Problems with oncolytic viruses




















Of note, the antibody question is complex, and in fact baseline neutralizing antibody titers have not prevented antitumor efficacy in humans [ 76 , 87 ]. The large amounts of virus that are produced by tumors into the bloodstream might simply overcome neutralizing antibodies producible into blood.

In wild type adenovirus infections, only small amounts of adenovirus enter the blood. Neutralizing antibodies are designed to block such virus, not the huge numbers produced by tumors infected with oncolytic virus. Nevertheless, in epidemiological analysis, lack of antibodies at baseline impacted survival statistically significantly [ 76 ], but not in a clinically meaningful manner, as responses and long survival could be seen regardless of baseline antibody titers.

Interestingly, sequential intravenous treatments by changing the virus or serotype might make a difference [ 88 ]. As expression of the Ad5 receptor CXADR appears limiting for efficacy in the context of advanced tumors, fully serotype 3-based oncolytic adenoviruses have been constructed [ 64 ]. This virus enters through non-CXADR-mediated mechanisms including desmoglein 2, which is highly expressed in advanced solid tumors [ 30 , 35 , 64 , 78 ].

Good safety and signs of efficacy were seen in patients treated with a fully serotype 3 adenovirus [ 64 ]. To achieve efficient dissemination of input virus and to minimize virus-related adverse events, adenoviruses have been modified to achieve increased tumor selectivity.

The strategies employed include transcriptional control of adenovirus early proteins such as the E1A or E1B. This blocks adenovirus DNA replication in quiescent normal tissues [ 61 , 66 ]. The fact that tumor cells contain several active oncogenes led to the realization that their resulting proteins could be harnessed to control transcription of adenovirus DNA.

For example, telomerase activity is a known feature of cancer cells, while activity in healthy cells is minimal [ 30 , 91 ]. Therefore, adenovirus replication has been successfully placed under the control of a human telomerase reverse transcriptase hTERT promoter showing antitumor efficacy in advanced cancers [ 64 , 68 ]. Similarly, p53 [ 69 ], carcinoembryonic antigen CEA [ 70 ] and prostate-specific antigen PSA [ 71 ] have been utilized to control expression of early adenovirus proteins.

The nature of viruses allows them to hijack the host cell to produce virus proteins. This allows therapeutic exploitation with the insertion of therapeutic transgenes into the adenovirus genome. Before the recognition of oncolytic adenoviruses as immunotherapy, one of the most common modifications was the insertion of the cytosine deaminase and herpes simplex-derived thymidine kinase [ 74 ]. More recently, the increased recognition of the immune system as an important component in the efficacy of oncolytic viruses led researchers to perceive oncolytic adenoviruses as potent vehicles for immune factors.

Adding a granulocyte macrophage colony stimulating factor GMCSF cytokine transgene into the adenoviral genome is a commonly used modification.

In this approach, virus replication is accompanied by GMCSF production, which results in the recruitment and maturation of dendritic cells DCs , and subsequent priming of T cells with tumor-associated antigens released by oncolysis [ 92 ]. Patient data confirms this notion, with reported increases in peripheral levels of T cells against tumor-associated antigens [ 66 ]. These finding suggest dendritic cell priming in humans as predicted by the established mechanism of action of GMCSF [ 93 ].

However, emerging human data suggests that GMCSF producing viruses might be safe and effective [ 76 , 77 ]. Beyond GMCSF, combined expression of IL and decorin in an oncolytic adenovirus allowed the recovery of antitumor immunity in a poorly immunogenic murine breast cancer model, via cytotoxic T cell infiltration and transforming growth factor beta TGFb reduction [ 82 ].

Coexpression of CD40L and BBL by an oncolytic adenovirus has also shown promising results, due to its ability to promote the destruction of pancreatic tumors, through repolarization of the tumor microenvironment. Such polarization enabled release of T cell attractants and immune stimulatory cytokines, allowing potent antitumor T cell responses [ 96 ].

Also, antibodies can be inserted as transgenes to enhance the efficacy of oncolytic virotherapy. For example, anti-CTLA4, a checkpoint inhibitor, has been successfully inserted in an oncolytic adenovirus platform. Its usage in murine models and ex vivo cultures of cancer patient peripheral blood mononuclear cells PBMCs resulted in increased antitumor activity of T cells [ 81 ]. More recently, dual targeted antibodies targeting T cells and cancer-specific cell surface antigens such as epidermal growth factor receptor EGFR [ 84 ], FR-a [ 85 ], familial adenomatous polyposis FAP [ 10 ] and CD44v6 [ 86 ], have demonstrated promising preclinical results [ 84 ].

Moreover, also other approaches have been studied. These include arming with fusogenic molecules, antibodies, T cell engagers, and ion channels capable of concentrating radioiodine. While in these cases, the transgenes are not necessarily immunologically active, and the oncolytic platform results in immunostimulation. It is important to note that clinical data suggests that oncolytic adenovirus single-agent efficacy has often been somewhat limited.

Several barriers that affect oncolytic adenovirus therapies have been suggested. These include antiviral interferons, which can be produced by the tumor stroma even if the cancer cells themselves lack such ability [ 50 ].

Other reasons include stromal barriers, hypoxia, hyperbaric, necrotic, and acidic areas [ 97 , 98 ]. However, some of these hurdles have been addressed in redesigned adenoviruses conditionally replicating in response to hypoxic factors or acidic tumor microenvironments [ 99 ]. Alternatively, oncolytic adenoviruses have been armed with hyaluronidase [ ], an enzyme that degrades hyaluronic acid which hampers virus dissemination.

Notably, treatment of a number of preclinical in vivo tumor models allowed increased antitumor efficacy. Neutralizing antibodies remain a concern for oncolytic immunotherapies. However, the use of bispecific adapters to retarget antiviral neutralizing antibodies can offer an attractive approach to increase the effectiveness of oncolytic adenovirus therapy [ ]. The coating of oncolytic adenoviruses with tumor derivatives has been reported to allow for successful delivery of particles into the tumor with potent antitumor responses [ ].

A long-term follow-up of these patients has been published [ 5 ]. Treatments were given in the context of an individualized treatment program under the EU Advanced Therapies directive [ 5 ].

While many objective responses were seen, no definite conclusions regarding overall survival benefit can be drawn as no reliable control group was available. However, some case-control analyses that were performed suggest survival benefit [ 88 ]. While taking into account the limitations of nonrandomized data, some interesting findings emerged [ 5 , 51 , 64 , 66 , 80 , 88 , 92 , , , ]. One of the most important observations was that all of the administered viruses appeared quite safe in patients with advanced cancer.

Good tolerability was seen across different serotype viruses, including various capsid modifications, and different immunological arming devices i. Concomitant low-dose cyclophosphamide and temozolomide were also well tolerated [ ]. The former was used to reduce regulatory T cells.

The latter aimed at increased induction of autophagy in infected cancer cells, as this appears to be an important mediator of oncolytic cell death. Moreover, virus replication could be increased by concomitant calcium channel blockage [ ]. No treatment-related patient deaths were observed [ 63 ]. Typical flu-like symptoms, such as fever and fatigue, were observed in most patients a few days after treatment.

These findings were confirmed in multiple subsequent clinical trials [ 61 , 94 , ]. Flu-like symptoms and fever could be effectively reduced with acetaminophen paracetamol. Biodistribution studies done with adenoviruses in rodents are unreliable as most animals lack entry receptors or their organ distribution is different from humans.

From ATAP patients, we have been able to collect important information about virus biodistribution in humans [ 63 ]. Many humans have neutralizing antibodies against different adenovirus serotypes, although often at low baseline titer [ 64 ]. However, as hundreds of billions of viruses are given in a typical treatment, pre-existing antibodies may be unable to completely block intravenous delivery.

In subsequent intravenous treatments with the same serotype, the situation is more unclear, which is one of the reasons intratumoral delivery is commonly used with oncolytic viruses. Of note, it has been proposed that antiviral immunity helps generate antitumor immunity [ 66 ]. It has been established in humans that adenovirus is able to travel through blood to metastases despite neutralizing antibodies [ 63 ].

For some viruses, the mechanism appears to relate to binding to blood cells [ ]. Interestingly, adenoviruses in blood qPCR data were most often found from blood clots, while some patients had significant amount of virus in the serum compartment [ 64 ].

Treatment responses or long survival are seen regardless of neutralizing antibody titers, although it should be noted that most patients were treated intratumorally [ 76 ]. Interestingly, we treated seven patients with the serotype 3 adenovirus using only intravenous administration.

This was seen also with patients who had pre-existing antibodies against the virus. Also, 5 of the 6 evaluable patients showed signs of possible benefit. This data indicates that viruses might be able to enter tumors also via the intravenous route [ 64 ]. This was later confirmed in an autopsy study where non-injected tumors were shown to have oncolytic adenovirus [ 63 ].

From a clinical standpoint, the use of viruses for cancer treatment in the modern era is in its infancy. Initially, wild type viruses were used, but this approach could result in adverse events caused by virus replication in normal tissues.

Nevertheless, Rigvir an ECHO-7 virus [ ], an oncolytic picornavirus with some innate tumor selectivity, was the first approved oncolytic virotherapy product for cancer approved in Latvia and later in a few other countries. The second oncolytic virus was rationally designed for tumor selectivity. Named H Oncorine , this adenovirus has been used in China since for the treatments of solid tumors [ ].

Of note, both of these viruses lack arming devices. The acknowledgement that repurposing the immune system to exert antitumor functions could provide a promising approach to treat cancer enabled scientists to employ the immunological capabilities of oncolytic viruses [ ]. This landmark approval in Western countries encouraged optimism in the medical community to continue developing and improving oncolytic viruses for cancer therapy, including adenoviruses. As expected, a high presence of cytotoxic T cell infiltration was observed in the tumors following treatment [ ].

Of note, and in contrast to combinations of checkpoint inhibitors [ ], adverse events were not compounded. This suggests that oncolytic viruses can be combined with checkpoint inhibition without a problematic decrease in safety. At the time of writing this review March , we did a search on clinicaltrials. Sixteen different oncolytic viruses were used in these completed or recruiting trials.

No active phase III clinical trials were found. Sixteen oncolytic adenoviruses used in phase I—II trials that have been completed or recruiting Mar clinicaltrials. It was designed based on human data from ATAP. Observations of hematological cancer regressions following virus infection were seen early last century [ 17 , 18 ]. Of note, response evaluation was possible for hematological malignancies, by microscopy and cell counting, while this was not true for most solid tumors at the time.

This might have caused overrepresentation of hematological cancer in early literature, due to observational bias. However, it cannot be denied that many reports suggested regression of hematological tumors after viral infections. As such, there is strong rationale to believe that oncolytic viruses could be used also in hematological malignancies. However, not all viruses are suitable for treatment of blood cell tumors.

For example, adenovirus does not appear to be able to lyse white blood cells [ ]. According to recent publications, growing interest towards oncolytic viruses is present also in the hematological field, as several viruses are being studied in preclinical settings [ ]. However, only a few trials have been published [ ]. A search from clinicaltrials. To conclude, to date, there have been few trials with oncolytic viruses in hematological cancers. Some noteworthy efforts include early phase trials with reovirus [ ], measles [ ], and vesicular stomatitis virus VSV in multiple myeloma [ ].

No hematological trials with oncolytic adenovirus were found. Arming with immunostimulatory cytokines has been one popular method to generate immunological synergy with the effects of oncolysis.

Clinical benefit of this approach was seen in the phase III OPTiM trial where 1 in 6 patients achieved complete responses with the oncolytic virus talimogene laherparepvec. Combining oncolytic viruses with T cell activating checkpoint inhibition can improve antitumor efficacy of oncolytic adenovirus therapy. Clinical proof-of-concept for the efficacy of combining oncolytic viruses with checkpoint inhibitors has been presented [ , ].

Furthermore, the safety profile of these treatments has been good and oncolytic viruses do not seem to increase the rate of serious adverse events. Although these combinations seem to yield increased potency and long-term benefits to some patients, not all benefit, and there is clearly a role for further improvement. Combining chemotherapy or radiation therapy in a rational way to improve treatment benefits, and even these conventional therapies seem to have an immunological component [ 27 , ].

However, clear clinical proof in support of this approach is currently limited. Combining other therapies such as adoptive cell therapy or targeted therapies might also result in better treatments. However, we are still lacking much information about the immune effects in individual tumors. This insufficiency of knowledge makes it hard to understand which patients would benefit most of what kind of treatment combinations.

This could be the greatest challenge in the field at the moment. Classic trial design is not well suited for understanding mechanisms on an individual tumor and patient level. An ideal cancer treatment should be so good that most patients would clearly benefit while side effects should be tolerable. Our own contribution to this quest is a novel oncolytic adenovirus, designed specifically with T cells in mind.

Utilizing tumor hypoxia to enhance oncolytic viral therapy in colorectal metastases. A hypoxia-and telomerase-responsive oncolytic adenovirus expressing secretable trimeric TRAIL triggers tumour-specific apoptosis and promotes viral dispersion in TRAIL-resistant glioblastoma.

Vojtechova Z. Davola M. Kaufman H. Oncolytic viruses: A new class of immunotherapy drugs. Drug Discov. Sokolowski N. Oncolytic virotherapy using herpes simplex virus: How far have we come? Shen B. Effect of hypoxia on Ad5 infection, transgene expression and replication.

Maroun J. Designing and building oncolytic viruses. Future Virol. Badrinath N. Viruses as nanomedicine for cancer. Marchini A. Tischer B. Herpesviruses—A zoonotic threat? Franci G. Haarr L. The herpes simplex virus type 1 particle: Structure and molecular functions.

Review article. Stelitano D. Amino Acids, Peptides and Proteins. HSV membrane glycoproteins, their function in viral entry and their use in vaccine studies; pp. Singh M. Coffin R. Liu B. Andtbacka R. Eissa I. Johnson D. Talimogene laherparepvec T-VEC for the treatment of advanced melanoma. Masoud S. Russell L. The emerging role of oncolytic virus therapy against cancer. Ahn D. Thomas S. Development of a new fusion-enhanced oncolytic immunotherapy platform based on herpes simplex virus type 1.

Senzer N. Phase II clinical trial of a granulocyte-macrophage colony-stimulating factor-encoding, second-generation oncolytic herpesvirus in patients with unresectable metastatic melanoma. Oncolytic herpes simplex virus vector g47delta in combination with androgen ablation for the treatment of human prostate adenocarcinoma. Mineta T. Foreman P. Oncolytic Virotherapy for the Treatment of Malignant Glioma. Kambara H. Saha B. The adenovirus genome contributes to the structural stability of the virion.

Mangel W. Structure, function and dynamics in adenovirus maturation. Harada J. Ries S. Garber K. Cancer Inst. Ganly I. Nemunaitis J. Makower D. Khuri F. Nwanegbo E. Prevalence of neutralizing antibodies to adenoviral serotypes 5 and 35 in the adult populations of The Gambia, South Africa, and the United States. Diagn Lab. Peter M. Oncolytic Adenovirus in Cancer Immunotherapy.

Philbrick B. Lang F. Green T. Structure of the vesicular stomatitis virus nucleoprotein-RNA complex. Stojdl D. VSV strains with defects in their ability to shutdown innate immunity are potent systemic anti-cancer agents. Cancer Cell. Sahin E. Development of oncolytic reovirus for cancer therapy. Cancer Ther. Gong J. Activated ras signaling pathways and reovirus oncolysis: An update on the mechanism of preferential reovirus replication in cancer cells.

Errington F. Reovirus activates human dendritic cells to promote innate antitumor immunity. Kaufmann B. The structure of human parvovirus B EMBO Mol. Nuesch J. Molecular pathways: Rodent parvoviruses--mechanisms of oncolysis and prospects for clinical cancer treatment. Hristov G. Josupeit R. Palgen J. Unity in diversity: Shared mechanism of entry among paramyxoviruses. Aguilar H. Paramyxovirus Glycoproteins and the Membrane Fusion Process.

Chatziandreou N. Relationships and host range of human, canine, simian and porcine isolates of simian virus 5 parainfluenza virus 5 J. Fox C. Wansley E. Foloppe J. Heinrich B. Immunogenicity of oncolytic vaccinia viruses JX-GFP and TG in a human melanoma in vitro model: Studying immunogenic cell death, dendritic cell maturation and interaction with cytotoxic T lymphocytes.

Onco Targets Ther. Lauer U. Minev B. First-in-human study of TK-positive oncolytic vaccinia virus delivered by adipose stromal vascular fraction cells.

Donina S. Melanoma Res. Ismailov Z. Case Rep. Cell Dev. Verza F. Milazzo G. Johnstone R. Histone-deacetylase inhibitors: Novel drugs for the treatment of cancer. Mariadason J. Park J. Epigenetic modulation with histone deacetylase inhibitors in combination with immunotherapy. Secrist J. HDAC inhibitors for the treatment of cancer.

Nehme Z. Control of viral infections by epigenetic-targeted therapy. Zhao C. Sanaei M. Hassell K. Histone Deacetylases and their Inhibitors in Cancer Epigenetics.

Abbas A. The role of histone deacetylases in prostate cancer. Bubna A. Vorinostat-An Overview. Indian J. Bali P. Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein A novel basis for antileukemia activity of histone deacetylase inhibitors.

Rivero-Cruz J. Cytotoxic constituents of the twigs of Simarouba glauca collected from a plot in Southern Florida. Rikiishi H. Autophagic and apoptotic effects of HDAC inhibitors on cancer cells. Chinnaiyan P. Modulation of radiation response by histone deacetylase inhibition. Munster P. The histone deacetylase inhibitor suberoylanilide hydroxamic acid induces differentiation of human breast cancer cells. Kim H. Histone deacetylase inhibitors: Molecular mechanisms of action and clinical trials as anti-cancer drugs.

Vanhaecke T. Trichostatin A-like hydroxamate histone deacetylase inhibitors as therapeutic agents: Toxicological point of view. Damaskos C. Wang G. Kuefer R. Sodium butyrate and tributyrin induce in vivo growth inhibition and apoptosis in human prostate cancer. Xiong F. Inhibition of mouse B16 melanoma by sodium butyrate correlated to tumor associated macrophages differentiation suppression. Fortunati N. Valproic acid is a selective antiproliferative agent in estrogen-sensitive breast cancer cells.

Cancer Lett. Jennings V. Nakashima H. Combining HDAC inhibitors with oncolytic virotherapy for cancer therapy. Otsuki A. Histone deacetylase inhibitors augment antitumor efficacy of herpes-based oncolytic viruses.

Connolly R. Entinostat: A promising treatment option for patients with advanced breast cancer. Future Oncol. Schech A. Histone deacetylase inhibitors: Molecular mechanisms of action.

Barbarotta L. Suraweera A. Furumai R. FK depsipeptide as a natural prodrug that inhibits class I histone deacetylases. Marrone A. A Narrative Review. Ruf B. Combination of the oral histone deacetylase inhibitor resminostat with oncolytic measles vaccine virus as a new option for epi-virotherapeutic treatment of hepatocellular carcinoma. Soukupova J. Resminostat induces changes in epithelial plasticity of hepatocellular carcinoma cells and sensitizes them to sorafenib-induced apoptosis.

Katsura T. The effects of trichostatin A on the oncolytic ability of herpes simplex virus for oral squamous cell carcinoma cells. Nguyen T. Oncolytic viruses and histone deacetylase inhibitors--a multi-pronged strategy to target tumor cells. Cytokine Growth Factor Rev. Liu T. Trichostatin A and oncolytic HSV combination therapy shows enhanced antitumoral and antiangiogenic effects.

Kim D. Combination therapy of conditionally replicating adenovirus and histone deacetylase inhibitors. Coxsackievirus and adenovirus receptor promotes antitumor activity of oncolytic adenovirus H in esophageal cancer.

Histone deacetylase inhibitor trichostatin A enhances the antitumor effect of the oncolytic adenovirus H on esophageal squamous cell carcinoma in vitro and in vivo. Kitazono M. Enhanced adenovirus transgene expression in malignant cells treated with the histone deacetylase inhibitor FR Han X. Synergistic combination of histone deacetylase inhibitor suberoylanilide hydroxamic acid and oncolytic adenovirus ZDTRAIL as a therapy against cervical cancer. Pei Z. Chemical targeting of the innate antiviral response by histone deacetylase inhibitors renders refractory cancers sensitive to viral oncolysis.

Shulak L. Histone deacetylase inhibitors potentiate vesicular stomatitis virus oncolysis in prostate cancer cells by modulating NF-kappaB-dependent autophagy. Eckschlager T. Histone Deacetylase Inhibitors as Anticancer Drugs. Thurn K. Rational therapeutic combinations with histone deacetylase inhibitors for the treatment of cancer. Michan S. Sirtuins in mammals: Insights into their biological function. Muscolini M. Kapoor P.

Impact of risk stratification on outcome among patients with multiple myeloma receiving initial therapy with lenalidomide and dexamethasone. Cai W. PMP22 regulates self-renewal and chemoresistance of gastric cancer cells. Jaime-Ramirez A. Bretscher C. Cold Spring Harb. Funding This research received no external funding. Conflicts of Interest The authors declare no conflict of interest.

References 1. Palumbo A. Persona E. Lenalidomide treatment for patients with multiple myeloma: Diagnosis and management of most frequent adverse events. Laubach J. Bortezomib in the management of multiple myeloma.

Cancer Manag. Sharma S. Panobinostat for the treatment of multiple myeloma: The evidence to date. Blood Med. Dima D. Frampton J. Starr P.

Health Drug Benefits. Munshi N. Morgan G. The genetic architecture of multiple myeloma. Bluming A. Taqi A. Kawa A. The effect of attenuated vaccinia virus AS strain on multiple myeloma; a case report. Conry R.

Talimogene laherparepvec: First in class oncolytic virotherapy. Vaccines Immunother. Hanahan D. The hallmarks of cancer. Oliva S. Systemic virotherapy for multiple myeloma. Expert Opin. Morris D. New Drugs. Gollamudi R. Kolb E. Blood Cancer. Guo Z. Bartee E. Potential of oncolytic viruses in the treatment of multiple myeloma. Oncolytic Virother. Sborov D. Cancer Res. Teoh G. Adenovirus vector-based purging of multiple myeloma cells. Fernandes M.

Senac J. Infection and killing of multiple myeloma by adenoviruses. Gene Ther. Drouin M. Lyle C. Integrin alphavbeta5 is a primary receptor for adenovirus in CAR-negative cells. Lavery D. Productive infection of cultured human lymphoid cells by adenovirus. Adenovirus E1A and E1B genes are regulated posttranscriptionally in human lymphoid cells. Wenthe J. Cancer Gene Ther. Zhang L. A novel immunocompetent murine model for replicating oncolytic ade-noviral therapy.

Ginsberg H. A mouse model for investigating the molecular pathogenesis of adenovirus pneumonia. Duncan S. Infection of mouse liver by human adeno-virus type 5. Deng H. Oncolytic virotherapy for multiple myeloma using a tumour-specific double-deleted vaccinia virus. Lei W. Combined expression of miRa and Smac mediated by oncolytic vaccinia virus synergistically promote anti-tumor effects in Multiple Myeloma. Xie S. Beclin1-armed oncolytic Vaccinia virus enhances the therapeutic efficacy of R-CHOP against lymphoma in vitro and in vivo.

Bartee M. Lymphoma Myeloma Leuk. Myxoma virus attenuates expression of activating transcription factor 4 ATF4 which has implications for the treatment of proteasome inhibitor—resistant multiple myeloma. Lilly C. Villa N. Ghose J. Oncolytic herpes simplex virus infects myeloma cells in vitro and in vivo. Oku M. Oncolytic herpes simplex virus type 1 HSV-1 in combi-nation with lenalidomide for plasma cell neoplasms.

Thirukkumaran C. Reovirus modulates autophagy during oncolysis of multiple myeloma. Reovirus as a successful ex vivo purging modality for multiple myeloma. Acta Med Scand Suppl. ONYX, an E1B gene-attenuated adenovirus, causes tumor-specific cytolysis and antitumoral efficacy that can be augmented by standard chemotherapeutic agents.

Nat Med. Ai Zheng. Garber K. J Natl Cancer Inst. Coffin R. Interview with Robert Coffin, inventor of T-VEC: the first oncolytic immunotherapy approved for the treatment of cancer.

Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J Clin Oncol. Oncolytic herpes virus with defective ICP6 specifically replicates in quiescent cells with homozygous genetic mutations in p Reovirus therapy of tumors with activated Ras pathway. Exploiting tumor-specific defects in the interferon pathway with a previously unknown oncolytic virus. Oncolytic vaccinia virus for the treatment of cancer. Expert Opin Biol Ther. Double-faceted mechanism of parvoviral oncosuppression.

Curr Opin Virol. In vivo imaging and tumor therapy with the sodium iodide symporter. J Cell Biochem. Weissleder R. Molecular imaging in cancer. Theranostic potential of oncolytic vaccinia virus. Oncolytic measles virus expressing the sodium iodide symporter to treat drug-resistant ovarian cancer.

Cancer Res. Phase I study of noninvasive imaging of adenovirus-mediated gene expression in the human prostate. Oncolytic vaccinia virus expressing the human somatostatin receptor SSTR2: molecular imaging after systemic delivery using In-pentetreotide. Oncolytic adenoviruses armed with thymidine kinase can be traced by PET imaging and show potent antitumoural effects by ganciclovir dosing.

PLoS One. Positron emission tomography-based imaging of transgene expression mediated by replication-conditional, oncolytic herpes simplex virus type 1 mutant vectors in vivo.

Radiovirotherapy: principles and prospects in oncology. Curr Pharm Des. Molecular imaging with bioluminescence and PET reveals viral oncolysis kinetics and tumor viability. Vaccinia virus GLV-1h is a novel agent for detection and effective local control of positive surgical margins for breast cancer.

Breast Cancer Res. Feasibility of adenovirus-mediated hNIS gene transfer and I radioiodine therapy as a definitive treatment for localized prostate cancer. A novel oncolytic viral therapy and imaging technique for gastric cancer using a genetically engineered vaccinia virus carrying the human sodium iodide symporter.

J Exp Clin Cancer Res. Doxycycline inducible melanogenic vaccinia virus as theranostic anti-cancer agent. Adapted ECHO-7 virus Rigvir immunotherapy oncolytic virotherapy prolongs survival in melanoma patients after surgical excision of the tumour in a retrospective study. Melanoma Res. Oncolytic virotherapy including Rigvir and standard therapies in malignant melanoma. Oncolytic Virother.

Liang M. Oncorine, the world first oncolytic virus medicine and its update in China. Curr Cancer Drug Targets. Ries S, Korn WM.

ONYX mechanisms of action and clinical potential of a replication-selective adenovirus. J Virol. The early region 1B kilodalton oncoprotein of adenovirus relieves growth restrictions imposed on viral replication by the cell cycle. Going viral: a review of replication-selective oncolytic adenoviruses. Seroprevalence of neutralizing antibodies to human adenoviruses type-5 and type and chimpanzee adenovirus type in healthy Chinese adults.

J Med Virol. Prevalence of neutralizing antibodies to adenoviral serotypes 5 and 35 in the adult populations of The Gambia, South Africa, and the United States. Clin Diagn Lab Immunol. Genome sequence of herpes simplex virus 1 strain KOS. Experimental therapy of human glioma by means of a genetically engineered virus mutant.

Mohr I, Gluzman Y. A herpesvirus genetic element which affects translation in the absence of the viral GADD34 function. EMBO J. Into the clinic: talimogene laherparepvec T-VEC , a first-in-class intratumoral oncolytic viral therapy. J Immunother Cancer. Intratumoral injection of HSV, an oncolytic herpes virus, is safe and shows evidence of immune response and viral replication in young cancer patients. Clin Cancer Res. A phase 1 trial of oncolytic HSV-1, G, given in combination with radiation for recurrent GBM demonstrates safety and radiographic responses.

Hum Gene Ther. Genomic signature of the natural oncolytic herpes simplex virus HF10 and its therapeutic role in preclinical and clinical trials. Ann Surg Oncol.

Gene Ther. Oncolytic virotherapy promotes intratumoral T cell infiltration and improves Anti-PD-1 immunotherapy. Randomized, open-label phase II study evaluating the efficacy and safety of talimogene laherparepvec in combination with ipilimumab versus ipilimumab alone in patients with advanced, unresectable melanoma.

The adenovirus capsid: major progress in minor proteins. J Gen Virol. Components of adenovirus genome packaging. Front Microbiol. Vesicular stomatitis virus: re-inventing the bullet. Trends Mol Med. A phase I study of telomerase-specific replication competent oncolytic adenovirus telomelysin for various solid tumors. An open label, single-arm, phase II multicenter study of the safety and efficacy of CG oncolytic vector regimen in patients with BCG-unresponsive non-muscle-invasive bladder cancer: interim results.

Urol Oncol. Conserved fiber-penton base interaction revealed by nearly atomic resolution cryo-electron microscopy of the structure of adenovirus provides insight into receptor interaction. Zhang Y, Bergelson JM.

Adenovirus receptors. Multiple adenovirus serotypes use alpha v integrins for infection. Arnberg N. Adenovirus receptors: implications for targeting of viral vectors. Trends Pharmacol Sci. Preclinical characterization of the antiglioma activity of a tropism-enhanced adenovirus targeted to the retinoblastoma pathway. Int J Cancer. The oncolytic adenovirus VCN as therapeutic approach against pediatric osteosarcoma. Replacement of adenovirus type 5 fiber shaft heparan sulfate proteoglycan-binding domain with RGD for improved tumor infectivity and targeting.

Oncolytic adenovirus research evolution: from cell-cycle checkpoints to immune checkpoints. Phase 1b trial of biweekly intravenous Pexa-Vec JX , an oncolytic and immunotherapeutic vaccinia virus in colorectal cancer. Seroprevalence of herpes simplex virus types 1 and 2—United States, J Infect Dis. Systemic therapy of malignant human melanoma tumors by a common cold-producing enterovirus, coxsackievirus a Type B coxsackieviruses and their interactions with the innate and adaptive immune systems.

Future Microbiol. A retroviral replicating vector encoding cytosine deaminase and 5-FC induces immune memory in metastatic colorectal cancer models. Mol Ther Oncolytics. Design and selection of Toca for clinical use: modified retroviral replicating vector with improved stability and gene expression. Toca plus 5-fluorocytosine in combination with lomustine shows chemotoxic and immunotherapeutic activity with no additive toxicity in rodent glioblastoma models.

Neuro Oncol. Going viral with cancer immunotherapy. Nat Rev Cancer. Viruses as anticancer drugs. The case of oncolytic viruses versus the immune system: waiting on the judgment of Solomon. Oncolytic viruses. Adv Virol.



0コメント

  • 1000 / 1000