Skip to main content
Oncoimmunology logoLink to Oncoimmunology
. 2013 Apr 16;2(6):e24612. doi: 10.4161/onci.24612

Trial watch

Oncolytic viruses for cancer therapy

Erika Vacchelli 1,2,3, Alexander Eggermont 2, Catherine Sautès-Fridman 4,5,6, Jérôme Galon 7,6,8,9, Laurence Zitvogel 2,10, Guido Kroemer 3,4,6,11,12,, Lorenzo Galluzzi 1,4,11,†,*
PMCID: PMC3716755  PMID: 23894720

Abstract

Oncolytic virotherapy is emerging as a promising approach for the treatment of several neoplasms. The term “oncolytic viruses” is generally employed to indicate naturally occurring or genetically engineered attenuated viral particles that cause the demise of malignant cells while sparing their non-transformed counterparts. From a conceptual standpoint, oncolytic viruses differ from so-called “oncotropic viruses” in that only the former are able to kill cancer cells, even though both display a preferential tropism for malignant tissues. Of note, such a specificity can originate at several different steps of the viral cycle, including the entry of virions (transductional specificity) as well as their intracellular survival and replication (post-transcriptional and transcriptional specificity). During the past two decades, a large array of replication-competent and replication-incompetent oncolytic viruses has been developed and engineered to express gene products that would specifically promote the death of infected (cancer) cells. However, contrarily to long-standing beliefs, the antineoplastic activity of oncolytic viruses is not a mere consequence of the cytopathic effect, i.e., the lethal outcome of an intense, productive viral infection, but rather involves the elicitation of an antitumor immune response. In line with this notion, oncolytic viruses genetically modified to drive the local production of immunostimulatory cytokines exert more robust therapeutic effects than their non-engineered counterparts. Moreover, the efficacy of oncolytic virotherapy is significantly improved by some extent of initial immunosuppression (facilitating viral replication and spread) followed by the administration of immunostimulatory molecules (boosting antitumor immune responses). In this Trial Watch, we will discuss the results of recent clinical trials that have evaluated/are evaluating the safety and antineoplastic potential of oncolytic virotherapy.

Keywords: GM-CSF, HSV, immunotherapy, JX594, reolysin, talimogene laherparepvec

Introduction

Oncolytic viruses

Using viruses against cancer is not a particularly new concept, as it was initially put forward at the beginning of the 20th century.1 Still, it was not until the 1950s that the antineoplastic potential of (naturally occurring) viruses begun to be tested in preclinical settings as well as in patients, most often with deceiving results in terms of either safety or efficacy.2-5 Throughout the following three decades, the clinical interest in viruses as antineoplastic agents declined, even though a few studies reported encouraging safety data and even anecdotic cases of tumor regression.5-7 The possibility of using viruses as highly specific tumor-targeting tools came back under the limelight only in the late 1990s, along with the establishment of modern genetic engineering technologies.8-10 Since then, the clinical interest in oncolytic (and oncotropic) viruses has never declined again.

The term “oncolytic viruses” is generally employed to indicate non-pathogenic viral particles that specifically infect cancer cells, and hence cause their demise, while leaving unaffected non-malignant tissues.11 From a strict conceptual standpoint, oncolytic viruses differ from their oncotropic counterparts in that while the latter only exhibit a specific tropism for malignant cells, the former (be they replication-competent or not) kill their hosts upon infection (via several mechanisms, see below).9 Thus, oncotropic viruses (e.g., baculovirus, canarypox virus and canine parvovirus) efficiently enter malignant cells (and hence can be used as vectors for anticancer gene therapy) but are unable to complete the viral cycle and hence fail to exert major cytotoxic effects.12-14 Conversely, bona fide oncolytic viruses selectively infect cancer cells and kill them, either as a mere consequence of viral replication (cytopathic effect, mediated by replication-competent viruses only) or following the expression of endogenous or exogenous cytotoxic gene products (suicide gene therapy, mediated by both replication-competent and replication-incompetent virions).9,11 A detailed description of all the viral species that have been tested for their oncolytic potential or genetically engineered in this sense—each of which is associated with specific advantages and drawbacks—exceed the scope of the present Trial Watch and can be found in refs. 9 and 11.

Tropism and selectivity of oncolytic viruses

The specific targeting of malignant cells is an absolute requirement for the clinical success of oncolytic virotherapy. In line with this notion, during the past two decades several distinct strategies have been pursued to improve the oncotropism of naturally occurring viruses (some of which exhibit an inherent preference for malignant cells), as well as to increase their oncoselective cytotoxic potential.9,11 The strategies that have been investigated to ameliorate the selectivity of oncolytic viruses include (1) the genetic manipulation of viral envelopes or capsids to provide viruses with a strict specificity for cells bearing a tumor-associated surface marker (transductional targeting);15,16 (2) the expression of one or more genes that are essential for the viral cycle under the control of promoters that functions only in transformed cells, including tissue-specific and hypoxia-responsive promoters (transcriptional targeting);17-20 (3) the insertion of essential viral genes downstream of internal ribosome entry sites (IRESs) that cannot be used as translational origins in specific tissues (translational targeting);21 (4) the use of so-called “destabilization domains,” rendering essential viral products inherently unstable unless a stabilizing chemical is administered (in a tissue-restricted manner) (post-translational targeting);22,23 (5) the deletion of one or a few virulence factors, resulting in attenuated viral strains that are able to replicate only in cells bearing cancer-associated alterations in specific signal transduction pathways, such as the hyperactivation of RAS or the inactivation of p53 (oncogenetic targeting);24-28 and (6) the insertion of tissue-specific microRNA-binding elements in the 3′-UTR of essential viral genes, allowing tissues that express such microRNAs to escape productive infection (microRNA targeting).29-32

Alongside, great efforts have been dedicated at rendering oncolytic viruses increasingly more toxic for cancer cells, well beyond the cytopathic effect normally mediated by replication-competent particles. Strategies that have been undertaken in this sense include the integration into the viral genome of sequences coding for: (1) enzymes that can transform a non-toxic pro-drug into a lethal cytotoxic chemical, such as the thymidine kinase (TK) of herpes simplex viruses (HSVs), converting ganciclovir into deoxyguanosine triphosphate;33,34 or cytosine deaminase, catalyzing the conversion of 5-fluorocytosine into 5-fluorouracil;35-37 (2) proteins that (at least potentially) exert tumor-restricted cytotoxic functions, such as wild-type p53 or death receptor ligands (e.g., FASL);38,39 and (3) short-hairpin RNAs targeting proteins that are specifically required for the survival of malignant cells, such as survivin.40 A detailed description of these approaches can be found in refs. 11 and 41. Of note, some viral particles are capable of inducing the demise of malignant (but not normal) cells upon uncoating even in the absence of viral gene expression.42 The precise molecular mechanisms underlying this phenomenon remain obscure, yet its efficacy appears to be linked to the number of virions actually entering target cells and being uncoated. Along similar lines, a long list of viral proteins exerts bona fide tumor-selective pro-apoptotic functions.43 For example, both apoptin, a 15 kDa protein encoded by the chicken anemia virus, and the polypeptide coded by the adenoviral E4 open reading frame have been shown to preferentially trigger the apoptotic demise of malignant, as opposed to non-transformed, cells.44-46

Immunogenic activity of oncolytic viruses

Contrarily to long-standing beliefs, the antineoplastic potential of oncolytic virotherapy is not a mere consequence of the cytopathic effect, but rather involves (at least in a majority of settings) the induction of a tumor-specific immune response.9,11,47,48 In line with this notion, oncolytic viruses genetically engineered to drive the production of various immunostimulatory factors have been shown to mediate more robust therapeutic effects than their non-engineered counterparts. Proof-of-principle evidence in support of this concept has been generated for oncolytic viruses that express membrane-bound co-stimulatory molecules, such as CD40 ligand (CD40L) and CD80,49-52 as well as multiple distinct immunostimulatory cytokines encompassing interleukin (IL)-2,53 IL-12,54-56 IL-1557-59 and granulocyte macrophage colony-stimulating factor (GM-CSF).55,60-62 In addition, the clinical efficacy of oncolytic virotherapy appears to be robustly ameliorated by some degree of initial immunosuppression (facilitating viral replication and spread) followed by the provision of immunostimulatory factor (boosting antitumor immunity).9,11

Obstacles against efficient oncolytic virotherapy

The administration of oncolytic viruses as a standalone therapeutic intervention has been shown to rarely induce the complete, long-term regression of established tumors in vivo, owing to several reasons.9,11 First, preclinical and clinical evidence indicates that oncolytic viruses often infect neoplastic lesions in a rather heterogeneous and incomplete fashion, irrespective of administration route and whether viruses are replication-competent or not.63-67 A prominent role in this context is played by tumor size68 as well as physicochemical barriers to infection, including the layers of connective tissue,69,70 the elevated interstitial pressure,71 the poorly permissive vasculature72 and the large areas of necrosis/calcification73,74 that generally characterize solid tumors. Second, oncolytic viruses often establish a “dynamic equilibrium” with tumors, that is, a state in which viral infection affects a spatially-restricted population of malignant cells, rather than the entire lesion, and is continuously counterbalanced by the proliferation of non-infected cancer cells.75,76 Of note, while in immunodeficient settings such an equilibrium may be relatively stable (and hence de facto impede tumor growth),77 at some stage the immune system is expected to eradicate the viral infection in immunocompetent animals, thereby tilting the tip of the balance in favor of malignant cells. Third, similar to what commonly observed in the course of chemo- and radiotherapy, malignant cells are prone to become resistant to oncolytic virotherapy over time,67,72,77-79 presumably linked to their intrinsic genomic instability and propensity to accumulate mutations.80,81 Interestingly, cancer cells that had been rendered resistant to the parvovirus H1 in vitro were found to be cross-protected against the cytotoxic effects of tumor necrosis factor α (TNFα),82-84 an observation with profound implications for the development of combination chemoimmunotherapeutic regimens involving oncolytic viruses. Fourth, a large fraction of the population has previously been exposed to the naturally occurring viruses that are commonly employed to generate therapeutic strains, de facto being fully protected against their infectious potential by high levels of neutralizing antibodies.85,86 Along similar lines, some viral particles, including HSV1- and murine leukemia virus-derived viruses, are particularly prone to inactivation by the complement system.87,88 Fifth, unless specific preventive measures are undertaken, oncolytic viruses administered i.v. are massively sequestered (most often upon opsonization) by the mononuclear phagocytic system (MPS) of the liver and spleen.89,90 On one hand, this significantly restricts the amount of infectious particles that are capable of reaching the tumor site, de facto compromising transduction efficiency and hence therapeutic effects.91 On the other hand, the accumulation of viral particles into the liver and the spleen may trigger the release of pro-inflammatory cytokines, potentially driving serious, dose-limiting toxicities.92,93 Sixth, although great progresses have already been made in this sense, the use of viruses remains intrinsically associated with some threats, in particular when cancer patients—who often exhibit some degree of immunosuppression—are concerned.94-96 In this respect, one problematic issue relates to the fact that all viruses are potentially immunogenic and hence can cause—at the therapeutic doses of 1012 particles—unwarranted side effects including transient hepatic inflammation (see above) and low-grade disseminated intravascular coagulation.97,98 In addition, great concerns have been raised by the case of three children who developed a leukemic syndrome following the reinfusion of retrovirally transduced T cells for the treatment of a severe monogenic immunodeficiency, most likely owing to the integration of the viral vector in the proximity of the LMO2 oncogene.99,100

Taken together, these observations highlight an urgent need for the development of ever more refined oncolytic viruses and the design of combinatorial strategies by which the obstacles described above would be—at least in part—circumvented. Several approaches have already been conceived to ameliorate the therapeutic potential of oncolytic virotherapy, including (among several others) (1) the use of coating polymers to shield viral particles from the neutralizing effects of circulating antibodies and the complement system as well as from sequestration by the MPS;101-103 (2) the pre-administration of molecules that selectively increase the permeability of tumor vessels (e.g., IL-2, TNFα, histamine, bradykinin analogs) or aggressive chemotherapy, to reduce interstitial pressure;104,105 (3) the intratumoral administration of antifibrotic agents, such as the FDA-approved antihypertensive drug losartan, or enzymes that degrade components of the extracellular matrix, such as hyaluronidase;106,107 (4) the development of oncolytic viruses that—in addition to cytotoxic factors—express extracellular matrix-degrading enzymes like hyaluronidase;108 and (5) the careful modulation of the immune system (for instance with cyclophosphamide, which at high doses exerts potent immunosuppressive effects)109-111 to avoid premature antiviral responses that would compromise viral spread (and hence therapeutic efficacy).112

The safety and preliminary antineoplastic effects of several naturally occurring and genetically modified viruses have been/are being tested in multiple distinct clinical trials (see below). As it stands, however, no oncolytic viruses are licensed by the U.S. FDA for use in cancer patients (source www.fda.gov). Conversely, gendicine, an oncotropic, recombinant adenovirus engineered to express wild-type p53, has been approved for the treatment of subjects affected by head and neck squamous cell carcinoma in China as early as in 2003.113,114 Along the lines of our monthly Trial Watch series,115-128 here we will briefly review the progress of recent clinical trials that have investigated/are investigating the antineoplastic potential of oncolytic viruses.

Clinical Development of Oncolytic Viruses

Literature

As mentioned above, the possibility of using viruses to specifically kill neoplastic cells begun to attract the attention of clinicians, and hence to be tested in patients, in the 1950s.1 In this context, hundreds of patients bearing a wide array of distinct neoplasms received highly impure viral preparations (including infected body fluids and so-called “oncolysates,” i.e., autologous malignant cells or established cancer cell lines killed by viruses ex vivo), via almost every conceivable route of administration.3,11,129,130 Most often, these attempts failed to drive any therapeutic response as viruses were rapidly inactivated by the immune system. Nevertheless, sporadic cases of tumor regression were reported, in particular among immunodeficient patients, who however were at increased risk of death owing to the spread of the viral infection to healthy tissues.3,11,129,130 Perhaps the most impressive results of this period were obtained by Asada and colleagues, who reported 37 tumor regressions among 90 terminal cancer patients treated with a non-attenuated strain of the mumps virus.131

In 1991, Martuza et al. were the first to demonstrate that a genetically manipulated variant of HSV lacking the TK-coding gene (and hence displaying attenuated virulence) could be successfully employed to treat glioblastoma in mice.132 This report de facto paved the way to modern virotherapy and drove an intense wave of preclinical and clinical investigation that has not yet come to an end. Since then indeed, dozens—if not hundreds—of distinct oncolytic and oncotropic viruses have been developed and tested in preclinical models. An exhaustive discussion of the preclinical results obtained during the last two decades with oncolytic viruses can be found in refs. 9, 11, 133 and 134. Alongside, several oncolytic viruses entered the clinics and their safety and preliminary antineoplastic potential begun to be investigated in patients affected by a wide variety of (mostly solid) tumors.11,135,136

Adenoviruses and HSVs represent by far the oncolytic viruses most extensively investigated (at both the preclinical and clinical level) and developed so far.137-139 Indeed, attenuated adenoviral strains, most often lacking the genes coding for E3 or the anti-apoptotic protein E1B55K,43 have already been used in patients affected by glioma,140 sarcoma,141 head and neck cancer (HNC),97,142-145 pancreatic cancer,146,147 colorectal carcinoma (CRC),147-149 prostate carcinoma,150-152 ovarian cancer,153,154 bladder carcinoma61 and multiple solid tumors.155,156 Along similar lines, the safety and therapeutic potential of attenuated HSVs, near to invariably lacking the gene coding for the main neurovirulence factor γ34.5,157 have been tested in subjects with glioma,158-162 breast carcinoma,163 HNC,164-166 melanoma,167-170 pancreatic carcinoma,171 CRC hepatic metastases172 and various solid malignancies.173 Most of the remaining clinical trials that have been completed so far were designed to evaluated the safety and the oncolytic profile of stains of Newcastle disease virus (in subjects affected by glioma and other solid tumors),174-176 parvovirus (in glioblastoma multiforme patients),177 reovirus (in individuals bearing glioma, melanoma and other solid tumors)178-181 and vaccinia virus (in hepatocellular carcinoma and melanoma patients).62,182-184 The majority of these studies were Phase I/II trials, most often reporting reassuring safety data and sporadic antineoplastic activity, even when oncolytic viruses were employed as standalone therapeutic interventions. In this setting, the most encouraging results have surely been recorded with talimogene laherparepvec (developed by Amgen, also known as OncoVex), an oncolytic γ34.5-deficient variant of HSV genetically manipulated to drive the expression of GM-CSF by infected cells.185 As a single agent administered i.t., talimogene laherparepvec induced the complete regression of both injected and distant lesions in 8 out of 50 metastatic melanoma patients.170 A similar efficacy has been observed with JX954, an oncolytic vaccinia virus engineered to express GM-CSF that was shown to induce objective responses in a consistent percentage of hepatocellular carcinoma patients.62,184 In addition, multiple studies have shown that combining oncolytic viruses with conventional radio- or chemotherapeutic is generally safe and improves (to some extent) the rates of clinical responses.141,143,145,146,149-152,166,174,179 In summary, accumulating clinical data strongly support the development of oncolytic virotherapy.

Recent, ongoing clinical trials

Nowadays (April 2013), official sources list no less than 52 recent (started after January 1, 2008), ongoing (not withdrawn, terminated or completed at the day of submission), clinical trials assessing the safety and antineoplastic potential of oncolytic viruses in cancer patients (Table 1). One third (17) of these studies are investigating the activity of a wild-type reovirus (serotype 3 Dearing, developed by Oncolytics Biotech under the name of Reolysin®)186 administered i.v. or (rarely) i.p., often in combination with conventional therapeutic regimens, to patients affected by multiple myeloma (NCT01533194), HNC (NCT00753038; NCT01166542), breast carcinoma (NCT01656538), melanoma (NCT00651157; NCT00984464), lung cancer (NCT00861627; NCT00998192; NCT01708993), pancreatic carcinoma (NCT00998322; NCT01280058), CRC (NCT01274624; NCT01622543), prostate cancer (NCT01619813), reproductive tract neoplasms (NCT00602277; NCT01199263) or pediatric solid tumors (NCT01240538). Eight clinical trials are assessing the safety and antineoplastic profile of JX594 (developed by Transgene as Pexa-Vec), administered i.v. most often as a standalone therapeutic intervention, in subjects bearing hepatocellular carcinoma (NCT01171651; NCT01387555; NCT01636284), CRC (NCT01380600, NCT01394939; NCT01469611) or chemorefractory solid tumors (NCT00625456; NCT01169584). The highly attenuated oncolytic vaccinia virus GL-ONC1 (developed by Genelux, also known as GLV-1h68)187 is being tested in combination with chemoradiotherapy for the treatment of HNC patients (NCT01584284), as a standalone therapeutic intervention administered intrapleurally in lung cancer patients (NCT01766739), and upon intravenous or intraperitoneal delivery in subjects affected by advanced solid tumors or peritoneal carcinomatosis, respectively (NCT00794131; NCT01443260).

Table 1. Recent clinical trials evaluating oncolytic virotherapy in cancer patients*.

Virus Indication(s) Phase Status Route Co-therapy Ref.
CG0070
Bladder carcinoma
II/III
Not yet recruiting
Intravesical
As single agent
NCT01438112
CGTG-102
Solid tumors
I
Recruiting
i.t. and i.v.
Combined with cyclophosphamide
NCT01598129
Not yet recruiting
i.t.
As single agent
NCT01437280
Coxsackievirus A21
Melanoma
II
Recruiting
i.t.
As single agent
NCT01227551
NCT01636882
DNX-2401
GBM
I/II
Recruiting
CED
As single agent
NCT01582516
Glioma
I
Recruiting
i.t.
As single agent
NCT00805376
GL-ONC1
HNC
I
Recruiting
i.v.
Combined with cisplatin and RT
NCT01584284
Lung cancer
I
Recruiting
Intrapleural
As single agent
NCT01766739
Peritoneal carcinomatosis
I/II
Recruiting
i.p.
As single agent
NCT01443260
Solid tumors
I
Recruiting
i.v.
As single agent
NCT00794131
HF10
HNC
Solid tumors
I
Recruiting
i.t.
As single agent
NCT01017185
HSV1716
Mesothelioma
I/II
Recruiting
Intrapleural
As single agent
NCT01721018
Non-CNS
solid tumors
I
Recruiting
i.t.
As single agent
NCT00931931
JX594
HCC
II
Recruiting
i.v.
As single agent
NCT01636284
n.a.
Combined with BSC
NCT01387555
Active not recruiting
i.v. and i.t.
Followed by sorafenib
NCT01171651
CRC
I
Active not recruiting
i.v.
As single agent
NCT01380600
Recruiting
i.v.
As single agent
NCT01469611
I/II
Recruiting
i.v. and i.t.
Combined with irinotecan
NCT01394939
Solid tumors
I
Active not recruiting
i.t.
As single agent
NCT01169584
i.v.
As single agent
NCT00625456
Measles virus
Mesothelioma
I
Recruiting
Intrapleural
As single agent
NCT01503177
NDV-HUJ
GBM
NB
Sarcoma
I/II
Not yet recruiting
i.v.
As single agent
NCT01174537
NTX-010
Lung cancer
II
Recruiting
i.v.
As single agent
NCT01017601
Neuroendocrine tumors
I
Recruiting
i.v.
Combined with cyclophosphamide
NCT01048892
ParvOryx
GBM
I/II
Recruiting
i.t. or i.v.
As single agent
NCT01301430
PVSRIPO
GBM
I
Recruiting
i.t.
As single agent
NCT01491893
Reolysin®
Breast carcinoma
II
Recruiting
n.a.
Combined with PTX
NCT01656538
CRC
I
Active not recruiting
i.v.
Combined with FOLFIRI regimen
NCT01274624
II
Recruiting
n.a.
Combined with BVC and FOLFOX regimen
NCT01622543
HNC
II
Active not recruiting
i.v.
Combined with
CBP and PTX
NCT00753038
III
Active not recruiting
i.v.
Combined with
CBP and PTX
NCT01166542
Lung cancer
II
Recruiting
i.v.
Combined wit
CBP and PTX
NCT00861627
NCT00998192
Combined with DCX or pemetrexed
NCT01708993
Melanoma
II
Active not recruiting
i.v.
As single agent
NCT00651157
Recruiting
i.v.
Combined with
CBP and PTX
NCT00984464
Multiple myeloma
I
Recruiting
i.v.
As single agent
NCT01533194
Pancreatic cancer
II
Recruiting
i.v.
Combined with
CBP and PTX
NCT01280058
Active not recruiting
i.v.
Combined with gemcitabine
NCT00998322
Pediatric solid tumors
I
Recruiting
i.v.
Combined with cyclophosphamide
NCT01240538
Prostate cancer
II
Recruiting
n.a.
Combined with DCX and prednisone
NCT01619813
Reproductive
tract tumors
I
Recruiting
i.v. + i.p.
As single agent
NCT00602277
II
Recruiting
i.v.
Combined with PTX
NCT01199263
Talimogene
laherparepvec
Melanoma
I/II
Recruiting
i.t.
Combined with ipilimumab
NCT01740297
III
Active not recruiting
i.t.
As single agent
NCT00769704
Enrolling by invitation
i.t.
As single agent
NCT01368276
Toca 511
Astrocytoma
GBM
Oligoastrocytoma
Oligodendroglioma
I
Recruiting
Resection cavity
As single agent
NCT01470794
I/II
Recruiting
i.t.
Combined with 5-FC
NCT01156584
VSV-IFN-β HCC I Recruiting i.t. As single agent NCT01628640

Abbreviations: 5-FC, 5-fluorocytosine; BSC, best supportive care; BVC, bevacizumab; CBP, carboplatin; CED, convection enhanced delivery; CNS, central nervous system tumor; CRC, colorectal cancer; DCX, docetaxel; FOLFIRI, folinic acid, 5-fluorouracil, irinotecan; FOLFOX, folinic acid, 5-flurouracil, oxaliplatin; GBM, glioblastoma multiforme; HCC, hepatocellular carcinoma; HNC, head and neck cancer; HSV, herpes simplex virus; IFN, interferon; n.a., not available; i.p., intra peritoneum; i.t., intra tumorem; i.v., intra venam; NB, neuroblastoma; NDV, Newcastle disease virus; PTX, paclitaxel; RT, radiotherapy; VSV, vesicular stomatitis virus. *Started after January 1, 2008, and not withdrawn, terminated or completed on the day of submission.

In addition, clinical trials are currently ongoing for investigating the safety and antineoplastic activity of: (1) talimogene laherparepvec,62 administered i.t.—as a standalone intervention or combined with systemic ipilimumab188,189—to melanoma patients (NCT00769704; NCT01368276; NCT01740297); (2) naturally occurring coxsackievirus A21 (developed by Viralytics as Cavatak™),190 administered i.t. as a single agent to melanoma patients (NCT01227551; NCT01636882); (3) CGTG-102 (an oncolytic adenovirus engineered to drive the local expression of GM-GSF, developed by Oncos),191 administered i.t. or i.v.—alone or in combination with metronomic cyclophosphamide—to patients affected by advanced solid tumors (NCT01437280; NCT01598129); (4) DNX-2401 (a genetically manipulated replication-competent adenovirus developed by DNAtrix),192,193 given i.t. as a standalone agent to glioblastoma and glioma patients (NCT00805376; NCT01582516); (5) HSV1716 (a γ34.5-deficient variant of HSV developed by Virttu as Seprehvir®),169 administered intrapleurally or intratumorally to individuals affected by mesothelioma or solid tumors (NCT01721018; NCT00931931); (6) Toca511® (a retrovirus engineered to express cytosine deaminase, developed by Tocagen), given alone or in combination with 5-fluorocytosine to subjects affected by astrocytoma, glioblastoma, oligoastrocytoma and oligodendrocytoma (NCT01156584; NCT01470794); (7) the Seneca Valley virus (a replication-competent oncolytic picornavirus also known as NTX-010),194 administered i.v. as a single agent or combined with metronomic cyclophosphamide to patients with lung cancer or neuroendocrine tumors, respectively (NCT01017601; NCT01048892); (8) PVSRIPO (an nonpathogenic recombinant poliovirus),195 given i.t. as a standalone intervention to glioblastoma patients (NCT01491893); (9) the attenuated measles virus commonly used as a prophylactic vaccine,75,196 given intrapleurally as a single therapeutic agent to mesothelioma patients (NCT01503177); (10) CG0070 (a conditionally replicating GM-CSF-armed oncolytic adenovirus),61,197 instilled intravesically as a single agent to subjects with bladder carcinoma (NCT01438112); (11) HF10 (a spontaneous attenuated variant of HSV1),198,199 administered i.t. as a standalone intervention to individuals affected by refractory HNC or other solid tumors (NCT01017185); (12) an attenuated lentogenic isolate of the Newcastle disease virus (known as NDV-HUJ),174,200 given i.v. as a single agent to glioblastoma, neuroblastoma and sarcoma patients (NCT01174537); (13) parvovirus H1 (a naturally occurring parvoviral variant developed by Oryx Verwaltungs as ParvOryx®),177,201 administered i.v. or i.t. as a standalone therapeutic intervention to glioblastoma patients (NCT01301430); and (14) a variant of the vesicular stomatitis virus engineered to drive the expression of interferon β (IFNβ),202,203 given i.t. as a single agent to subjects affected by hepatocellular carcinoma (NCT01628640). The large majority of these studies are Phase I/II clinical trials, with the notable exceptions of NCT01438112 (assessing the antineoplastic activity of CG0070 in bladder carcinoma patients), NCT01166542 (evaluating the efficacy of Reolysin® in HNC patients) as well as NCT00769704 and NCT01368276 (both investigating the antineoplastic potential of talimogene laherparepvec in melanoma patients). Of note, Amgen representatives have very recently declared that NCT00769704 met its primary endpoint of durable response rate, defined as the rate of complete or partial response lasting continuously for at least six months (source www.amgen.com/media/media_pr_detail.jsp?releaseID=1798143). Thus, talimogene laherparepvec appears relatively close to being approved by FDA for use in melanoma patients.

Concluding Remarks

During the last two decades, great efforts have been dedicated at the development of viruses that would selectively and efficiently kill malignant cells while sparing their normal counterparts. Accumulating clinical evidence indicates that oncolytic virotherapy is generally well tolerated and, at least under specific circumstances, exerts durable antineoplastic effects. Importantly, although some degree of immunosuppression initially favors transduction and viral spread, the antineoplastic potential of oncolytic viruses in fine appears to rely on the elicitation of tumor-specific immune responses.9,11,47,48 This notion is best exemplified by the fact that oncolytic viruses engineered to drive the expression of immunostimulatory factors, such as GM-CSF and IFNβ, are usually more efficient at promoting tumor regression than their naturally occurring counterparts.52,173,202,203

As oncolytic virotherapy may soon move from the bench to the beside, future studies will have to elucidate to which extent, if any, the clinical efficacy of oncolytic viruses can be improved by combining them with appropriate immunomodulatory interventions. At least theoretically, both (pre-conditioning) immunosuppressive regimens and robust immunostimulatory interventions such as the local or systemic administration of recombinant cytokines,118,204 Toll-like receptor (TLR) agonists,119,120,205,206 or immune checkpoint-blocking antibodies,115,124,188,207 may significantly boost the therapeutic effects of oncolytic virotherapy, yet optimal schedules and administration routes to achieve this objective will have to be carefully defined. Rommelfanger and colleagues have indeed demonstrated that the intratumoral administration of lipopolysaccharide (a natural TLR2/TLR4 agonist) significantly enhances the antineoplastic potential of an oncolytic vesicular stomatitis virus administered i.t. to melanoma-bearing mice, yet drives a lethal inflammatory syndrome in the majority of animals when viral particles are given i.v.208 As several distinct oncolytic viruses delivered i.t. have been shown to induce clinical responses even in distant, non-injected lesions,62,168,170 the local—as opposed to systemic—route of administration stands out as a safe and efficient alternative in this setting. Of note, encouraging results have also been obtained by combining oncolytic viruses with selected chemotherapeutics, such as the microtubular inhibitor paclitaxel.209 It is tempting to speculate, yet remains to be formally demonstrated, that such an effect may originate from the capacity of some combinatorial antineoplastic regimens to elicit immunogenic cell death.210,211 Irrespective of this unresolved issue, the rational combination of oncolytic virotherapy with immunotherapy and chemotherapy is expected to drive the development of ever more efficient clinical protocols for the treatment of cancer.

Acknowledgments

Authors are supported by the Ligue Nationale contre le Cancer (équipe labellisée), Agence Nationale de la Recherche, AXA Chair for Longevity Research, Association pour la Recherche sur le Cancer, Cancéropôle Ile-de-France, Institut National du Cancer (INCa), Fondation Bettencourt-Schueller, Fondation de France, Fondation pour la Recherche Médicale, the European Commission (ArtForce), the European Research Council, the LabEx Immuno-Oncology, the SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination (Socrate), the Cancer Research and Personalized Medicine (Carpem) consortium, and the Paris Alliance of Cancer Research Institutes (Pacri).

Glossary

Abbreviations:

CD40L

CD40 ligand

CRC

colorectal carcinoma

GM-CSF

granulocyte monocyte colony-stimulating factor

HNC

head and neck cancer

HSV

herpes simplex virus

IFN

interferon

IL

interleukin

IRES

internal ribosome entry site

MPS

mononuclear phagocytic system

TK

thymidine kinase

TLR

Toll-like receptor

TNFα

tumor necrosis factor α

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Footnotes

These authors share senior co-authorship.

References

  • 1.Kelly E, Russell SJ. History of oncolytic viruses: genesis to genetic engineering. Mol Ther. 2007;15:651–9. doi: 10.1038/sj.mt.6300108. [DOI] [PubMed] [Google Scholar]
  • 2.Pack GT. Note on the experimental use of rabies vaccine for melanomatosis. AMA Arch Derm Syphilol. 1950;62:694–5. doi: 10.1001/archderm.1950.01530180083015. [DOI] [PubMed] [Google Scholar]
  • 3.Southam CM. Present status of oncolytic virus studies. Trans N Y Acad Sci. 1960;22:657–73. doi: 10.1111/j.2164-0947.1960.tb00739.x. [DOI] [PubMed] [Google Scholar]
  • 4.Southam CM, Hilleman MR, Werner JH. Pathogenicity and oncolytic capacity of RI virus strain RI-67 in man. J Lab Clin Med. 1956;47:573–82. [PubMed] [Google Scholar]
  • 5.Huebner RJ, Rowe WP, Schatten WE, Smith RR, Thomas LB. Studies on the use of viruses in the treatment of carcinoma of the cervix. Cancer. 1956;9:1211–8. doi: 10.1002/1097-0142(195611/12)9:6<1211::AID-CNCR2820090624>3.0.CO;2-7. [DOI] [PubMed] [Google Scholar]
  • 6.Newman W, Southam CM. Virus treatment in advanced cancer; a pathological study of fifty-seven cases. Cancer. 1954;7:106–18. doi: 10.1002/1097-0142(195401)7:1<106::AID-CNCR2820070112>3.0.CO;2-L. [DOI] [PubMed] [Google Scholar]
  • 7.Wheelock EF, Dingle JH. Observations on the repeated administration of viruses to a patient with acute leukemia. A preliminary report. N Engl J Med. 1964;271:645–51. doi: 10.1056/NEJM196409242711302. [DOI] [PubMed] [Google Scholar]
  • 8.Stanford MM, Bell JC, Vähä-Koskela MJ. Novel oncolytic viruses: riding high on the next wave? Cytokine Growth Factor Rev. 2010;21:177–83. doi: 10.1016/j.cytogfr.2010.02.012. [DOI] [PubMed] [Google Scholar]
  • 9.Vähä-Koskela MJ, Heikkilä JE, Hinkkanen AE. Oncolytic viruses in cancer therapy. Cancer Lett. 2007;254:178–216. doi: 10.1016/j.canlet.2007.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Quetglas JI, John LB, Kershaw MH, Alvarez-Vallina L, Melero I, Darcy PK, et al. Virotherapy, gene transfer and immunostimulatory monoclonal antibodies. Oncoimmunology. 2012;1:1344–54. doi: 10.4161/onci.21679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Russell SJ, Peng KW, Bell JC. Oncolytic virotherapy. Nat Biotechnol. 2012;30:658–70. doi: 10.1038/nbt.2287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Griffith TS, Kawakita M, Tian J, Ritchey J, Tartaglia J, Sehgal I, et al. Inhibition of murine prostate tumor growth and activation of immunoregulatory cells with recombinant canarypox viruses. J Natl Cancer Inst. 2001;93:998–1007. doi: 10.1093/jnci/93.13.998. [DOI] [PubMed] [Google Scholar]
  • 13.Mäkelä AR, Matilainen H, White DJ, Ruoslahti E, Oker-Blom C. Enhanced baculovirus-mediated transduction of human cancer cells by tumor-homing peptides. J Virol. 2006;80:6603–11. doi: 10.1128/JVI.00528-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Singh P, Destito G, Schneemann A, Manchester M. Canine parvovirus-like particles, a novel nanomaterial for tumor targeting. J Nanobiotechnology. 2006;4:2. doi: 10.1186/1477-3155-4-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ayala-Breton C, Barber GN, Russell SJ, Peng KW. Retargeting vesicular stomatitis virus using measles virus envelope glycoproteins. Hum Gene Ther. 2012;23:484–91. doi: 10.1089/hum.2011.146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Muik A, Kneiske I, Werbizki M, Wilflingseder D, Giroglou T, Ebert O, et al. Pseudotyping vesicular stomatitis virus with lymphocytic choriomeningitis virus glycoproteins enhances infectivity for glioma cells and minimizes neurotropism. J Virol. 2011;85:5679–84. doi: 10.1128/JVI.02511-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Passer BJ, Cheema T, Zhou B, Wakimoto H, Zaupa C, Razmjoo M, et al. Identification of the ENT1 antagonists dipyridamole and dilazep as amplifiers of oncolytic herpes simplex virus-1 replication. Cancer Res. 2010;70:3890–5. doi: 10.1158/0008-5472.CAN-10-0155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lee CY, Bu LX, DeBenedetti A, Williams BJ, Rennie PS, Jia WW. Transcriptional and translational dual-regulated oncolytic herpes simplex virus type 1 for targeting prostate tumors. Mol Ther. 2010;18:929–35. doi: 10.1038/mt.2010.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Cuevas Y, Hernández-Alcoceba R, Aragones J, Naranjo-Suárez S, Castellanos MC, Esteban MA, et al. Specific oncolytic effect of a new hypoxia-inducible factor-dependent replicative adenovirus on von Hippel-Lindau-defective renal cell carcinomas. Cancer Res. 2003;63:6877–84. [PubMed] [Google Scholar]
  • 20.Post DE, Van Meir EG. A novel hypoxia-inducible factor (HIF) activated oncolytic adenovirus for cancer therapy. Oncogene. 2003;22:2065–72. doi: 10.1038/sj.onc.1206464. [DOI] [PubMed] [Google Scholar]
  • 21.Yang X, Chen E, Jiang H, Muszynski K, Harris RD, Giardina SL, et al. Evaluation of IRES-mediated, cell-type-specific cytotoxicity of poliovirus using a colorimetric cell proliferation assay. J Virol Methods. 2009;155:44–54. doi: 10.1016/j.jviromet.2008.09.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Banaszynski LA, Sellmyer MA, Contag CH, Wandless TJ, Thorne SH. Chemical control of protein stability and function in living mice. Nat Med. 2008;14:1123–7. doi: 10.1038/nm.1754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Glass M, Busche A, Wagner K, Messerle M, Borst EM. Conditional and reversible disruption of essential herpesvirus proteins. Nat Methods. 2009;6:577–9. doi: 10.1038/nmeth.1346. [DOI] [PubMed] [Google Scholar]
  • 24.Stojdl DF, Lichty BD, tenOever BR, Paterson JM, Power AT, Knowles S, et al. VSV strains with defects in their ability to shutdown innate immunity are potent systemic anti-cancer agents. Cancer Cell. 2003;4:263–75. doi: 10.1016/S1535-6108(03)00241-1. [DOI] [PubMed] [Google Scholar]
  • 25.Sarinella F, Calistri A, Sette P, Palù G, Parolin C. Oncolysis of pancreatic tumour cells by a gamma34.5-deleted HSV-1 does not rely upon Ras-activation, but on the PI 3-kinase pathway. Gene Ther. 2006;13:1080–7. doi: 10.1038/sj.gt.3302770. [DOI] [PubMed] [Google Scholar]
  • 26.Krishnamurthy S, Takimoto T, Scroggs RA, Portner A. Differentially regulated interferon response determines the outcome of Newcastle disease virus infection in normal and tumor cell lines. J Virol. 2006;80:5145–55. doi: 10.1128/JVI.02618-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Stojdl DF, Lichty B, Knowles S, Marius R, Atkins H, Sonenberg N, et al. Exploiting tumor-specific defects in the interferon pathway with a previously unknown oncolytic virus. Nat Med. 2000;6:821–5. doi: 10.1038/77558. [DOI] [PubMed] [Google Scholar]
  • 28.Coffey MC, Strong JE, Forsyth PA, Lee PW. Reovirus therapy of tumors with activated Ras pathway. Science. 1998;282:1332–4. doi: 10.1126/science.282.5392.1332. [DOI] [PubMed] [Google Scholar]
  • 29.Sugio K, Sakurai F, Katayama K, Tashiro K, Matsui H, Kawabata K, et al. Enhanced safety profiles of the telomerase-specific replication-competent adenovirus by incorporation of normal cell-specific microRNA-targeted sequences. Clin Cancer Res. 2011;17:2807–18. doi: 10.1158/1078-0432.CCR-10-2008. [DOI] [PubMed] [Google Scholar]
  • 30.Kelly EJ, Nace R, Barber GN, Russell SJ. Attenuation of vesicular stomatitis virus encephalitis through microRNA targeting. J Virol. 2010;84:1550–62. doi: 10.1128/JVI.01788-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Edge RE, Falls TJ, Brown CW, Lichty BD, Atkins H, Bell JC. A let-7 MicroRNA-sensitive vesicular stomatitis virus demonstrates tumor-specific replication. Mol Ther. 2008;16:1437–43. doi: 10.1038/mt.2008.130. [DOI] [PubMed] [Google Scholar]
  • 32.Kelly EJ, Hadac EM, Greiner S, Russell SJ. Engineering microRNA responsiveness to decrease virus pathogenicity. Nat Med. 2008;14:1278–83. doi: 10.1038/nm.1776. [DOI] [PubMed] [Google Scholar]
  • 33.Wildner O, Blaese RM, Morris JC. Therapy of colon cancer with oncolytic adenovirus is enhanced by the addition of herpes simplex virus-thymidine kinase. Cancer Res. 1999;59:410–3. [PubMed] [Google Scholar]
  • 34.Tseng JC, Zanzonico PB, Levin B, Finn R, Larson SM, Meruelo D. Tumor-specific in vivo transfection with HSV-1 thymidine kinase gene using a Sindbis viral vector as a basis for prodrug ganciclovir activation and PET. J Nucl Med. 2006;47:1136–43. [PubMed] [Google Scholar]
  • 35.Foloppe J, Kintz J, Futin N, Findeli A, Cordier P, Schlesinger Y, et al. Targeted delivery of a suicide gene to human colorectal tumors by a conditionally replicating vaccinia virus. Gene Ther. 2008;15:1361–71. doi: 10.1038/gt.2008.82. [DOI] [PubMed] [Google Scholar]
  • 36.Liu Y, Deisseroth A. Oncolytic adenoviral vector carrying the cytosine deaminase gene for melanoma gene therapy. Cancer Gene Ther. 2006;13:845–55. doi: 10.1038/sj.cgt.7700962. [DOI] [PubMed] [Google Scholar]
  • 37.Leveille S, Samuel S, Goulet ML, Hiscott J. Enhancing VSV oncolytic activity with an improved cytosine deaminase suicide gene strategy. Cancer Gene Ther. 2011;18:435–43. doi: 10.1038/cgt.2011.14. [DOI] [PubMed] [Google Scholar]
  • 38.Shinoura N, Yoshida Y, Asai A, Kirino T, Hamada H. Adenovirus-mediated transfer of p53 and Fas ligand drastically enhances apoptosis in gliomas. Cancer Gene Ther. 2000;7:732–8. doi: 10.1038/sj.cgt.7700160. [DOI] [PubMed] [Google Scholar]
  • 39.Zhao L, Dong A, Gu J, Liu Z, Zhang Y, Zhang W, et al. The antitumor activity of TRAIL and IL-24 with replicating oncolytic adenovirus in colorectal cancer. Cancer Gene Ther. 2006;13:1011–22. doi: 10.1038/sj.cgt.7700969. [DOI] [PubMed] [Google Scholar]
  • 40.Jiang G, Li J, Zeng Z, Xian L. Lentivirus-mediated gene therapy by suppressing survivin in BALB/c nude mice bearing oral squamous cell carcinoma. Cancer Biol Ther. 2006;5:435–40. doi: 10.4161/cbt.5.4.2542. [DOI] [PubMed] [Google Scholar]
  • 41.Cattaneo R, Miest T, Shashkova EV, Barry MA. Reprogrammed viruses as cancer therapeutics: targeted, armed and shielded. Nat Rev Microbiol. 2008;6:529–40. doi: 10.1038/nrmicro1927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Labrada L, Bodelón G, Viñuela J, Benavente J. Avian reoviruses cause apoptosis in cultured cells: viral uncoating, but not viral gene expression, is required for apoptosis induction. J Virol. 2002;76:7932–41. doi: 10.1128/JVI.76.16.7932-7941.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Galluzzi L, Brenner C, Morselli E, Touat Z, Kroemer G. Viral control of mitochondrial apoptosis. PLoS Pathog. 2008;4:e1000018. doi: 10.1371/journal.ppat.1000018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Olijslagers S, Dege AY, Dinsart C, Voorhoeve M, Rommelaere J, Noteborn MH, et al. Potentiation of a recombinant oncolytic parvovirus by expression of Apoptin. Cancer Gene Ther. 2001;8:958–65. doi: 10.1038/sj.cgt.7700392. [DOI] [PubMed] [Google Scholar]
  • 45.Pietersen AM, van der Eb MM, Rademaker HJ, van den Wollenberg DJ, Rabelink MJ, Kuppen PJ, et al. Specific tumor-cell killing with adenovirus vectors containing the apoptin gene. Gene Ther. 1999;6:882–92. doi: 10.1038/sj.gt.3300876. [DOI] [PubMed] [Google Scholar]
  • 46.Branton PE, Roopchand DE. The role of adenovirus E4orf4 protein in viral replication and cell killing. Oncogene. 2001;20:7855–65. doi: 10.1038/sj.onc.1204862. [DOI] [PubMed] [Google Scholar]
  • 47.Chinnery F, King CA, Elliott T, Bateman AR, James E. Viral antigen mediated NKp46 activation of NK cells results in tumor rejection via NK-DC crosstalk. Oncoimmunology. 2012;1:874–83. doi: 10.4161/onci.20636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Woller N, Kühnel F, Kubicka S. Virus infections in tumors pave the way for tumor-directed DC-vaccines. Oncoimmunology. 2012;1:208–10. doi: 10.4161/onci.1.2.18099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Diaconu I, Cerullo V, Hirvinen ML, Escutenaire S, Ugolini M, Pesonen SK, et al. Immune response is an important aspect of the antitumor effect produced by a CD40L-encoding oncolytic adenovirus. Cancer Res. 2012;72:2327–38. doi: 10.1158/0008-5472.CAN-11-2975. [DOI] [PubMed] [Google Scholar]
  • 50.Pesonen S, Diaconu I, Kangasniemi L, Ranki T, Kanerva A, Pesonen SK, et al. Oncolytic immunotherapy of advanced solid tumors with a CD40L-expressing replicating adenovirus: assessment of safety and immunologic responses in patients. Cancer Res. 2012;72:1621–31. doi: 10.1158/0008-5472.CAN-11-3001. [DOI] [PubMed] [Google Scholar]
  • 51.Gomes EM, Rodrigues MS, Phadke AP, Butcher LD, Starling C, Chen S, et al. Antitumor activity of an oncolytic adenoviral-CD40 ligand (CD154) transgene construct in human breast cancer cells. Clin Cancer Res. 2009;15:1317–25. doi: 10.1158/1078-0432.CCR-08-1360. [DOI] [PubMed] [Google Scholar]
  • 52.Choi KJ, Kim JH, Lee YS, Kim J, Suh BS, Kim H, et al. Concurrent delivery of GM-CSF and B7-1 using an oncolytic adenovirus elicits potent antitumor effect. Gene Ther. 2006;13:1010–20. doi: 10.1038/sj.gt.3302759. [DOI] [PubMed] [Google Scholar]
  • 53.Zamarin D, Vigil A, Kelly K, García-Sastre A, Fong Y. Genetically engineered Newcastle disease virus for malignant melanoma therapy. Gene Ther. 2009;16:796–804. doi: 10.1038/gt.2009.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Wong RJ, Chan MK, Yu Z, Ghossein RA, Ngai I, Adusumilli PS, et al. Angiogenesis inhibition by an oncolytic herpes virus expressing interleukin 12. Clin Cancer Res. 2004;10:4509–16. doi: 10.1158/1078-0432.CCR-04-0081. [DOI] [PubMed] [Google Scholar]
  • 55.Zhang SN, Choi IK, Huang JH, Yoo JY, Choi KJ, Yun CO. Optimizing DC vaccination by combination with oncolytic adenovirus coexpressing IL-12 and GM-CSF. Mol Ther. 2011;19:1558–68. doi: 10.1038/mt.2011.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Parker JN, Gillespie GY, Love CE, Randall S, Whitley RJ, Markert JM. Engineered herpes simplex virus expressing IL-12 in the treatment of experimental murine brain tumors. Proc Natl Acad Sci U S A. 2000;97:2208–13. doi: 10.1073/pnas.040557897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Stephenson KB, Barra NG, Davies E, Ashkar AA, Lichty BD. Expressing human interleukin-15 from oncolytic vesicular stomatitis virus improves survival in a murine metastatic colon adenocarcinoma model through the enhancement of anti-tumor immunity. Cancer Gene Ther. 2012;19:238–46. doi: 10.1038/cgt.2011.81. [DOI] [PubMed] [Google Scholar]
  • 58.Liu J, Wennier S, Reinhard M, Roy E, MacNeill A, McFadden G. Myxoma virus expressing interleukin-15 fails to cause lethal myxomatosis in European rabbits. J Virol. 2009;83:5933–8. doi: 10.1128/JVI.00204-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.van Rikxoort M, Michaelis M, Wolschek M, Muster T, Egorov A, Seipelt J, et al. Oncolytic effects of a novel influenza A virus expressing interleukin-15 from the NS reading frame. PLoS One. 2012;7:e36506. doi: 10.1371/journal.pone.0036506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Pesonen S, Diaconu I, Cerullo V, Escutenaire S, Raki M, Kangasniemi L, et al. Integrin targeted oncolytic adenoviruses Ad5-D24-RGD and Ad5-RGD-D24-GMCSF for treatment of patients with advanced chemotherapy refractory solid tumors. Int J Cancer. 2012;130:1937–47. doi: 10.1002/ijc.26216. [DOI] [PubMed] [Google Scholar]
  • 61.Burke JM, Lamm DL, Meng MV, Nemunaitis JJ, Stephenson JJ, Arseneau JC, et al. A first in human phase 1 study of CG0070, a GM-CSF expressing oncolytic adenovirus, for the treatment of nonmuscle invasive bladder cancer. J Urol. 2012;188:2391–7. doi: 10.1016/j.juro.2012.07.097. [DOI] [PubMed] [Google Scholar]
  • 62.Heo J, Reid T, Ruo L, Breitbach CJ, Rose S, Bloomston M, et al. Randomized dose-finding clinical trial of oncolytic immunotherapeutic vaccinia JX-594 in liver cancer. Nat Med. 2013;19:329–36. doi: 10.1038/nm.3089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Colombo F, Barzon L, Franchin E, Pacenti M, Pinna V, Danieli D, et al. Combined HSV-TK/IL-2 gene therapy in patients with recurrent glioblastoma multiforme: biological and clinical results. Cancer Gene Ther. 2005;12:835–48. doi: 10.1038/sj.cgt.7700851. [DOI] [PubMed] [Google Scholar]
  • 64.Lun X, Alain T, Zemp FJ, Zhou H, Rahman MM, Hamilton MG, et al. Myxoma virus virotherapy for glioma in immunocompetent animal models: optimizing administration routes and synergy with rapamycin. Cancer Res. 2010;70:598–608. doi: 10.1158/0008-5472.CAN-09-1510. [DOI] [PubMed] [Google Scholar]
  • 65.Ram Z, Culver KW, Oshiro EM, Viola JJ, DeVroom HL, Otto E, et al. Therapy of malignant brain tumors by intratumoral implantation of retroviral vector-producing cells. Nat Med. 1997;3:1354–61. doi: 10.1038/nm1297-1354. [DOI] [PubMed] [Google Scholar]
  • 66.Smyth JW, Fleeton MN, Sheahan BJ, Atkins GJ. Treatment of rapidly growing K-BALB and CT26 mouse tumours using Semliki Forest virus and its derived vector. Gene Ther. 2005;12:147–59. doi: 10.1038/sj.gt.3302390. [DOI] [PubMed] [Google Scholar]
  • 67.Vähä-Koskela MJ, Kallio JP, Jansson LC, Heikkilä JE, Zakhartchenko VA, Kallajoki MA, et al. Oncolytic capacity of attenuated replicative semliki forest virus in human melanoma xenografts in severe combined immunodeficient mice. Cancer Res. 2006;66:7185–94. doi: 10.1158/0008-5472.CAN-05-2214. [DOI] [PubMed] [Google Scholar]
  • 68.Cordaro TA, de Visser KE, Tirion FH, Graus YM, Haanen JB, Kioussis D, et al. Tumor size at the time of adoptive transfer determines whether tumor rejection occurs. Eur J Immunol. 2000;30:1297–307. doi: 10.1002/(SICI)1521-4141(200005)30:5<1297::AID-IMMU1297>3.0.CO;2-C. [DOI] [PubMed] [Google Scholar]
  • 69.Li ZY, Ni S, Yang X, Kiviat N, Lieber A. Xenograft models for liver metastasis: Relationship between tumor morphology and adenovirus vector transduction. Mol Ther. 2004;9:650–7. doi: 10.1016/j.ymthe.2004.01.021. [DOI] [PubMed] [Google Scholar]
  • 70.Shayakhmetov DM, Li ZY, Ni S, Lieber A. Targeting of adenovirus vectors to tumor cells does not enable efficient transduction of breast cancer metastases. Cancer Res. 2002;62:1063–8. [PubMed] [Google Scholar]
  • 71.Stohrer M, Boucher Y, Stangassinger M, Jain RK. Oncotic pressure in solid tumors is elevated. Cancer Res. 2000;60:4251–5. [PubMed] [Google Scholar]
  • 72.Bilbao R, Bustos M, Alzuguren P, Pajares MJ, Drozdzik M, Qian C, et al. A blood-tumor barrier limits gene transfer to experimental liver cancer: the effect of vasoactive compounds. Gene Ther. 2000;7:1824–32. doi: 10.1038/sj.gt.3301312. [DOI] [PubMed] [Google Scholar]
  • 73.Cairns R, Papandreou I, Denko N. Overcoming physiologic barriers to cancer treatment by molecularly targeting the tumor microenvironment. Mol Cancer Res. 2006;4:61–70. doi: 10.1158/1541-7786.MCR-06-0002. [DOI] [PubMed] [Google Scholar]
  • 74.Galluzzi L, Vitale I, Abrams JM, Alnemri ES, Baehrecke EH, Blagosklonny MV, et al. Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ. 2012;19:107–20. doi: 10.1038/cdd.2011.96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Grote D, Russell SJ, Cornu TI, Cattaneo R, Vile R, Poland GA, et al. Live attenuated measles virus induces regression of human lymphoma xenografts in immunodeficient mice. Blood. 2001;97:3746–54. doi: 10.1182/blood.V97.12.3746. [DOI] [PubMed] [Google Scholar]
  • 76.Peng KW, TenEyck CJ, Galanis E, Kalli KR, Hartmann LC, Russell SJ. Intraperitoneal therapy of ovarian cancer using an engineered measles virus. Cancer Res. 2002;62:4656–62. [PubMed] [Google Scholar]
  • 77.Kanerva A, Zinn KR, Peng KW, Ranki T, Kangasniemi L, Chaudhuri TR, et al. Noninvasive dual modality in vivo monitoring of the persistence and potency of a tumor targeted conditionally replicating adenovirus. Gene Ther. 2005;12:87–94. doi: 10.1038/sj.gt.3302387. [DOI] [PubMed] [Google Scholar]
  • 78.Alain T, Kim M, Johnston RN, Urbanski S, Kossakowska AE, Forsyth PA, et al. The oncolytic effect in vivo of reovirus on tumour cells that have survived reovirus cell killing in vitro. Br J Cancer. 2006;95:1020–7. doi: 10.1038/sj.bjc.6603363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kim YT, Ganly I, Brown R, Stuart D. Acquired resistance to cytolysis of the E1B-attenuated adenovirus, dl1520, in ovarian tumour cell lines. Cancer Gene Ther. 2003;10:589–90. doi: 10.1038/sj.cgt.7700607. [DOI] [PubMed] [Google Scholar]
  • 80.Vitale I, Galluzzi L, Castedo M, Kroemer G. Mitotic catastrophe: a mechanism for avoiding genomic instability. Nat Rev Mol Cell Biol. 2011;12:385–92. doi: 10.1038/nrm3115. [DOI] [PubMed] [Google Scholar]
  • 81.Vitale I, Galluzzi L, Senovilla L, Criollo A, Jemaà M, Castedo M, et al. Illicit survival of cancer cells during polyploidization and depolyploidization. Cell Death Differ. 2011;18:1403–13. doi: 10.1038/cdd.2010.145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.López-Guerrero JA, Rayet B, Tuynder M, Rommelaere J, Dinsart C. Constitutive activation of U937 promonocytic cell clones selected for their resistance to parvovirus H-1 infection. Blood. 1997;89:1642–53. [PubMed] [Google Scholar]
  • 83.Rayet B, Lopez-Guerrero JA, Rommelaere J, Dinsart C. Induction of programmed cell death by parvovirus H-1 in U937 cells: connection with the tumor necrosis factor alpha signalling pathway. J Virol. 1998;72:8893–903. doi: 10.1128/jvi.72.11.8893-8903.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Telerman A, Tuynder M, Dupressoir T, Robaye B, Sigaux F, Shaulian E, et al. A model for tumor suppression using H-1 parvovirus. Proc Natl Acad Sci U S A. 1993;90:8702–6. doi: 10.1073/pnas.90.18.8702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Fisher KD, Stallwood Y, Green NK, Ulbrich K, Mautner V, Seymour LW. Polymer-coated adenovirus permits efficient retargeting and evades neutralising antibodies. Gene Ther. 2001;8:341–8. doi: 10.1038/sj.gt.3301389. [DOI] [PubMed] [Google Scholar]
  • 86.Massari I, Donnini A, Argentati K, Straino S, Mangoni A, Gaetano C, et al. Age-dependent effects of repeated immunization with a first generation adenovirus vector on the immune response and transgene expression in young and old rats. Exp Gerontol. 2002;37:823–31. doi: 10.1016/S0531-5565(02)00011-6. [DOI] [PubMed] [Google Scholar]
  • 87.Ikeda K, Wakimoto H, Ichikawa T, Jhung S, Hochberg FH, Louis DN, et al. Complement depletion facilitates the infection of multiple brain tumors by an intravascular, replication-conditional herpes simplex virus mutant. J Virol. 2000;74:4765–75. doi: 10.1128/JVI.74.10.4765-4775.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Pensiero MN, Wysocki CA, Nader K, Kikuchi GE. Development of amphotropic murine retrovirus vectors resistant to inactivation by human serum. Hum Gene Ther. 1996;7:1095–101. doi: 10.1089/hum.1996.7.9-1095. [DOI] [PubMed] [Google Scholar]
  • 89.Bernt KM, Ni S, Gaggar A, Li ZY, Shayakhmetov DM, Lieber A. The effect of sequestration by nontarget tissues on anti-tumor efficacy of systemically applied, conditionally replicating adenovirus vectors. Mol Ther. 2003;8:746–55. doi: 10.1016/j.ymthe.2003.07.006. [DOI] [PubMed] [Google Scholar]
  • 90.Underhill DM, Ozinsky A. Phagocytosis of microbes: complexity in action. Annu Rev Immunol. 2002;20:825–52. doi: 10.1146/annurev.immunol.20.103001.114744. [DOI] [PubMed] [Google Scholar]
  • 91.Doronin K, Shashkova EV, May SM, Hofherr SE, Barry MA. Chemical modification with high molecular weight polyethylene glycol reduces transduction of hepatocytes and increases efficacy of intravenously delivered oncolytic adenovirus. Hum Gene Ther. 2009;20:975–88. doi: 10.1089/hum.2009.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Ferguson MS, Lemoine NR, Wang Y. Systemic delivery of oncolytic viruses: hopes and hurdles. Adv Virol. 2012;2012:805629. doi: 10.1155/2012/805629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Brenner C, Galluzzi L, Kepp O, Kroemer G. Decoding cell death signals in liver inflammation. J Hepatol. 2013 doi: 10.1016/j.jhep.2013.03.033. In press. [DOI] [PubMed] [Google Scholar]
  • 94.Chisholm J, Howe K, Taj M, Zambon M. Influenza immunisation in children with solid tumours. Eur J Cancer. 2005;41:2280–7. doi: 10.1016/j.ejca.2005.07.006. [DOI] [PubMed] [Google Scholar]
  • 95.Cooksley CD, Avritscher EB, Bekele BN, Rolston KV, Geraci JM, Elting LS. Epidemiology and outcomes of serious influenza-related infections in the cancer population. Cancer. 2005;104:618–28. doi: 10.1002/cncr.21203. [DOI] [PubMed] [Google Scholar]
  • 96.Hicks KL, Chemaly RF, Kontoyiannis DP. Common community respiratory viruses in patients with cancer: more than just “common colds”. Cancer. 2003;97:2576–87. doi: 10.1002/cncr.11353. [DOI] [PubMed] [Google Scholar]
  • 97.Nemunaitis J, Khuri F, Ganly I, Arseneau J, Posner M, Vokes E, et al. Phase II trial of intratumoral administration of ONYX-015, a replication-selective adenovirus, in patients with refractory head and neck cancer. J Clin Oncol. 2001;19:289–98. doi: 10.1200/JCO.2001.19.2.289. [DOI] [PubMed] [Google Scholar]
  • 98.Geevarghese SK, Geller DA, de Haan HA, Hörer M, Knoll AE, Mescheder A, et al. Phase I/II study of oncolytic herpes simplex virus NV1020 in patients with extensively pretreated refractory colorectal cancer metastatic to the liver. Hum Gene Ther. 2010;21:1119–28. doi: 10.1089/hum.2010.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Hacein-Bey-Abina S, von Kalle C, Schmidt M, Le Deist F, Wulffraat N, McIntyre E, et al. A serious adverse event after successful gene therapy for X-linked severe combined immunodeficiency. N Engl J Med. 2003;348:255–6. doi: 10.1056/NEJM200301163480314. [DOI] [PubMed] [Google Scholar]
  • 100.Hacein-Bey-Abina S, Von Kalle C, Schmidt M, McCormack MP, Wulffraat N, Leboulch P, et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science. 2003;302:415–9. doi: 10.1126/science.1088547. [DOI] [PubMed] [Google Scholar]
  • 101.Eto Y, Yoshioka Y, Mukai Y, Okada N, Nakagawa S. Development of PEGylated adenovirus vector with targeting ligand. Int J Pharm. 2008;354:3–8. doi: 10.1016/j.ijpharm.2007.08.025. [DOI] [PubMed] [Google Scholar]
  • 102.Duncan R. Polymer conjugates as anticancer nanomedicines. Nat Rev Cancer. 2006;6:688–701. doi: 10.1038/nrc1958. [DOI] [PubMed] [Google Scholar]
  • 103.Morrison J, Briggs SS, Green N, Fisher K, Subr V, Ulbrich K, et al. Virotherapy of ovarian cancer with polymer-cloaked adenovirus retargeted to the epidermal growth factor receptor. Mol Ther. 2008;16:244–51. doi: 10.1038/sj.mt.6300363. [DOI] [PubMed] [Google Scholar]
  • 104.Fang J, Nakamura H, Maeda H. The EPR effect: Unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect. Adv Drug Deliv Rev. 2011;63:136–51. doi: 10.1016/j.addr.2010.04.009. [DOI] [PubMed] [Google Scholar]
  • 105.Jain RK, Stylianopoulos T. Delivering nanomedicine to solid tumors. Nat Rev Clin Oncol. 2010;7:653–64. doi: 10.1038/nrclinonc.2010.139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Diop-Frimpong B, Chauhan VP, Krane S, Boucher Y, Jain RK. Losartan inhibits collagen I synthesis and improves the distribution and efficacy of nanotherapeutics in tumors. Proc Natl Acad Sci U S A. 2011;108:2909–14. doi: 10.1073/pnas.1018892108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Ganesh S, Gonzalez-Edick M, Gibbons D, Van Roey M, Jooss K. Intratumoral coadministration of hyaluronidase enzyme and oncolytic adenoviruses enhances virus potency in metastatic tumor models. Clin Cancer Res. 2008;14:3933–41. doi: 10.1158/1078-0432.CCR-07-4732. [DOI] [PubMed] [Google Scholar]
  • 108.Guedan S, Rojas JJ, Gros A, Mercade E, Cascallo M, Alemany R. Hyaluronidase expression by an oncolytic adenovirus enhances its intratumoral spread and suppresses tumor growth. Mol Ther. 2010;18:1275–83. doi: 10.1038/mt.2010.79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov. 2012;11:215–33. doi: 10.1038/nrd3626. [DOI] [PubMed] [Google Scholar]
  • 110.Kaluza KM, Vile R. Improving the outcome of adoptive cell transfer by targeting tumor escape. Oncoimmunology. 2013;2:e22059. doi: 10.4161/onci.22059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Pere H, Tanchot C, Bayry J, Terme M, Taieb J, Badoual C, et al. Comprehensive analysis of current approaches to inhibit regulatory T cells in cancer. Oncoimmunology. 2012;1:326–33. doi: 10.4161/onci.18852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Chiocca EA. The host response to cancer virotherapy. Curr Opin Mol Ther. 2008;10:38–45. [PubMed] [Google Scholar]
  • 113.Wilson JM. Gendicine: the first commercial gene therapy product. Hum Gene Ther. 2005;16:1014–5. doi: 10.1089/hum.2005.16.1014. [DOI] [PubMed] [Google Scholar]
  • 114.Peng Z. Current status of gendicine in China: recombinant human Ad-p53 agent for treatment of cancers. Hum Gene Ther. 2005;16:1016–27. doi: 10.1089/hum.2005.16.1016. [DOI] [PubMed] [Google Scholar]
  • 115.Galluzzi L, Vacchelli E, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, et al. Trial Watch: Monoclonal antibodies in cancer therapy. Oncoimmunology. 2012;1:28–37. doi: 10.4161/onci.1.1.17938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Vacchelli E, Galluzzi L, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, et al. Trial watch: Chemotherapy with immunogenic cell death inducers. Oncoimmunology. 2012;1:179–88. doi: 10.4161/onci.1.2.19026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial Watch: Adoptive cell transfer immunotherapy. Oncoimmunology. 2012;1:306–15. doi: 10.4161/onci.19549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Vacchelli E, Galluzzi L, Eggermont A, Galon J, Tartour E, Zitvogel L, et al. Trial Watch: Immunostimulatory cytokines. Oncoimmunology. 2012;1:493–506. doi: 10.4161/onci.20459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial Watch: Experimental Toll-like receptor agonists for cancer therapy. Oncoimmunology. 2012;1:699–716. doi: 10.4161/onci.20696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Vacchelli E, Galluzzi L, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial watch: FDA-approved Toll-like receptor agonists for cancer therapy. Oncoimmunology. 2012;1:894–907. doi: 10.4161/onci.20931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Galluzzi L, Senovilla L, Vacchelli E, Eggermont A, Fridman WH, Galon J, et al. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology. 2012;1:1111–34. doi: 10.4161/onci.21494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Senovilla L, Vacchelli E, Galon J, Adjemian S, Eggermont A, Fridman WH, et al. Trial watch: Prognostic and predictive value of the immune infiltrate in cancer. Oncoimmunology. 2012;1:1323–43. doi: 10.4161/onci.22009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Vacchelli E, Martins I, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial watch: Peptide vaccines in cancer therapy. Oncoimmunology. 2012;1:1557–76. doi: 10.4161/onci.22428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Vacchelli E, Eggermont A, Galon J, Sautès-Fridman C, Zitvogel L, Kroemer G, et al. Trial watch: Monoclonal antibodies in cancer therapy. Oncoimmunology. 2013;2:e22789. doi: 10.4161/onci.22789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Menger L, Vacchelli E, Kepp O, Eggermont A, Tartour E, Zitvogel L, et al. Trial watch: Cardiac glycosides and cancer therapy. Oncoimmunology. 2013;2:e23082. doi: 10.4161/onci.23082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Vacchelli E, Galluzzi L, Fridman WH, Galon J, Sautés-Fridman C, Tartour E, et al. Trial Watch: Chemotherapy with immunogenic cell death inducers. OncoImmunology. 2012;1:179–88. doi: 10.4161/onci.1.2.19026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Senovilla L, Vacchelli E, Garcia P, Eggermont A, Fridman WH, Galon J, et al. Trial Watch: DNA vaccines for cancer therapy. OncoImmunology. 2013;2:e23803. doi: 10.4161/onci.23803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial Watch: Adoptive cell transfer immunotherapy. Oncoimmunology. 2012;1:306–15. doi: 10.4161/onci.19549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Sinkovics JG. Viral oncolysates as human tumor vaccines. Int Rev Immunol. 1991;7:259–87. doi: 10.3109/08830189109114875. [DOI] [PubMed] [Google Scholar]
  • 130.Sinkovics JG, Horvath JC. Evidence accumulating in support of cancer vaccines combined with chemotherapy: a pragmatic review of past and present efforts. Int J Oncol. 2006;29:765–77. [PubMed] [Google Scholar]
  • 131.Asada T. Treatment of human cancer with mumps virus. Cancer. 1974;34:1907–28. doi: 10.1002/1097-0142(197412)34:6<1907::AID-CNCR2820340609>3.0.CO;2-4. [DOI] [PubMed] [Google Scholar]
  • 132.Martuza RL, Malick A, Markert JM, Ruffner KL, Coen DM. Experimental therapy of human glioma by means of a genetically engineered virus mutant. Science. 1991;252:854–6. doi: 10.1126/science.1851332. [DOI] [PubMed] [Google Scholar]
  • 133.Bradbury J. Oncolytic viral anti-cancer therapy: a magic bullet? Lancet. 2001;357:614. doi: 10.1016/S0140-6736(05)71407-0. [DOI] [PubMed] [Google Scholar]
  • 134.Chiocca EA. Oncolytic viruses. Nat Rev Cancer. 2002;2:938–50. doi: 10.1038/nrc948. [DOI] [PubMed] [Google Scholar]
  • 135.Bourke MG, Salwa S, Harrington KJ, Kucharczyk MJ, Forde PF, de Kruijf M, et al. The emerging role of viruses in the treatment of solid tumours. Cancer Treat Rev. 2011;37:618–32. doi: 10.1016/j.ctrv.2010.12.003. [DOI] [PubMed] [Google Scholar]
  • 136.Donnelly OG, Errington-Mais F, Prestwich R, Harrington K, Pandha H, Vile R, et al. Recent clinical experience with oncolytic viruses. Curr Pharm Biotechnol. 2012;13:1834–41. doi: 10.2174/138920112800958904. [DOI] [PubMed] [Google Scholar]
  • 137.Cerullo V, Vähä-Koskela M, Hemminki A. Oncolytic adenoviruses: A potent form of tumor immunovirotherapy. Oncoimmunology. 2012;1:979–81. doi: 10.4161/onci.20172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Kaur B, Chiocca EA, Cripe TP. Oncolytic HSV-1 virotherapy: clinical experience and opportunities for progress. Curr Pharm Biotechnol. 2012;13:1842–51. doi: 10.2174/138920112800958814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Li H, Zhang X. Oncolytic HSV as a vector in cancer immunotherapy. Methods Mol Biol. 2010;651:279–90. doi: 10.1007/978-1-60761-786-0_16. [DOI] [PubMed] [Google Scholar]
  • 140.Chiocca EA, Abbed KM, Tatter S, Louis DN, Hochberg FH, Barker F, et al. A phase I open-label, dose-escalation, multi-institutional trial of injection with an E1B-Attenuated adenovirus, ONYX-015, into the peritumoral region of recurrent malignant gliomas, in the adjuvant setting. Mol Ther. 2004;10:958–66. doi: 10.1016/j.ymthe.2004.07.021. [DOI] [PubMed] [Google Scholar]
  • 141.Galanis E, Okuno SH, Nascimento AG, Lewis BD, Lee RA, Oliveira AM, et al. Phase I-II trial of ONYX-015 in combination with MAP chemotherapy in patients with advanced sarcomas. Gene Ther. 2005;12:437–45. doi: 10.1038/sj.gt.3302436. [DOI] [PubMed] [Google Scholar]
  • 142.Ganly I, Kirn D, Eckhardt G, Rodriguez GI, Soutar DS, Otto R, et al. A phase I study of Onyx-015, an E1B attenuated adenovirus, administered intratumorally to patients with recurrent head and neck cancer. Clin Cancer Res. 2000;6:798–806. [PubMed] [Google Scholar]
  • 143.Khuri FR, Nemunaitis J, Ganly I, Arseneau J, Tannock IF, Romel L, et al. a controlled trial of intratumoral ONYX-015, a selectively-replicating adenovirus, in combination with cisplatin and 5-fluorouracil in patients with recurrent head and neck cancer. Nat Med. 2000;6:879–85. doi: 10.1038/78638. [DOI] [PubMed] [Google Scholar]
  • 144.Xia ZJ, Chang JH, Zhang L, Jiang WQ, Guan ZZ, Liu JW, et al. [Phase III randomized clinical trial of intratumoral injection of E1B gene-deleted adenovirus (H101) combined with cisplatin-based chemotherapy in treating squamous cell cancer of head and neck or esophagus] Ai Zheng. 2004;23:1666–70. [PubMed] [Google Scholar]
  • 145.Xu RH, Yuan ZY, Guan ZZ, Cao Y, Wang HQ, Hu XH, et al. [Phase II clinical study of intratumoral H101, an E1B deleted adenovirus, in combination with chemotherapy in patients with cancer] Ai Zheng. 2003;22:1307–10. [PubMed] [Google Scholar]
  • 146.Hecht JR, Bedford R, Abbruzzese JL, Lahoti S, Reid TR, Soetikno RM, et al. A phase I/II trial of intratumoral endoscopic ultrasound injection of ONYX-015 with intravenous gemcitabine in unresectable pancreatic carcinoma. Clin Cancer Res. 2003;9:555–61. [PubMed] [Google Scholar]
  • 147.Reid T, Galanis E, Abbruzzese J, Sze D, Wein LM, Andrews J, et al. Hepatic arterial infusion of a replication-selective oncolytic adenovirus (dl1520): phase II viral, immunologic, and clinical endpoints. Cancer Res. 2002;62:6070–9. [PubMed] [Google Scholar]
  • 148.Hamid O, Varterasian ML, Wadler S, Hecht JR, Benson A, 3rd, Galanis E, et al. Phase II trial of intravenous CI-1042 in patients with metastatic colorectal cancer. J Clin Oncol. 2003;21:1498–504. doi: 10.1200/JCO.2003.09.114. [DOI] [PubMed] [Google Scholar]
  • 149.Reid TR, Freeman S, Post L, McCormick F, Sze DY. Effects of Onyx-015 among metastatic colorectal cancer patients that have failed prior treatment with 5-FU/leucovorin. Cancer Gene Ther. 2005;12:673–81. doi: 10.1038/sj.cgt.7700819. [DOI] [PubMed] [Google Scholar]
  • 150.DeWeese TL, van der Poel H, Li S, Mikhak B, Drew R, Goemann M, et al. A phase I trial of CV706, a replication-competent, PSA selective oncolytic adenovirus, for the treatment of locally recurrent prostate cancer following radiation therapy. Cancer Res. 2001;61:7464–72. [PubMed] [Google Scholar]
  • 151.Freytag SO, Khil M, Stricker H, Peabody J, Menon M, DePeralta-Venturina M, et al. Phase I study of replication-competent adenovirus-mediated double suicide gene therapy for the treatment of locally recurrent prostate cancer. Cancer Res. 2002;62:4968–76. [PubMed] [Google Scholar]
  • 152.Freytag SO, Stricker H, Pegg J, Paielli D, Pradhan DG, Peabody J, et al. Phase I study of replication-competent adenovirus-mediated double-suicide gene therapy in combination with conventional-dose three-dimensional conformal radiation therapy for the treatment of newly diagnosed, intermediate- to high-risk prostate cancer. Cancer Res. 2003;63:7497–506. [PubMed] [Google Scholar]
  • 153.Kimball KJ, Preuss MA, Barnes MN, Wang M, Siegal GP, Wan W, et al. A phase I study of a tropism-modified conditionally replicative adenovirus for recurrent malignant gynecologic diseases. Clin Cancer Res. 2010;16:5277–87. doi: 10.1158/1078-0432.CCR-10-0791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Vasey PA, Shulman LN, Campos S, Davis J, Gore M, Johnston S, et al. Phase I trial of intraperitoneal injection of the E1B-55-kd-gene-deleted adenovirus ONYX-015 (dl1520) given on days 1 through 5 every 3 weeks in patients with recurrent/refractory epithelial ovarian cancer. J Clin Oncol. 2002;20:1562–9. doi: 10.1200/JCO.20.6.1562. [DOI] [PubMed] [Google Scholar]
  • 155.Nemunaitis J, Senzer N, Sarmiento S, Zhang YA, Arzaga R, Sands B, et al. A phase I trial of intravenous infusion of ONYX-015 and enbrel in solid tumor patients. Cancer Gene Ther. 2007;14:885–93. doi: 10.1038/sj.cgt.7701080. [DOI] [PubMed] [Google Scholar]
  • 156.Nemunaitis J, Tong AW, Nemunaitis M, Senzer N, Phadke AP, Bedell C, et al. A phase I study of telomerase-specific replication competent oncolytic adenovirus (telomelysin) for various solid tumors. Mol Ther. 2010;18:429–34. doi: 10.1038/mt.2009.262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Markovitz NS, Baunoch D, Roizman B. The range and distribution of murine central nervous system cells infected with the gamma(1)34.5- mutant of herpes simplex virus 1. J Virol. 1997;71:5560–9. doi: 10.1128/jvi.71.7.5560-5569.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Harrow S, Papanastassiou V, Harland J, Mabbs R, Petty R, Fraser M, et al. HSV1716 injection into the brain adjacent to tumour following surgical resection of high-grade glioma: safety data and long-term survival. Gene Ther. 2004;11:1648–58. doi: 10.1038/sj.gt.3302289. [DOI] [PubMed] [Google Scholar]
  • 159.Markert JM, Liechty PG, Wang W, Gaston S, Braz E, Karrasch M, et al. Phase Ib trial of mutant herpes simplex virus G207 inoculated pre-and post-tumor resection for recurrent GBM. Mol Ther. 2009;17:199–207. doi: 10.1038/mt.2008.228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Markert JM, Medlock MD, Rabkin SD, Gillespie GY, Todo T, Hunter WD, et al. Conditionally replicating herpes simplex virus mutant, G207 for the treatment of malignant glioma: results of a phase I trial. Gene Ther. 2000;7:867–74. doi: 10.1038/sj.gt.3301205. [DOI] [PubMed] [Google Scholar]
  • 161.Papanastassiou V, Rampling R, Fraser M, Petty R, Hadley D, Nicoll J, et al. The potential for efficacy of the modified (ICP 34.5(-)) herpes simplex virus HSV1716 following intratumoural injection into human malignant glioma: a proof of principle study. Gene Ther. 2002;9:398–406. doi: 10.1038/sj.gt.3301664. [DOI] [PubMed] [Google Scholar]
  • 162.Todo T, Martuza RL, Rabkin SD, Johnson PA. Oncolytic herpes simplex virus vector with enhanced MHC class I presentation and tumor cell killing. Proc Natl Acad Sci U S A. 2001;98:6396–401. doi: 10.1073/pnas.101136398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Kimata H, Imai T, Kikumori T, Teshigahara O, Nagasaka T, Goshima F, et al. Pilot study of oncolytic viral therapy using mutant herpes simplex virus (HF10) against recurrent metastatic breast cancer. Ann Surg Oncol. 2006;13:1078–84. doi: 10.1245/ASO.2006.08.035. [DOI] [PubMed] [Google Scholar]
  • 164.Mace AT, Ganly I, Soutar DS, Brown SM. Potential for efficacy of the oncolytic Herpes simplex virus 1716 in patients with oral squamous cell carcinoma. Head Neck. 2008;30:1045–51. doi: 10.1002/hed.20840. [DOI] [PubMed] [Google Scholar]
  • 165.Fujimoto Y, Mizuno T, Sugiura S, Goshima F, Kohno S, Nakashima T, et al. Intratumoral injection of herpes simplex virus HF10 in recurrent head and neck squamous cell carcinoma. Acta Otolaryngol. 2006;126:1115–7. doi: 10.1080/00016480600702100. [DOI] [PubMed] [Google Scholar]
  • 166.Harrington KJ, Hingorani M, Tanay MA, Hickey J, Bhide SA, Clarke PM, et al. Phase I/II study of oncolytic HSV GM-CSF in combination with radiotherapy and cisplatin in untreated stage III/IV squamous cell cancer of the head and neck. Clin Cancer Res. 2010;16:4005–15. doi: 10.1158/1078-0432.CCR-10-0196. [DOI] [PubMed] [Google Scholar]
  • 167.White RR, Stanley WE, Johnson JL, Tyler DS, Seigler HF. Long-term survival in 2,505 patients with melanoma with regional lymph node metastasis. Ann Surg. 2002;235:879–87. doi: 10.1097/00000658-200206000-00017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Kaufman HL, Kim DW, DeRaffele G, Mitcham J, Coffin RS, Kim-Schulze S. Local and distant immunity induced by intralesional vaccination with an oncolytic herpes virus encoding GM-CSF in patients with stage IIIc and IV melanoma. Ann Surg Oncol. 2010;17:718–30. doi: 10.1245/s10434-009-0809-6. [DOI] [PubMed] [Google Scholar]
  • 169.MacKie RM, Stewart B, Brown SM. Intralesional injection of herpes simplex virus 1716 in metastatic melanoma. Lancet. 2001;357:525–6. doi: 10.1016/S0140-6736(00)04048-4. [DOI] [PubMed] [Google Scholar]
  • 170.Senzer NN, Kaufman HL, Amatruda T, Nemunaitis M, Reid T, Daniels G, et al. Phase II clinical trial of a granulocyte-macrophage colony-stimulating factor-encoding, second-generation oncolytic herpesvirus in patients with unresectable metastatic melanoma. J Clin Oncol. 2009;27:5763–71. doi: 10.1200/JCO.2009.24.3675. [DOI] [PubMed] [Google Scholar]
  • 171.Nakao A, Kasuya H, Sahin TT, Nomura N, Kanzaki A, Misawa M, et al. A phase I dose-escalation clinical trial of intraoperative direct intratumoral injection of HF10 oncolytic virus in non-resectable patients with advanced pancreatic cancer. Cancer Gene Ther. 2011;18:167–75. doi: 10.1038/cgt.2010.65. [DOI] [PubMed] [Google Scholar]
  • 172.Fong Y, Kim T, Bhargava A, Schwartz L, Brown K, Brody L, et al. A herpes oncolytic virus can be delivered via the vasculature to produce biologic changes in human colorectal cancer. Mol Ther. 2009;17:389–94. doi: 10.1038/mt.2008.240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Hu JC, Coffin RS, Davis CJ, Graham NJ, Groves N, Guest PJ, et al. A phase I study of OncoVEXGM-CSF, a second-generation oncolytic herpes simplex virus expressing granulocyte macrophage colony-stimulating factor. Clin Cancer Res. 2006;12:6737–47. doi: 10.1158/1078-0432.CCR-06-0759. [DOI] [PubMed] [Google Scholar]
  • 174.Freeman AI, Zakay-Rones Z, Gomori JM, Linetsky E, Rasooly L, Greenbaum E, et al. Phase I/II trial of intravenous NDV-HUJ oncolytic virus in recurrent glioblastoma multiforme. Mol Ther. 2006;13:221–8. doi: 10.1016/j.ymthe.2005.08.016. [DOI] [PubMed] [Google Scholar]
  • 175.Laurie SA, Bell JC, Atkins HL, Roach J, Bamat MK, O’Neil JD, et al. A phase 1 clinical study of intravenous administration of PV701, an oncolytic virus, using two-step desensitization. Clin Cancer Res. 2006;12:2555–62. doi: 10.1158/1078-0432.CCR-05-2038. [DOI] [PubMed] [Google Scholar]
  • 176.Pecora AL, Rizvi N, Cohen GI, Meropol NJ, Sterman D, Marshall JL, et al. Phase I trial of intravenous administration of PV701, an oncolytic virus, in patients with advanced solid cancers. J Clin Oncol. 2002;20:2251–66. doi: 10.1200/JCO.2002.08.042. [DOI] [PubMed] [Google Scholar]
  • 177.Geletneky K, Huesing J, Rommelaere J, Schlehofer JR, Leuchs B, Dahm M, et al. Phase I/IIa study of intratumoral/intracerebral or intravenous/intracerebral administration of Parvovirus H-1 (ParvOryx) in patients with progressive primary or recurrent glioblastoma multiforme: ParvOryx01 protocol. BMC Cancer. 2012;12:99. doi: 10.1186/1471-2407-12-99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Vidal L, Pandha HS, Yap TA, White CL, Twigger K, Vile RG, et al. A phase I study of intravenous oncolytic reovirus type 3 Dearing in patients with advanced cancer. Clin Cancer Res. 2008;14:7127–37. doi: 10.1158/1078-0432.CCR-08-0524. [DOI] [PubMed] [Google Scholar]
  • 179.Karapanagiotou EM, Roulstone V, Twigger K, Ball M, Tanay M, Nutting C, et al. Phase I/II trial of carboplatin and paclitaxel chemotherapy in combination with intravenous oncolytic reovirus in patients with advanced malignancies. Clin Cancer Res. 2012;18:2080–9. doi: 10.1158/1078-0432.CCR-11-2181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Galanis E, Markovic SN, Suman VJ, Nuovo GJ, Vile RG, Kottke TJ, et al. Phase II trial of intravenous administration of Reolysin(®) (Reovirus Serotype-3-dearing Strain) in patients with metastatic melanoma. Mol Ther. 2012;20:1998–2003. doi: 10.1038/mt.2012.146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Morris DG, Feng X, Difrancesco LM, Fonseca K, Forsyth PA, Paterson AH, et al. REO-001: A phase I trial of percutaneous intralesional administration of reovirus type 3 dearing (Reolysin®) in patients with advanced solid tumors. Invest New Drugs. 2012 doi: 10.1007/s10637-012-9865-z. In press. [DOI] [PubMed] [Google Scholar]
  • 182.Hwang TH, Moon A, Burke J, Ribas A, Stephenson J, Breitbach CJ, et al. A mechanistic proof-of-concept clinical trial with JX-594, a targeted multi-mechanistic oncolytic poxvirus, in patients with metastatic melanoma. Mol Ther. 2011;19:1913–22. doi: 10.1038/mt.2011.132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Mastrangelo MJ, Maguire HC, Jr., Eisenlohr LC, Laughlin CE, Monken CE, McCue PA, et al. Intratumoral recombinant GM-CSF-encoding virus as gene therapy in patients with cutaneous melanoma. Cancer Gene Ther. 1999;6:409–22. doi: 10.1038/sj.cgt.7700066. [DOI] [PubMed] [Google Scholar]
  • 184.Park BH, Hwang T, Liu TC, Sze DY, Kim JS, Kwon HC, et al. Use of a targeted oncolytic poxvirus, JX-594, in patients with refractory primary or metastatic liver cancer: a phase I trial. Lancet Oncol. 2008;9:533–42. doi: 10.1016/S1470-2045(08)70107-4. [DOI] [PubMed] [Google Scholar]
  • 185.Liu BL, Robinson M, Han ZQ, Branston RH, English C, Reay P, et al. ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumour properties. Gene Ther. 2003;10:292–303. doi: 10.1038/sj.gt.3301885. [DOI] [PubMed] [Google Scholar]
  • 186.Kelly K, Nawrocki S, Mita A, Coffey M, Giles FJ, Mita M. Reovirus-based therapy for cancer. Expert Opin Biol Ther. 2009;9:817–30. doi: 10.1517/14712590903002039. [DOI] [PubMed] [Google Scholar]
  • 187.Zhang Q, Liang C, Yu YA, Chen N, Dandekar T, Szalay AA. The highly attenuated oncolytic recombinant vaccinia virus GLV-1h68: comparative genomic features and the contribution of F14.5L inactivation. Mol Genet Genomics. 2009;282:417–35. doi: 10.1007/s00438-009-0475-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Zitvogel L, Kroemer G. Targeting PD-1/PD-L1 interactions for cancer immunotherapy. Oncoimmunology. 2012;1:1223–5. doi: 10.4161/onci.21335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Madan RA, Heery CR, Gulley JL. Combination of vaccine and immune checkpoint inhibitor is safe with encouraging clinical activity. Oncoimmunology. 2012;1:1167–8. doi: 10.4161/onci.20591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Skelding KA, Barry RD, Shafren DR. Enhanced oncolysis mediated by Coxsackievirus A21 in combination with doxorubicin hydrochloride. Invest New Drugs. 2012;30:568–81. doi: 10.1007/s10637-010-9614-0. [DOI] [PubMed] [Google Scholar]
  • 191.Kanerva A, Nokisalmi P, Diaconu I, Koski A, Cerullo V, Liikanen I, et al. Anti-viral and anti-tumor T-cell immunity in patients treated with GMCSF coding oncolytic adenovirus. Clin Cancer Res. 2013 doi: 10.1158/1078-0432.CCR-12-2546. In press. [DOI] [PubMed] [Google Scholar]
  • 192.Bauerschmitz GJ, Kanerva A, Wang M, Herrmann I, Shaw DR, Strong TV, et al. Evaluation of a selectively oncolytic adenovirus for local and systemic treatment of cervical cancer. Int J Cancer. 2004;111:303–9. doi: 10.1002/ijc.20217. [DOI] [PubMed] [Google Scholar]
  • 193.Fueyo J, Alemany R, Gomez-Manzano C, Fuller GN, Khan A, Conrad CA, et al. Preclinical characterization of the antiglioma activity of a tropism-enhanced adenovirus targeted to the retinoblastoma pathway. J Natl Cancer Inst. 2003;95:652–60. doi: 10.1093/jnci/95.9.652. [DOI] [PubMed] [Google Scholar]
  • 194.Reddy PS, Burroughs KD, Hales LM, Ganesh S, Jones BH, Idamakanti N, et al. Seneca Valley virus, a systemically deliverable oncolytic picornavirus, and the treatment of neuroendocrine cancers. J Natl Cancer Inst. 2007;99:1623–33. doi: 10.1093/jnci/djm198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Goetz C, Dobrikova E, Shveygert M, Dobrikov M, Gromeier M. Oncolytic poliovirus against malignant glioma. Future Virol. 2011;6:1045–58. doi: 10.2217/fvl.11.76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Msaouel P, Opyrchal M, Domingo Musibay E, Galanis E. Oncolytic measles virus strains as novel anticancer agents. Expert Opin Biol Ther. 2013;13:483–502. doi: 10.1517/14712598.2013.749851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Ramesh N, Ge Y, Ennist DL, Zhu M, Mina M, Ganesh S, et al. CG0070, a conditionally replicating granulocyte macrophage colony-stimulating factor--armed oncolytic adenovirus for the treatment of bladder cancer. Clin Cancer Res. 2006;12:305–13. doi: 10.1158/1078-0432.CCR-05-1059. [DOI] [PubMed] [Google Scholar]
  • 198.Mori I, Liu B, Goshima F, Ito H, Koide N, Yoshida T, et al. HF10, an attenuated herpes simplex virus (HSV) type 1 clone, lacks neuroinvasiveness and protects mice against lethal challenge with HSV types 1 and 2. Microbes Infect. 2005;7:1492–500. doi: 10.1016/j.micinf.2005.05.007. [DOI] [PubMed] [Google Scholar]
  • 199.Takakuwa H, Goshima F, Nozawa N, Yoshikawa T, Kimata H, Nakao A, et al. Oncolytic viral therapy using a spontaneously generated herpes simplex virus type 1 variant for disseminated peritoneal tumor in immunocompetent mice. Arch Virol. 2003;148:813–25. doi: 10.1007/s00705-002-0944-x. [DOI] [PubMed] [Google Scholar]
  • 200.Yaacov B, Eliahoo E, Lazar I, Ben-Shlomo M, Greenbaum I, Panet A, et al. Selective oncolytic effect of an attenuated Newcastle disease virus (NDV-HUJ) in lung tumors. Cancer Gene Ther. 2008;15:795–807. doi: 10.1038/cgt.2008.31. [DOI] [PubMed] [Google Scholar]
  • 201.Grekova SP, Rommelaere J, Raykov Z. Parvoviruses-tools to fine-tune anticancer immune responses. Oncoimmunology. 2012;1:1417–9. doi: 10.4161/onci.21097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Duntsch CD, Zhou Q, Jayakar HR, Weimar JD, Robertson JH, Pfeffer LM, et al. Recombinant vesicular stomatitis virus vectors as oncolytic agents in the treatment of high-grade gliomas in an organotypic brain tissue slice-glioma coculture model. J Neurosurg. 2004;100:1049–59. doi: 10.3171/jns.2004.100.6.1049. [DOI] [PubMed] [Google Scholar]
  • 203.Obuchi M, Fernandez M, Barber GN. Development of recombinant vesicular stomatitis viruses that exploit defects in host defense to augment specific oncolytic activity. J Virol. 2003;77:8843–56. doi: 10.1128/JVI.77.16.8843-8856.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Egilmez NK, Harden JL, Rowswell-Turner RB. Chemoimmunotherapy as long-term maintenance therapy for cancer. Oncoimmunology. 2012;1:563–5. doi: 10.4161/onci.19369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Amiset L, Fend L, Gatard-Scheikl T, Rittner K, Duong V, Rooke R, et al. TLR2 ligation protects effector T cells from regulatory T-cell mediated suppression and repolarizes T helper responses following MVA-based cancer immunotherapy. Oncoimmunology. 2012;1:1271–80. doi: 10.4161/onci.21479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Holcmann M, Drobits B, Sibilia M. How imiquimod licenses plasmacytoid dendritic cells to kill tumors. Oncoimmunology. 2012;1:1661–3. doi: 10.4161/onci.22033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Peng W, Lizée G, Hwu P. Blockade of the PD-1 pathway enhances the efficacy of adoptive cell therapy against cancer. Oncoimmunology. 2013;2:e22691. doi: 10.4161/onci.22691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Rommelfanger DM, Grau MC, Diaz RM, Ilett E, Alvarez-Vallina L, Thompson JM, et al. The efficacy versus toxicity profile of combination virotherapy and TLR immunotherapy highlights the danger of administering TLR agonists to oncolytic virus-treated mice. Mol Ther. 2013;21:348–57. doi: 10.1038/mt.2012.204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Kottke T, Chester J, Ilett E, Thompson J, Diaz R, Coffey M, et al. Precise scheduling of chemotherapy primes VEGF-producing tumors for successful systemic oncolytic virotherapy. Mol Ther. 2011;19:1802–12. doi: 10.1038/mt.2011.147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol. 2013;31:51–72. doi: 10.1146/annurev-immunol-032712-100008. [DOI] [PubMed] [Google Scholar]
  • 211.Martins I, Kepp O, Schlemmer F, Adjemian S, Tailler M, Shen S, et al. Restoration of the immunogenicity of cisplatin-induced cancer cell death by endoplasmic reticulum stress. Oncogene. 2011;30:1147–58. doi: 10.1038/onc.2010.500. [DOI] [PubMed] [Google Scholar]

Articles from Oncoimmunology are provided here courtesy of Taylor & Francis

RESOURCES