothelium and into submucosal tumor cells. No dose-limiting toxicity was observed, and side effects were local and of transient nature. CONCLUSION: Intravesical instillation of SCH 58500 combined with a transduction-enhancing agent is safe, feasible, and biologically active in patients with bladder cancer. Studies to evaluate the clinical efficacy of this treatment in patients with localized high-risk bladder cancer are warranted.
Article
Introduction: Transurethral resection of bladder tumor (TURBT) is the gold standard initial diagnostic intervention for bladder cancer and provides diagnostic, therapeutic and prognostic benefit in non-muscle-invasive bladder cancer (NMIBC). However, TURBT alone is inadequate for optimal management of NMIBC, as patients will experience recurrence or progression depending on tumor characteristics. Adjuvant intravesical therapy with either immunotherapy or chemotherapy has been shown to reduce recurrence and/or progression in appropriately selected patients through immunostimulation or direct cell ablation. Areas covered: This review will discuss risk stratification of patients with NMIBC and role of intravesical therapies in reducing recurrence and progression of disease in these patients. A Medline search was performed to identify the best available evidence available from various systematic reviews, meta-analyses, and clinical trials on various immunotherapy and chemotherapy agents. In addition, the main aspects of drug pharmacology (mechanism of action, dosing and administration) and side effects will be reviewed. Expert opinion: The selection of the appropriate intravesical agent for NMIBC is complex and is dependent on risk stratification and intravesical agent toxicity. Intravesical induction and maintenance immunotherapy with Bacillus Calmette-Guerin (BCG) is the preferred and most effective agent for patients with high-risk NMIBC (carcinoma in situ and high-grade disease) and reduces both recurrence and progression.
Article
IL-15 has a pivotal role in life and death of natural killer (NK) and CD8 memory T cells. IL-15 signals through a heterotrimeric receptor involving the common gamma chain (γc) shared with IL-2, IL-4, IL-7, IL-9, and IL-21, IL-2/IL-15 receptor β (IL-15Rβ) shared with IL-2 and a private IL-15Rα subunit. IFN- or CD40 ligand-stimulated dendritic cells coordinately express IL-15 and IL-15Rα. Cell surface IL-15Rα presents IL-15 in trans to cells that express IL-2/IL-15Rβ and γc. IL-15 is being used to treat patients with metastatic malignancy. However, IL-15 is an inflammatory cytokine involved in immunological memory including that to self, thereby playing a role in autoimmune diseases. These insights provide the scientific basis for clinical strategies directed toward diminishing IL-15 action. Dysregulated IL-15 expression was demonstrated in patients with rheumatoid arthritis, inflammatory bowel disease, psoriasis, celiac disease, and alopecia areata. The monoclonal antibody Hu-Mik-β-1 targets the cytokine receptor subunit IL-2/IL-15Rβ (CD122), blocks IL-15 transpresentation, and is being used in clinical trials in patients with autoimmune diseases. In parallel, clinical trials have been initiated involving the Jak2/3 (Janus kinase-2/3) inhibitor tofacitinib and Jak1/2 inhibitor ruxolitinib to block IL-15 signaling.
Article
The widely used viral gene therapy vectors include retrovirus, lentivirus, adenovirus and adeno-associated virus, which have proven to be clinically valuable in numerous trials. These vectors, however, possess respective drawbacks and may raise safety concerns [1] although recent clinical trials have shown encouraging results [2]. Moreover, the production of these vectors has posed challenges in large scale production. In contrast to the commonly employed viral vectors, baculovirus is an insect virus and has been extensively employed for recombinant protein production by infecting its natural host insect cells since 1980s. However, two groups in the mid-1990s uncovered that baculovirus can enter mammalian cells and mediate transgene expression as long as the transgene is driven by a promoter active in mammalian cells [3,4]. Since then, following studies have demonstrated that baculovirus can efficiently transduce cells originating from human, rodent, porcine, bovine, rabbit, fish and avian species [5]. Baculovirus also transduces dividing and nondividing cells as well as primary cells including mesenchymal stem cells [6] and embryonic stem cells [7]. In mammalian cells, baculovirus neither replicates nor imposes apparent cytotoxicity, hence minimizing the possible side effects. Also baculovirus is non-pathogenic, hence the handling and production can be carried out in biosafety level 1 laboratories. Baculovirus is easy to construct and produced to high titers and can accommodate a single large insert or multiple genes encompassing the regulatory elements [5,8,9]. These findings and attributes have fueled the interests to explore baculovirus as a vector for gene therapy and a plethora of emerging applications. In this special issue, 3 papers review the development of baculovirus vectors for in vitro applications including cell-based assay development for drug screening [10], display of eucaryotic proteins [11] and recombinant protein production [12]. Baculovirus has been used for in vivo gene delivery, hence its in vivo applications and monitoring are crucial and reviewed in this issue [13]. Inspired by the accumulation of more fundamental knowledge and discovery of new permissive cell types, baculovirus has also been exploited in emerging fields such as cancer therapy [14], vaccine development [15] and tissue regeneration [16], which are reviewed by Madhan et al., Wang and Balasundaram, and Lin et al., respectively. Due to the mounting interests and applications, whether baculovirus is truly safe and what immune responses are provoked after baculovirus transduction of cells or administration into animals have emerged as an important issue, which is reviewed by Abe and Matsuura [17]. Finally, the translation of baculovirus technology from bench to bedside hinges critically on whether the baculoviral vectors can be produced and purified in large scale at reasonable cost, therefore, the bioprocessing of baculoviral vectors is reviewed by Aucoin et al. [18]. Compared to other well-known viral vectors, the development of baculoviral vector for gene delivery is still in its infancy. To date baculovirus-mediated gene therapy remains unknown to a large population of scientific community and has yet to advance to clinical trials. Nevertheless, the potential of baculovirus has attracted a rapidly increasing number of research groups to enter this field, with research topics covering the fundamental virology studies, viral vector development, new applications, to downstream production and purification. Although more needs to be done to elucidate and ameliorate the safety profile and efficacy, it is anticipated that this technology will yield dividends in coming years.