Sunteți pe pagina 1din 18

Advanced Techniques Ahmed, Adv Tech Biol Med 2013, 1:2

http://dx.doi.org/10.4172/atbm.1000107

in
Biology & Medicine
Research
Review ArticleAr ticle Open
OpenAccess
Access

0ncolytic Viruses as Therapeutic Agents for Prostate Cancer


Maryam Ahmed*
Department of Biology, Appalachian State University, USA

Abstract
Prostate cancer remains the leading cause of cancer-related morbidity and mortality for men in the Western
world. Conventional anti-cancer therapies like chemotherapy, irradiation, and hormone ablation often slow tumor
growth but do not engender long term benefits on patient survival. These therapies are limited by the fact that
tumor re-growth and spread to distal sites usually occurs following the conclusion of treatment. Therefore, there is
an increasing demand for the development of alternative therapeutic regiments. The use of oncolytic viruses for
the treatment of prostate cancer is an attractive option due to the natural ability of viruses to target and kill cancer
cells. Furthermore, oncolytic viruses may be genetically manipulated to transfer exogenous genes into cancer cells
in order to provide new generations of biological controls. This brief review highlights the potential of select
oncolytic viruses as promising modalities for prostate cancer treatments and presents the advantages and
practicalities of such viruses as therapeutic agents.

Keywords: Oncolytic viruses; Prostate cancer; Cancer therapies; One of the most widely studied oncolytic viruses is adenovirus.
Adenovirus; Herpes simplex virus; Vaccinia virus; Newcastle disease This virus was initially isolated in the early 1950s from adenoid-
virus; Vesicular stomatitis virus infected

Oncolytic Viruses
Viruses are commonly thought of as opportunistic organisms that
hijack cellular machinery with the ultimate goal of replicating and
causing destruction in the host. However, viruses are increasingly
being explored as therapeutic agents for a variety of disorders and
diseases, including cancers, due to their natural ability to spread to
various cell types. Oncolytic viruses are replication-competent viruses
that have the ability to selectively target cancerous growths, either
naturally through adaptation or through genetic manipulation. The
field of oncolytic virus development has spanned approximately
twenty years, with numerous viruses currently in clinical trials and in
various stages of development as anti-cancer agents. Although
oncolytic viruses exhibit diversity in their biologics and host range,
oncolytic viruses share common features of selective replication in
tumors, effective lysis of tumor cells, either directly or through
activation of anti-tumor immunity, and dispersion within tumor
masses. Strategies employed by viruses to target cancer cells include
exploiting the defective antiviral responses in cancer cells, the use of
cancer-specific surface markers as viral entry receptors, as well as
the use of cancer-specific promoters to activate viral gene products.
Many viruses that are currently being developed as agents against
prostate cancers have taken advantage of these strategies.
Depending on the disease state, the current treatments for prostate
cancer include radiotherapy, surgery, and hormone-deprivation
therapy [1]. Prostate cancer is particularly well-suited for oncolytic
therapies due to the fact that prostate removal or ablation is not life-
threatening. Furthermore, the major cause of death from prostate
cancers results from metastatic spread. Therefore, the natural ability
of viruses to spread to distal sites and seek out susceptible cancerous
tissues makes them attractive therapeutic agents for this disease. The
response to oncolytic therapies may also be monitored by serum
prostate-specific antigen (PSA) levels. This review will focus on
select oncolytic viruses and the advances that have been made in
developing them as therapeutic agents for the treatment of prostate
cancer.

Adenovirus
Adv Tech Biol Med
ISSN: ATBM, an open access journal Volume 1 • Issue 2 • 1000107
cell cultures, thus leading to the name adenovirus. It is recognized
as the etiological agent for diverse syndromes due to the presence of
approximately 50 serotypes. Not only does adenovirus possess
inherent oncolytic activity, it is widely being developed as a vaccine
and gene therapy agent.
Adenoviruses are attractive therapeutic vectors due to their wide
host range and the ease by which they may be manipulated
genetically [2]. Furthermore, a decade worth of clinical trials have
tested the safety and efficacy of various adenoviral vectors, thus
providing a framework from which to engineer novel agents. As an
oncolytic virus, much research has revealed that unmodified
adenovirus is insufficient to effectively treat neoplastic tissue due to
a variety of factors, including clearance from the immune system,
hepatic virus sequestration and down regulation of the coxsackie-
adenovirus receptor (CAR) in tumor cells [3-5], thus leading to the
engineering of second- and third- generation viruses with greater
therapeutic efficacy.
The first oncolytic adenovirus used to treat human cancers is
ONYX-15, containing a deletion of the viral EIB 55 KD gene. The lack
of EIB 55 KD expression restricts ONYX-15 replication and killing in
cells infected in the G1 phase of the cell cycle [6-8]. This poses a
limitation to cancer therapies because a significant number of cells
within tumors exist in the G1 phase. Because of this, ONYX-15
therapies have been more successful when combined with
chemotherapy or radiation therapies [7,9]. Additional adenoviruses
with mutations in the E1A gene product have been generated. E1A
stimulates S phase entry and serves to transactivate both cellular and
viral genes essential for a productive viral infection [10]. For
targeting prostate cancers, investigators have taken advantage of
prostate specific promoters and inserted them upstream of the EIA
gene of adenovirus type 5 (Ad5), thus restricting viral replication to
prostate cancer cells. An example of such a virus is

*Corresponding author: Maryam Ahmed, Department of Biology Appalachian


State University, 572 Rivers Street, Boone, NC 28607, USA, Tel: 828-262-2677;
E-mail: ahmedm@appstate.edu

Received April 16, 2013; Accepted May 24, 2013; Published May 27, 2013

Citation: Ahmed M (2013) Oncolytic Viruses as Therapeutic Agents for Prostate


Cancer. Adv Tech Biol Med 1: 107. doi:10.4172/atbm.1000107

Copyright: © 2013 Ahmed M. This is an open-access article distributed under the


terms of the Creative Commons Attribution License, which permits unrestricted
use, distribution, and reproduction in any medium, provided the original author
and source are credited.

Adv Tech Biol Med


ISSN: ATBM, an open access journal Volume 1 • Issue 2 • 1000107
Citation: Ahmed M (2013) Oncolytic Viruses as Therapeutic Agents for Prostate Cancer. Adv Tech Biol Med 1: 107. doi:10.4172/atbm.1000107

Page 2 of 7
Citation: Ahmed M (2013) Oncolytic Viruses as Therapeutic Agents for Prostate Cancer. Adv Tech Biol Med 1: 107. doi:10.4172/atbm.1000107
CN706 which was created by engineered to express other studies highlight the include immune and suicide
inserting the prostate specific sTGFβRIIFc, a protein which success of combination gene therapies. Adenoviruses
enhancer (PSE) derived from directly targets and inhibits the approaches in cancer have been developed to express
the 5’ end of the PSA gene TGF-β pathway. TGF-β has treatments. cytokines, chemokines, tumor-
into Ad5 [11]. In treatments been shown to play an associated antigens or other
Further therapeutic
with CN706, virus replication important role in the control of immunomodulatory factors. For
regiments employed for
correlates with the level of bone metastases [21,22] and example, adenoviruses armed
prostate cancers
PSA expression within given high levels of this factor in the with immune-therapeutic genes
prostate cancer cells. Studies blood circulation are poor such as IL-12 and IL-24 have
show that this virus is effective prognostic markers of prostate shown some efficacy in
at restricting growth of PSA- cancer [23,24]. This virus preclinical studies for the
producing LNCaP prostate induces significant reduction treatment of prostate cancers
tumors in cell culture and of tumor burden, osteoclast [16,33]. Immune cells including
animal model studies. number and bone destruction macrophages have also been
Numerous studies have also in a bone metastasis mouse utilized to deliver adenoviruses
shown efficacy by targeting model [19], thus displaying its to hypoxic areas of prostate
prostate cell surface markers potential as a therapeutic for tumors [31]. In terms of suicide
that are shown to be up- prostate cancer metastases. gene therapy, the two most
regulated during widely used prodrug therapies
Combination approaches
tumorigenesis. Prime candidates for prostate cancers include
have also been explored as HSV thymidine kinase (HSV-
include prostate-specific
membrane antigen (PMSA), means to promote adenovirus tk) together with ganciclovirir
whose increased expression replication and killing at tumor (GCV) or acyclovir, cytosine
correlates with aggressive sites. Numerous studies have deaminase (CD) and
tumors, prostate stem cell reported synergistic interaction 5-fluorocytosine (5-FC)
antigen (PSCA), the urokinase- of adenovirus with cytotoxic [34,35]. Each of these
type plasminogen activator drugs such as cisplatin, therapeutic regiments
receptor (uPAR), which is gemcitabine, docetaxel and represents a targeted approach
involved in tumor angiogenesis mitoxantrone for the treatment for prostate cancers that have
[12-15], and differential display of prostate cancers [25-27]. acquired resistance to
code 3 (DD3 or DD3(PCA)) While exact mechanisms are conventional treatments.
[16]. ONYX15 and the poorly understood, studies have
prostate-specific adenoviruses implicated the early viral EIA H
illustrate the two main gene in chemosensitization e
strategies employed to restrict [28]. By screening a panel of r
oncolytic adenovirus replication replicating mutants with EIA p
to tumor tissues: deletions, Miranda et al. [27]
e
1) By inactivating viral genes showed that adenovirus-
whose functions can be mediated sensitization to
s
compensated in cancer cells, cytotoxic drugs is dependent on
and 2) By placing essential regulatory domains in the S
viral genes under control of EIA conserved region 1 i
cancer or tissue-specific domain, which may have m
promoters [17]. functions favoring viral p
In addition to the success amplification [29]. There is also
l
of engineered adenoviruses interest in enhancing the
effects of oncolytic e
utilizing targetable prostate
cancer specific receptors, virotherapies with dietary x
prostate cancers have been phytochemicals with known
targeted with viruses containing anti-cancer properties. Natural V
the promoter for human dietary compounds with low i
telomerase reverse transcriptase toxicity, such as curcumin,
r
(hTERT), the catalytic genistein and resveratrol, have
componenent of the telomerase shown efficacy at killing
u
ribonucleoprotein complex prostate cancer cells in s
found in cancer cells. An combination with an oncolytic
example of such a virus is adenoviral mutant both in T
OBP-301, which shows strong vitro and in vivo [30]. y
anticancer effects by inducing Additional therapies include
the lysis of human prostate
p
the delivery of oncolytic
cancer cells and also adenoviruses to cancer cells or
e
demonstrates antimetastatic tumor tissue following
effects by eradicating sensitization with irradiation, I
detectable contralateral LNCaP which is linked to DNA
tumors in vivo [18]. More Herpes simplex virus type I
damage recognition and repair (HSV-1) is a natural human
recently, Hu et al. [19,20] [31,32]. The results of these
developed an hTERT promoter- pathogen which has been
studies, as well as numerous studied as an oncolytic agent
containing adenovirus
Citation: Ahmed M (2013) Oncolytic Viruses as Therapeutic Agents for Prostate Cancer. Adv Tech Biol Med 1: 107. doi:10.4172/atbm.1000107
for over two decades. During human primates [45]. NV1020
this time, increased strides is a multimutant HSV-1 strain
have been made in that contains several genetic
developing HSV for the modifications including deletion
treatment of a variety of of the UL24 gene and one copy
different cancers. This progress of the γ34.5 gene [46]. This
is highlighted by the translation virus has also shown efficacy in
of at least six oncolytic HSV reducing prostate tumor growth
vectors to the clinic, some in vivo and significantly
having progressed to Phase decreasing serum PSA levels
II/III clinical trials [36]. A [47]. Additional attenuated,
benefit to oncolytic HSV-1 replication competent viruses
therapies is the availability of derived from first generation
anti-HSV specific drugs oncolytic HSV-1, such as
(acyclovir) that may be NV1023 and G47Δ, are being
administered upon detection of evaluated for their ability to
a life threatening infection.
promote greater antitumor
Early studies of oncolytic HSV
activity against prostate cancers
focused on developing safe
[43,48].
anti-cancer agents by deleting
the γ34.5 gene, which governs In addition to testing the
neuropathogenicity [37,38]. ability of oncolytic HSV-1
Further vectors were strains to induce tumor cell
developed by introducing killing, they have also been
mutations or deletions in used as a platform to deliver
specific genes to prevent transgenes of interest. The
reversions to wild-type strains. integration of membrane-
However, these changes led to fusion activity into these
limited success due to viruses has been shown to
attenuation of replication in promote anti-tumor effects in
susceptible tissues including prostate cancer cells [29,49].
prostate carcinoma cells [39], Furthermore, viruses have been
indicating that greater potency armed with agents commonly
was necessary to promote used for prostate cancer
oncolytic efficacy. Current
vaccinations such as prostatic
studies are focusing on
acid phosphatase (PAP) [50]
synergizing the effects of
immune modulators
oncolytic HSV with a variety of
agents.
Several oncolytic HSV-1
strains have shown promise at
treatment of prostate cancers.
G207 is one of the first
onoclytic HSV-1 strains taken
into clinical trials. This virus,
derived from strain F, contains
deletions in both copies of the
γ34.5 gene and has an
inactivated ICP6 gene, which
encodes a viral ribonucleotide
reductase function [40]. The
double mutations permit viral
replication within quiescent
tumor cells carrying specific
oncogene deletions but not in
normal cells [41]. G207 has
been shown to be effective at
killing human prostate cancer
cells in vitro, as well as in vivo
in both subcutaneous xenograft
and transgenic mouse models
[42-44]. Additionally, it
displayed no evidence of
clinical disease and virus spread
into other organs when injected
into the prostates of HSV-1
susceptible mice and non-
Citation: Ahmed M (2013) Oncolytic Viruses as Therapeutic Agents for Prostate Cancer. Adv Tech Biol Med 1: 107. doi:10.4172/atbm.1000107

Page 3 of 7
Citation: Ahmed M (2013) Oncolytic Viruses as Therapeutic Agents for Prostate Cancer. Adv Tech Biol Med 1: 107. doi:10.4172/atbm.1000107
such as IL-12 [51], as well as to express large genes. cause of cancer-related deaths. N
factors that enhance virus Furthermore, studies have e
In addition to GLV-1h68, a
replication such as Ing4 shown that it is able to enter recombinant vaccinia virus w
(inhibitor of growth 4) [52,53]. and replicate efficiently within expressing PSA (rV-PSA) was c
numerous cell types without constructed by inserting the
Studies have focused on a
enhancing virus replication at causing natural disease in PSA gene into the viral
humans [55]. However, to s
prostate cancer tissues as well genome of the Wyeth strain
as exploring combination promote safety, attenuated, of vaccinia. rV-PSA has t
approaches. Lee et al. [54] avirulent versions of vaccinia shown some success in Phase I l
have developed recombinant viruses, including those clinical trials as indicated by e
viruses whose expression is lacking replication capacity, limited toxicity and evidence of
regulated by the presence of have been utilized as delivery immunological activity in
vectors for gene therapies or as D
the prostate specific promoter patients with rising PSA levels
(AAR(2) PB) and the 5’UTR vaccine vectors for the i
after local therapy, and in
of rFGF-2, thus promoting expression of immunizing patients with metastatic
s
tumor specificity. G47Δ, a antigens. More recently, androgen- independent prostate e
multimutated, replication vaccinia virus has also gained cancer [60,61]. a
competent HSV-1 vector popularity as an anti-cancer s
derived from G207, was agent. Beginning in 2007,
engineered by creating an Zhang et al. [56] described the
e
additional deletion within the oncolytic potential of the
non-essential α47gene [54]. The attenuated recombinant V
combination of androgen vaccinia virus GLV-1h68 in i
ablation with G47D therapy breast tumors. Since then, the r
resulted in greater tumor oncolytic effect of this virus u
growth suppression than either has been demonstrated in
therapy alone in the TRAMP- s
numerous cancer models,
C2 subcutaneous model. These including in the treatment of Newcastle disease virus
are a few of the many lymph node metastases (NDV) is a negative-sense
examples of approaches to originating from prostate single-stranded virus that causes
enhance the oncolytic potential carcinoma cells [57]. deadly infection in various
of HSV-1 vectors, similar to species of birds but is non-
those outlined for adenoviruses. Current studies are pathogenic to humans and
interested in determining the domestic animals. NDV has
V mechanisms by which GLV- been applied for the treatment
a 1h68 promotes anti-cancer of human cancers since the
c activity. GLV-1h68 was early 1960s with studies on
c engineered by inserting three uterine carcinoma [62]. Since
expression cassettes into then, it has been reported to
i
different loci of the viral possess oncolytic activity
n genome. Further genomic against a range of cancer types
i analysis confirmed that these and various strains have been
a insertions reduced the tested in clinical trials in
virulence of this virus and different human cancers
V promoted cancer cell tropism including glioblastoma
[58]. Recent studies have multiforme and colorectal
i
attributed the ability of GLV- cancer [63-65]. Studies have
r shown that NDV exhibits
1h68 to effectively treat lymph
u node metastases of prostate inherent selectivity for a
s carcinoma cells to the elevated diverse group of tumors over
vascular permeability in normal cells due to defects in
Vaccinia virus is a large,
metastases leading to greater antiviral responses, such as the
enveloped virus belonging to
release of virus particles and type I interferon (IFN)
the poxvirus family. The study
spread to susceptible tissues response, in certain cancer cells
of vaccinia virus began with its [66,67]. However, it has also
popularity as the choice for [59]. Furthermore, the presence
of increased number of been proposed that tumor
smallpox vaccination and in its specificity may be dependent
role in the successful global immune cells and the
proliferation of cancer cells at upon tumor cell resistance to
eradication of smallpox by apoptosis [68]. These tumor-
1979. Since then, there has metastatic areas are thought
specific defects serve to enhance
been great interest in to provide favourable conditions
replication of NDV in cancer
developing vaccinia virus a for virus infection and
cells to promote virus-induced
vector for the expression of replication. Taken together,
cytotoxicity.
foreign genes. This virus is these data indicate that vaccinia
attractive as a delivery vehicle virus GLV-1h68 may be used for The mechanisms underlying
because of its ability to stably the preferential destruction of the antitumor activity of NDV
accept as much as 25 kb of metastatic prostate carcinoma have been investigated in
cells, which represent a major numerous studies. Multiple
foreign DNA, thus enabling it
Citation: Ahmed M (2013) Oncolytic Viruses as Therapeutic Agents for Prostate Cancer. Adv Tech Biol Med 1: 107. doi:10.4172/atbm.1000107
studies have revealed the role of determinant of virulence is the
apoptosis in cell death by NDV, cleavage site in the F protein,
including both the intrinsic and which becomes fusogenic only
extrinsic pathways. The exact upon proteolytic cleavage into
mechanisms of apoptotic death two disulfide- linked
are dependent on the strain of polypeptides by host cellular
NDV, the cell lines and the proteases [80]. In an attempt to
detection assays [69-74]. In improve antitumor efficacy,
addition to direct killing Shobana et al. [81] have
induced by the virus, NDV engineered the F protein
also stimulates robust innate cleavage site to target the serine
and adaptive immunity. Various protease, PSA, such that F
strains of NDV are capable of protein is cleavable exclusively
stimulating macrophage by PSA in prostate cancer
activity as indicated by the cells. This strategy enhanced
detection of macrophage pathogenicity of oncolytic NDV
enzymes such as iNOS,
in prostate cancer cells as a
lysozyme and acid phosphatase
result of restricted viral
as well as the production of
replication and fusogenicity
nitric oxide and TNF-α [75-77].
[82].
Natural killer (NK) cells have
also been shown to mediate V
cytotoxicity against multiple
e
tumor cell lines following
infection of peripheral blood s
mononuclear cells (PBMCs) i
with NDV strain 73-T, one of c
the most well-characterized u
oncolytic strain of NDV [78]. l
NDV as an oncolytic agent a
for the treatment of prostate r
cancer is currently in the early
stages of development. Studies
S
with NDV
73-T have demonstrated t
antitumor effects in prostate o
carcinoma (PC3) xenografts m
upon systemic administration a
[79]. Furthermore, significant t
inhibition of tumor growth i
(77-96%) was also observed in
t
epidermoid, colon, large cell
lung, breast and low passage i
colon carcinoma xenograft s
models. Although phase I
clinical trials using naturally V
attenuated NDV strains such as i
PV701 have been conducted,
r
they have not included patients
with prostate cancers.
u
Nevertheless, clinical trials s
have revealed that PV701 was Vesicular stomatitis virus
well tolerated by patients when (VSV), a negative-strand RNA
administered intravenously virus of the Rhabdovirus
[35]. Side effects, including flu- family, has been studied as an
like symptoms, localized anti-cancer agent
adverse effects at the tumor
site and infusion reactions,
were observed. However, there
was no toxicity from the
oncolytic virus treatment.
Infection with NDV is
dependent on two viral
glycoproteins;
hemagglutininneuraminidase
(HN) and fusion (F). A major
Citation: Ahmed M (2013) Oncolytic Viruses as Therapeutic Agents for Prostate Cancer. Adv Tech Biol Med 1: 107. doi:10.4172/atbm.1000107

Page 4 of 7
Citation: Ahmed M (2013) Oncolytic Viruses as Therapeutic Agents for Prostate Cancer. Adv Tech Biol Med 1: 107. doi:10.4172/atbm.1000107
for several years. VSV exhibits numerous properties of an effective PC3 cells, Nguyen et al. [92] pretreated prostate cancer cells with
oncolytic agent including its well-defined biology, ability to induce histone deacetylase inhibitors (HDIs) known to suppress the type I IFN
apoptosis in a wide array of cancer cells and the lack of preexisting response [93]. Using the HDIs, HDI-MS-275 and SAHA (Vorinostat),
immunity in humans [67,82-85]. Similar to Newcastle disease virus, which have shown promising anti-cancer results in preclinical or
it has been proposed that the susceptibility of tumors to VSV is due clinical trials, they were able to augment the oncolytic activity of VSV-
to development of defects in antiviral responses during tumorigenesis Δ51-GFP both in vitro and in vivo xenograft models. Another approach
[67,82,85-87]. While normal cells may be infected by VSV, they
involved engineering the recombinant (VSV)-MΔ51 virus to express
respond to the virus by enhancing the type I IFN response, leading
the cytosine deaminase/uracil phosphoribosyltransferase (CD::UPRT)
to the attenuation of virus replication. However, wt strains of VSV
suicide gene and 5-fluorocytosine (5FC) prodrug [93]. This virus had
have the ability to suppress the antiviral response, induce systemic
immunity and replicate in the central nervous system [86,88,89], thus an enhanced ability to kill PC3 cells as compared to viruses lacking
leading to safety concerns. Over the last decade, increasing strides the suicide gene. Furthermore, it was effective at killing additional
have been made in the understanding of the interaction between the tumor cell lines derived from the breast. These, and similar studies
virus, cancers, and the immune response. This has led to the with other oncolytic viruses demonstrate the concept that in order
development of a number of recombinant attenuated VSVs with the to promote oncolysis, synergistic combination approaches must be
goal of enhancing the oncolytic potential of the virus, either directly investigated. Immunocompetent transgenic mice have served as
or indirectly through stimulation of the immune response, while useful model systems for measuring the safety and efficacy of VSV
maintaining safety. VSV has been tested as a candidate oncolytic treatment of prostate tumors. Moussavi et al. [94] demonstrated that
virus for prostate cancer by several groups. Early studies with prostate an IFN- sensitive VSV (AV3 strain) expressing luciferase effectively
cancers tested the ability of a matrix (M) protein mutant of VSV spreads in tumor-bearing prostate-specific PTEN(-/-) mice to
(rM51R-M virus) to kill LNCaP and PC3 prostate cancer cells in cell selectively infect and kill prostate tumor cells while sparing normal
culture and xenograft model systems [86]. The M51R M protein cells in control mice [95]. In these studies the virus was injected at
mutation disrupts the ability of VSV to shut- off the host antiviral the prostate site, thus demonstrating the utility of this administration
response in infected cells [85,86,90]. Xenograft studies showed that route. More recently, this same group showed that AV3 effectively
rM51R-M virus exhibits enhanced selectivity for tumor over normal targets metastatic lesions arising in the transgenic adenocarcinoma of
cells as compared to wt VSV strains, as indicated by the ability of
the mouse prostate (TRAMP) model [95]. The TRAMP C2 cell line
the virus to effectively kill tumor cells with limited signs of disease
derived from TRAMP mice was also utilized to demonstrate the
[86]. However, the efficacy of the virus depends on the cell type.
enhanced oncolytic properties of a recombinant VSV encoding
LNCaP cells are extremely sensitive to the effects of the virus while
PC3 cells remain resistant to infection and killing by rM51R-M virus SV5-F able to induce syncytial formation [96]. The SV5-F
perhaps due to the constitutive expression of numerous antiviral gene recombinant virus was constructed by replacing VSV glycoprotein
products in this cell line [91] (Figure 1). Several M51 protein (G) with that of the SV5-F to generate rVSV-DeltaG-SV5-F. rVSV-
mutants of VSV have been used to explore combination approaches DeltaG-SV5-F virus replication was restricted to TRAMP-C2 tumors
for the treatment of a variety of cancers, including prostate cancers. where it showed enhanced apoptotic and cytotoxic effects relative to a
In an attempt to augment the ability of VSV-Δ51-GFP to kill VSV- control virus lacking SV5-F.
resistant

Antiviral
A. Normal Cell -Viral replication inhibited
response -Type I IFN response
- Surrounding cells protected

Intact antiviral response

Antiviral
response -Viral replication
-Cell killing
B. Cancer Cell - virus spread
(i.e. LNCaP)
Cancer-specific genetic
alterations in antiviral
pathways

Antiviral
response -Viral replication inhibited
-Type I IFN response
C. Cancer Cell - Surronding cells protected
(i.e. PC3)
Cancer-specific genetic
alterations but antiviral
pathways intact

Figure 1: Selective killing of cancer cells by oncolytic M protein mutant strains of VSV. Oncolytic M protein mutant VSVs act as selective anti-cancer agents due to
their inability to inhibit host gene expression in infected cells. As a result, infected cells produce type I IFN and other antiviral cytokines in response to virus infection.
A. Normal cells contain intact antiviral response pathways that are induced by M protein mutant VSV leading to the attenuation of viral replication and prevention of
spread to surrounding tissue. B. Some cancer cells acquire genetic defects in antiviral pathways that render them susceptible to the oncolytic activity of M protein
mutant viruses. C. Other cancers retain intact antiviral pathways that protect them from the oncolytic activity of VSV. The resistance of these cancer cells to VSV
may be due to the constitutive expression of antiviral factors or their ability to mount an antiviral response upon infection with VSV, similar to that observed in
normal cells.
Page 5 of 7
by standard chemotherapeutic
In order to direct VSV to enhancement of anti-tumor 8. Thomas MA, Broughton RS,
agents. Nat Med 3: 639-645. Goodrum FD, Ornelles DA
prostate cancer cells, immunity, various methods to (2009) E4orf1 limits the oncolytic
investigators have pseudotyped modify the immune response potential of the E1B-55K deletion
replication defective VSV have been tested, including the mutant adenovirus. J Virol 83:
lacking its glycoprotein viral delivery of 2406-2416.

(VSVΔG) with MV-F and MV- immunostimulatory molecules 9. Kumar S, Gao L, Yeagy B, Reid T
H displaying single-chain and the co-administration of (2008) Virus combinations and
chemotherapy for the treatment of
antibodies (scFv) specific for reagents to enhance immune human cancers. Curr Opin Mol
or prostate membrane-specific function. Additionally, the Ther 10: 371-379.
antigen (PSMA) [97]. Results multifaceted tumor 10. Whyte P, Ruley HE, Harlow E
indicated that VSV replication environment has been shown (1988) Two regions of the
was restricted to prostate to impact viral infection, adenovirus early region 1A
proteins are required for
cancer cells expressing the replication and spread within
transformation. J Virol 62: 257-
PSMA surface marker. In the tumor. Therefore, studies 265.
addition, upon engineering are also exploring means to
11. Rodriguez R, Schuur ER, Lim
VSV to express antibodies for overcome environmental HY, Henderson GA, Simons JW,
the epidermal growth factor restrictions imposed on et al. (1997) Prostate attenuated
receptor (EGFR) and folate oncolytic therapies. Each of replication competent adenovirus
(ARCA) CN706: a selective
receptor (FR), this group these approaches offers great cytotoxic for prostate-specific
confirmed that retargeted VSV promise, either alone, or in antigen-positive prostate cancer
only replicated in cells combination with established cells. Cancer Res 57: 2559-2563.
expressing the target receptor antitumor therapies such as 12. Kraaij R, van Rijswijk AL, Oomen
[98]. Therefore, taking radiation or chemotherapy. MH, Haisma HJ, Bangma CH
advantage of the presence of Therefore, together with results (2005) Prostate specific
membrane antigen (PSMA) is a
cancer-specific surface markers obtained from numerous tissue-specific target for
represents and effective strategy clinical trials, the future of adenoviral transduction of
to restrict the ability of the oncolytic therapies for prostate prostate cancer in vitro. Prostate
62: 253-259.
virus to kill cancer cells over cancers remains promising.
normal cells. 13. Li Y, Cozzi PJ (2007) Targeting
References uPA/uPAR in prostate cancer. Cancer
Treat
Conclusions 1. Gomella LG, Johannes J, Trabulsi
Rev 33: 521-527.
EJ (2009) Current prostate
Oncolytic viruses represent cancer treatments: effect on 14. Morgenroth A, Cartellieri M,
promising modalities for the quality of life. Urology 73: S28- Schmitz M, Günes S, Weigle B,
S35. et al. (2007) Targeting of tumor
treatment of prostate cancers
2. Bachtarzi H, Stevenson M, Fisher cells expressing the prostate stem
due their ability to seek out cell antigen (PSCA) using
K (2008) Cancer gene therapy
and infect tumors, replicate in with targeted adenoviruses. genetically engineered T-cells.
target cells and spread to Expert Opin Drug Deliv 5: 1231- Prostate 67: 1121-1131.
surrounding cancerous or 1240. 15. Romer J, Nielsen BS, Ploug M
metastatic tissues. Numerous 3. Anderson WF (1998) Human (2004) The urokinase receptor
gene therapy. Nature 392: 25-30. as a potential target in cancer
strategies have been employed therapy. Curr Pharm Des 10:
to enable oncolytic viruses to 4. Jongmans W, Tiemessen DM, 2359-2376.
selectively target, replicate and Oosterwijk E, Mulders PF (2003)
Correspondence re: Y. S. Haviv, 16. Fan JK, Wei N, Ding M, Gu JF,
kill cancer cells. Promising J. L. Blackwell, A. Kanerva, P. Liu XR, et al. (2010) Targeting
approaches include the Nagi, V. Krasnykh, I. Dmitriev, M. Gene-ViroTherapy for prostate
targeting of viruses to Wang, S. Naito, X. Lei, A. cancer by DD3-driven oncolytic
Hemminki, D. Carey, and D. T. virus-harboring interleukin-24
prostate tumor-specific surface Curiel, Adenoviral gene therapy gene. Int J Cancer 127: 707-717.
markers or the use of tumor- for renal cancer requires
17. Stanizzi MA, Hall SJ (2007)
specific promoters to restrict retargeting to alternative cellular
receptors. Cancer Res., 62: 4273- Clinical experience with gene
viral gene expression to prostate 4281, 2002. Cancer Res 63: therapy for the treatment of
tumors. In order to enhance 1994-1995. prostate cancer. Rev Urol 9 Suppl
1: S20-28.
killing of cancer cells, suicide 5. Thomas MA, Spencer JF, Toth K,
gene therapies such as the use Sagartz JE, Phillips NJ, et al. 18. Huang P, Watanabe M, Kaku H,
Kashiwakura Y, Chen J, et al.
of HSV thymidine kinase and (2008) Immunosuppression
enhances oncolytic adenovirus (2008) Direct and distant
gancyclovir have been replication and antitumor efficacy antitumor effects of a telomerase-
explored. In addition, the in the Syrian hamster model. Mol selective oncolytic adenoviral
delivery of tumor necrosis Ther 16: 1665-1673. agent, OBP-301, in a mouse
prostate cancer model. Cancer
factor-related apoptosis- 6. Goodrum FD, Ornelles DA (1998) Gene Ther 15:
inducing ligand (TRAIL) to p53 status does not determine 315-322.
outcome of
tumors has been shown to E1B 55-kilodalton mutant 19. Hu Z, Gupta J, Zhang Z,
enhance the release of progeny adenovirus lytic infection. J Gerseny H, Berg A, et al. (2012)
virions from infected cells in Virol 72: 9479-9490. Systemic delivery of oncolytic
adenoviruses targeting
order to promote spread of 7. Heise C, Sampson-Johannes A, transforming growth factor-β
virus to surrounding Williams A, McCormick F, Von inhibits established bone
Hoff DD et al. (1997) ONYX-015,
susceptible tissue. Because the an E1B gene-attenuated
metastasis in a prostate cancer
efficacy of oncolytic therapies mouse model. Hum Gene Ther
adenovirus, causes tumor-
23: 871-882.
is greatly dependent on the specific cytolysis and antitumoral
efficacy that can be augmented 20. Hu Z, Robbins JS, Pister A,
Zafar MB, Zhang ZW et al.
(2010) A modified hTERT
promoter-directed oncolytic
adenovirus replication with
concurrent inhibition of TGFbeta
signaling for breast cancer
therapy. Cancer Gene Ther
17: 235-243.

21. Juárez P, Guise TA (2011)


TGF-β in cancer and bone:
implications for treatment of
bone metastases. Bone 48: 23-
29.

22. Sato S, Futakuchi M, Ogawa K,


Asamoto M, Nakao K, et al.
(2008) Transforming growth
factor beta derived from bone
matrix promotes cell proliferation
of prostate cancer and osteoclast
activation-associated osteolysis in
the bone microenvironment.
Cancer Sci 99: 316-323.

23. Schroten C, Dits NF, Steyerberg


EW, Kranse R, van Leenders AG,
et al. (2012) The additional value
of TGFβ1 and IL-7 to predict the
course of prostate cancer
progression. Cancer Immunol
Immunother 61: 905-910.

24. Shariat SF, Shalev M,


Menesses-Diaz A, Kim IY,
Kattan MW, et al. (2001)
Preoperative plasma levels of
transforming growth factor beta(1)
(TGF-beta(1)) strongly predict
progression in patients
undergoing radical prostatectomy.
J Clin Oncol 19: 2856-2864.

25. Gjerset R, Haghighi A,


Lebedeva S, Mercola D
(2001) Gene therapy
approaches to sensitization of
human prostate carcinoma to
cisplatin by adenoviral expression
of p53 and by antisense jun
kinase oligonucleotide methods.
Methods Mol Biol 175: 495-520.

26. Halldén G (2009) Optimisation


of replication-selective oncolytic
adenoviral mutants in
combination with
chemotherapeutics. J BUON 14
Suppl 1: S61-67.

27. Miranda E, Maya Pineda H,


Öberg D, Cherubini G, Garate
Z, et al. (2012) Adenovirus-
mediated sensitization to the
cytotoxic drugs docetaxel and
mitoxantrone is dependent on
regulatory domains in the
E1ACR1 gene-region. PLoS One
7: e46617.
28. Cook JL, Miura TA, Iklé DN,
Lewis AM Jr, Routes JM (2003)
E1A oncogene- induced
sensitization of human tumor
cells to innate immune defenses
and
Page 6 of 7
chemotherapy-induced apoptosis Replication-competent herpes N, Danner RL, et al. (2007)
in vitro and in vivo. Cancer Res 38. Chou J, Kern ER, Whitley RJ, virus NV1020 as direct Eradication of solid human breast
63: 3435- Roizman B (1990) Mapping of treatment of pleural cancer in a tumors in nude mice with an
3443. herpes simplex virus-1 rat model. J Thorac Cardiovasc intravenously injected light-
neurovirulence to gamma 134.5, Surg 124: 123-129. emitting oncolytic vaccinia virus.
29. Fu X, Tao L, Jin A, Vile R, a gene nonessential for growth in Cancer Res 67: 10038-10046.
Brenner MK, et al. (2003) culture. Science 250: 1262-1266. 48. Cozzi PJ, Burke PB, Bhargav A,
Expression of a fusogenic Heston WD, Huryk B, et al. 58. Gentschev I, Donat U,
membrane glycoprotein by an 39. Thompson RL, Wagner EK, (2002) Oncolytic viral gene Hofmann E, Weibel S,
oncolytic herpes simplex virus Stevens JG (1983) Physical therapy for prostate cancer using Adelfinger M, et al. (2010)
potentiates the viral antitumor location of a herpes simplex virus two attenuated, replication- Regression of human prostate
effect. Mol Ther 7: 748-754. type-1 gene function(s) competent, genetically tumors and metastases in nude
specifically associated with a 10 engineered herpes simplex mice following treatment with the
30. Adam V, Ekblad M, Sweeney K, million- fold increase in HSV viruses. Prostate 53: 95- recombinant oncolytic vaccinia
Müller H, Busch KH, et al. (2012) neurovirulence. Virology 131: 1 virus GLV-1h68. J Biomed
Synergistic and Selective Cancer 180-192. 0 Biotechnol 2010: 489759.
Cell Killing Mediated by the 0
Oncolytic Adenoviral Mutant 40. Taneja S, MacGregor J, Markus . 59. Zhang Q, Liang C, Yu YA,
AdΔΔ and Dietary S, Ha S, Mohr I (2001) Chen N, Dandekar T, et al.
Phytochemicals in Prostate Enhanced antitumor efficacy of a 49. Todo T, Martuza RL, Rabkin SD, (2009) The highly attenuated
Cancer Models. Hum Gene Ther herpes simplex virus mutant Johnson PA (2001) Oncolytic oncolytic recombinant vaccinia
23: 1003-1015. isolated by genetic selection in herpes simplex virus vector with virus GLV-1h68: comparative
cancer cells. Proc Natl Acad Sci enhanced MHC class I genomic features and the
31. Muthana M, Rodrigues S, U S A 98: 8804-8808. presentation and tumor cell contribution of F14.5L
Chen YY, Welford A, Hughes killing. Proc Natl Acad Sci U S A inactivation. Mol Genet Genomics
R, et al. (2013) Macrophage 41. Mineta T, Rabkin SD, Yazaki T, 98: 6396-6401. 282: 417-435.
delivery of an oncolytic virus Hunter WD, Martuza RL (1995)
abolishes tumor regrowth and Attenuated multi-mutated herpes 50. Li QX, Liu G, Zhang X (2012) 60. Donat U, Weibel S, Hess M,
metastasis after chemotherapy or simplex virus-1 for the treatment Fusogenic oncolytic herpes Stritzker J, Härtl B, et al.
irradiation. Cancer Res 73: 490- of malignant gliomas. Nat Med 1: simplex viruses as a potent and (2012) Preferential colonization
495. 938-943. personalized cancer vaccine. of metastases by oncolytic
Curr Pharm Biotechnol 13: 1773- vaccinia virus strain GLV-1h68 in
32. Nokisalmi P, Rajecki M, Pesonen 42. Aghi M, Visted T, Depinho RA, 1 a human PC-3 prostate cancer
S, Escutenaire S, Soliymani R, et Chiocca EA (2008) Oncolytic 7 model in nude mice. PLoS One 7:
al. (2012) Radiation-induced herpes virus with defective ICP6 8 e45942.
upregulation of gene expression specifically replicates in quiescent 5
from adenoviral vectors mediated cells with homozygous genetic . 61. Eder JP, Kantoff PW, Roper K, Xu
by DNA damage repair and mutations in p16. Oncogene 27: GX, Bubley GJ, et al. (2000) A
4249-4254. 51. Castelo-Branco P, Passer BJ, phase I trial of a recombinant
regulation. Int J Radiat Oncol
Buhrman JS, Antoszczyk S, vaccinia virus expressing
Biol Phys 43. Oyama M, Ohigashi T, Hoshi M, Marinelli M, et al. (2010)
83: 376-384. prostate-specific antigen in
Murai M, Uyemura K, et al. Oncolytic herpes simplex virus advanced prostate cancer. Clin
33. Nokisalmi P, Rajecki M, Pesonen (2000) Oncolytic viral therapy for armed with xenogeneic Cancer Res 6: 1632-1638.
S, Escutenaire S, Soliymani R, et human prostate cancer by homologue of prostatic acid
al. (2012) Radiation-induced conditionally replicating herpes phosphatase enhances antitumor 62. Gulley J, Chen AP, Dahut W,
upregulation of gene expression simplex virus 1 vector G207. Jpn efficacy in prostate cancer. Gene Arlen PM, Bastian A et al. (2002).
from adenoviral vectors mediated J Cancer Res 91: 1339-1344. Ther 17: 805-810. Phase I study of a vaccine using
recombinant vaccinia virus
by DNA damage repair and 44. Varghese S, Rabkin SD, 52. Passer BJ, Cheema T, Wu S, Wu expressing PSA (rVPSA) in
regulation. Int J Radiat Oncol Nielsen GP, MacGarvey U, Liu CL, Rabkin SD, et al. (2013) patients with metastatic
Biol Phys R, et al. (2007) Systemic Combination of vinblastine and androgen-independent prostate
83: 376-384. therapy of spontaneous prostate oncolytic herpes simplex virus cancer. Prostate 53: 109-117.
34. Nasu Y, Bangma CH, Hull GW, cancer in transgenic mice with vector expressing IL-12 therapy
oncolytic herpes simplex viruses. increases antitumor and 63. CASSEL WA, GARRETT RE
Lee HM, Hu J, et al. (1999)
Cancer Res 67: 9371-9379. antiangiogenic effects in prostate (1965) NEWCASTLE DISEASE
Adenovirus- mediated interleukin-
cancer models. Cancer Gene VIRUS AS AN
12 gene therapy for prostate 45. Walker JR, McGeagh KG, Ther 20: 17-24. ANTINEOPLASTIC AGENT.
cancer: suppression of orthotopic Sundaresan P, Jorgensen TJ, Cancer 18: 863-868.
tumor growth and pre-established Rabkin SD, et al. (1999) Local 53. Conner J, Braidwood L (2012)
lung metastases in an orthotopic and systemic therapy of human Expression of inhibitor of growth 4 by 64. Freeman AI, Zakay-Rones Z,
model. Gene Ther 6: 338-349. prostate adenocarcinoma with HSV1716 Gomori JM, Linetsky E, Rasooly
the conditionally replicating improves oncolytic potency and L, et al. (2006) Phase I/II trial of
35. Anello R, Cohen S, Atkinson G, enhances efficacy. Cancer Gene
herpes simplex virus vector intravenous NDV-HUJ oncolytic
Hall SJ (2000) Adenovirus Ther 19: 499- virus in recurrent glioblastoma
mediated cytosine deaminase G207. Hum Gene Ther
10: 2237-2243. 5 multiforme. Mol Ther 13: 221-228.
gene transduction and 5- 0
fluorocytosine therapy sensitizes 46. Varghese S, Newsome JT, 7 65. Hotte SJ, Lorence RM, Hirte
mouse prostate cancer cells to Rabkin SD, McGeagh K, . HW, Polawski SR, Bamat MK, et
irradiation. J Urol 164: 2173- Mahoney D, et al. (2001) al. (2007) An optimized clinical
2177. 54. Lee CY, Bu LX, DeBenedetti regimen for the oncolytic virus
Preclinical safety evaluation of
A, Williams BJ, Rennie PS, et PV701. Clin Cancer Res 13:
36. Freytag SO, Stricker H, Pegg J, G207, a replication-competent
al. (2010) Transcriptional and 9
Paielli D, Pradhan DG et al. herpes simplex virus type 1,
translational dual-regulated 7
(2003). Phase I study of inoculated intraprostatically in
oncolytic herpes simplex virus 7
replication-competent mice and nonhuman primates.
type 1 for targeting prostate -
adenovirus-mediated double- Hum Gene Ther 12: 999-1010.
tumors. Mol Ther 18: 929-935. 9
suicide gene therapy in 47. Ebright MI, Zager JS, Malhotra 8
combination with conventional- 55. Fukuhara H, Martuza RL, Rabkin 5
S, Delman KA, Weigel TL, et al.
dose three-dimensional SD, Ito Y, Todo T (2005) .
(2002)
conformal radiation therapy for Oncolytic herpes simplex virus
the treatment of newly diagnosed, vector g47delta in combination 66. Schulze T, Kemmner W, Weitz J,
intermediate- to high- risk with androgen ablation for the Wernecke KD, Schirrmacher V et al.
treatment of human prostate (2009).
prostate cancer. Cancer Res 63:
adenocarcinoma. Clin Cancer Efficiency of adjuvant active
7497-7506.
Res 11: 7886-7890. specific immunization with
37. Kanai R, Wakimoto H, Cheema T, Newcastle disease virus modified
Rabkin SD (2010) Oncolytic 56. Shen Y, Nemunaitis J (2005) tumor cells in colorectal cancer
herpes simplex virus vectors and Fighting cancer with vaccinia patients following resection of
chemotherapy: are combinatorial virus: teaching new tricks to an liver metastases: results of a
strategies more effective for old dog. Mol Ther 11: 180-195. prospective randomized trial.
cancer? Future Oncol 6: 619-634. Cancer Immunol Immunother 58:
57. Zhang Q, Yu YA, Wang E, Chen
61-69.

67. Fiola C, Peeters B, Fournier P,


Arnold A, Bucur M, et al. (2006) Tumor
selective
Page 7 of 7
replication of Newcastle disease 79. Zorn U, Dallmann I, Grosse J, 88. Carey BL, Ahmed M, Puckett S, 98. Ayala-Breton C, Barber GN,
virus: association with defects of Kirchner H, Poliwoda H, et al. Lyles DS (2008) Early Steps of Russell SJ, Peng KW (2012)
tumor cells in antiviral defence. (1994) Induction of cytokines and the Virus Replication Cycle Are Retargeting vesicular stomatitis
Int J Cancer 119: 328-338. cytotoxicity against tumor cells by Inhibited in Prostate Cancer Cells virus using measles virus
Newcastle disease virus. Cancer Resistant to Oncolytic Vesicular envelope glycoproteins. Hum
68. Stojdl DF, Lichty B, Knowles S, Biother 9: 225-235. Stomatitis Virus. J Virol 82: Gene Ther
Marius R, Atkins H, et al. (2000) 12104-12115. 23: 484-491.
Exploiting tumor-specific defects 80. Phuangsab A, Lorence RM,
in the interferon pathway with a Reichard KW, Peeples ME, 89. Huneycutt BS, Bi Z, Aoki CJ,
previously unknown oncolytic Walter RJ (2001) Newcastle Reiss CS (1993) Central
virus. Nat Med 6: 821-825. disease virus therapy of human neuropathogenesis of
tumor xenografts: antitumor vesicular stomatitis virus infection
69. Mansour M, Palese P, Zamarin effects of local or systemic of immunodeficient mice. J Virol
D (2011) Oncolytic specificity of administration. Cancer Lett 172: 67: 6698-
Newcastle 27-36. 6706.
disease virus is mediated by
selectivity for apoptosis-resistant 81. Shobana R, Samal SK, 90. Huneycutt BS, Plakhov IV,
cells. J Virol 85: Elankumaran S (2013) Shusterman Z, Bartido SM,
6015-6023. Prostate-specific antigen- Huang A, et al. (1994)
retargeted recombinant newcastle Distribution of vesicular stomatitis
70. Ali R, Alabsi AM, Ali AM, Ideris A, virus proteins in the brains of
disease virus for prostate cancer
Omar AR, et al. (2011) Cytolytic BALB/c mice following intranasal
virotherapy. J Virol 87: 3792-
effects and apoptosis induction of inoculation: an
3800.
Newcastle disease virus strain immunohistochemical analysis.
AF2240 on anaplastic 82. Sergel-Germano T, McQuain C, Brain Res
astrocytoma brain tumor cell line. Submit your next
Morrison T (1994) Mutations in 635: 81-95.
Neurochem Res 36: 2051-2062. the fusion peptide and heptad manuscript and get
repeat regions of the Newcastle 91. Ahmed M, McKenzie MO, Puckett
71. Fábián Z, Csatary CM, advantages of OMICS
disease virus fusion protein block S, Hojnacki M, Poliquin L, et al.
Szeberényi J, Csatary LK
fusion. J Virol 68: 7654-7658. (2003) Ability of the matrix protein Group submissions
(2007) p53-independent of vesicular stomatitis virus to
endoplasmic reticulum stress- Unique features:
83. Balachandran S, Porosnicu M, suppress beta interferon gene
mediated cytotoxicity of a Barber GN (2001) Oncolytic expression is genetically • User friendly/feasible website-
Newcastle disease virus strain in activity of vesicular stomatitis correlated with the inhibition of translation of your paper to 50
tumor cell lines. J Virol 81: 2817- virus is effective against tumors host RNA and protein synthesis. J world’s leading languages
2830. exhibiting aberrant p53, Ras, or • Audio Version of published paper
Virol 77: 4646-4657.
myc function and involves the • Digital articles to share and
72. Molouki A, Hsu YT, Jahanshiri F, 92. Nguyen TL, Abdelbary H, explore
induction of apoptosis. J Virol 75:
Rosli R, Yusoff K (2010) Arguello M, Breitbach C, Special features:
3474-3479.
Newcastle disease virus infection Leveille S et al. (2008) Chemical
promotes Bax redistribution to • 250 Open Access Journals
84. Kopecky SA, Willingham MC, targeting of the innate antiviral • 20,000 editorial team
mitochondria and cell death in Lyles DS (2001) Matrix protein response by histone deacetylase • 21 days rapid review process
HeLa cells. Intervirology 53: 87- and another viral component inhibitors renders refractory
eview and publication
• Quality and quick editorial, r
94. contribute to induction of cancers sensitive processing
to viral • Indexing at PubMed (partial),
apoptosis in cells infected with oncolysis. Proceedings of the us and Google Scholar etc
73. Ravindra PV, Tiwari AK, Ratta B,
vesicular stomatitis virus. J Virol National Academy of Sciences • Sharing Option: Social
Bais MV, Chaturvedi U, et al. Networking Enabled
75: 12169-12181. of the United States of America
(2009) Time course of Newcastle • Authors, Reviewers and Editors
disease virus-induced apoptotic 105: rewarded with online Scientific
85. Koyama AH (1995) Induction of
pathways. Virus Res 144: 14981-14986. Credits
apoptotic DNA fragmentation by
350-354. • Better discount for your
the infection of vesicular 93. Cary ZD, Willingham MC, subsequent articles
stomatitis virus. Virus Res 37: Lyles DS (2011) Oncolytic
74. Ravindra PV, Tiwari AK, Ratta Submit your manuscript at:
285-290. vesicular stomatitis virus induces
B, Chaturvedi U, Palia SK, et http://www.omicsonline.org/submissio
al. (2009) Newcastle disease apoptosis in U87 glioblastoma n/
86. Stojdl DF, Lichty BD, tenOever
virus-induced cytopathic effect in cells by a type II death receptor
BR, Paterson JM, Power AT, et al.
infected cells is caused by mechanism and induces cell
(2003) VSV strains with defects in
apoptosis. Virus Res 141: 13-20. death and tumor clearance in
their ability to shutdown innate
vivo. J Virol 85:
immunity are potent systemic
75. Ravindra PV, Tiwari AK, Ratta B, 5708-5717.
anti-cancer agents. Cancer Cell
Chaturvedi U, Palia SK et al.
4: 263-275. 94. Moussavi M, Fazli L, Tearle H,
(2008) Induction of apoptosis in
Vero cells by Newcastle disease Guo Y, Cox M, et al. (2010)
87. Ahmed M, Cramer SD, Lyles DS
virus requires viral replication, Oncolysis of prostate cancers
(2004) Sensitivity of prostate
denovo protein synthesis and induced by vesicular stomatitis
tumors to wild type and M protein
caspase activation. Virus virus in PTEN knockout mice.
mutant vesicular stomatitis
research 133:285-290. Cancer Res 70: 1367-1376.
viruses. Virology 330: 34-49.
76. Hrabák A, Csuka I, Bajor T, 95. Leveille S, Samuel S, Goulet
Csatáry LK (2006) The cytotoxic ML, Hiscott J (2011) Enhancing
Citation: Ahmed M (2013) VSV oncolytic activity with an
anti-tumor effect of MTH-68/H, a Oncolytic Viruses as Therapeutic
live attenuated Newcastle improved cytosine deaminase
Agents for Prostate
disease virus is mediated by the suicide gene strategy. Cancer
Cancer. Adv Tech Biol Med 1: 107.
induction of nitric oxide synthesis Gene Ther 18: 435-443.
doi:10.4172/atbm.1000107
in rat peritoneal macrophages in 96. Moussavi M, Tearle H, Fazli L,
vitro. Cancer Lett 231: 279-289. Bell JC, Jia W, et al. (2013)
77. Lorence RM, Rood PA, Kelley Targeting and killing of metastatic
KW (1988) Newcastle disease cells in the transgenic
virus as an antineoplastic agent: adenocarcinoma of mouse
induction of tumor necrosis prostate model with vesicular
factor-alpha and augmentation of stomatitis virus. Mol Ther 21: 842-
its cytotoxicity. J Natl Cancer Inst 848.
80: 1305-1312. 97. Chang G, Xu S, Watanabe M,
78. Schirrmacher V, Bai L, Jayakar HR, Whitt MA, et al.
Umansky V, Yu L, Xing Y, et al. (2010) Enhanced oncolytic
(2000) Newcastle disease virus activity of vesicular stomatitis
activates macrophages for anti- virus encoding SV5-F protein
tumor activity. Int J Oncol 16: against prostate cancer. J Urol
363-373. 183: 1611-1618.

S-ar putea să vă placă și