INTRODUCTION
Glioblastoma multiforme (GBM) represents nearly half of all primary malignant brain tumors in adults, and malignant gliomas are a leading cause of cancer-related morbidity and mortality in children.1–3 Outcomes for patients with GBM are poor, and effective treatment options are limited with individuals having a median survival of approximately 15 months.2,4 The current treatment protocol focuses on maximal safe resection, radiotherapy, and concurrent tumor-treating fields/chemotherapy with temozolomide (TMZ) with only a modest effect on outcomes.4–8 There are multiple factors that contribute to treatment resistance and recurrence of GBM. It is highly invasive, with glioma cells spreading widely within normal brain tissue at early stages.9–11 GBMs contain tumorigenic glioma stem cells that contribute to tumor initiation, therapeutic resistance, and recurrence.12 GBM also exhibits both intertumoral and intratumoral heterogeneity, which contributes to diagnostic complexity and limits the application of personalized, targeted therapies.12
There is a substantial need for novel therapeutic approaches that address several of these challenges. Immunovirotherapy has emerged as a targeted approach for treatment of GBM and other malignant gliomas with promising results.5,13,14 Multiple viral vectors have been genetically altered and developed as oncolytic viruses and for targeted drug delivery. There are currently several ongoing clinical trials for the treatment of GBM with immunovirotherapy.12,15,16 In this review, we discuss the recent advances and current state of viral vectors developed for the targeted treatment of GBM and malignant gliomas including their mechanism of action and clinical applications.
HUMAN ONCOLYTIC VIRUS MODELS
Adenovirus
Adenovirus (Adv) is a double-stranded nonenveloped DNA virus causing mild upper respiratory symptoms in humans that typically self-resolve. Within the realm of immunovirotherapy, recombinants of Adv that show conditional replication are some of the most studied oncolytic viruses.16,17 The key to the multiple immunovirotherapy applications of the oncolytic Adv comes from its E1A gene, which is essential in its replication and is the first gene expressed on viral infection.18 The Ki67 promoter for E1A expression can be upregulated in conjunction with arming the oncolytic Adv with interleukin (IL)-15 gene expression against GBM cells with resultant enhanced anti-GBM efficacy via activation of microglial cells.18 Adenovirus can also be used to deliver suicide gene therapy.19 These suicide genes have successfully induced apoptosis via conversion of the prodrug 5-FC into 5-fluorouracil in the presence of Escherichia coli cytosine deaminase (CD) and have encoded proteins that terminate protein synthesis within tumor cells.19 Adenovirus, therefore, represents a multifaceted vector in the immunovirotherapy arsenal against GBM.
In 2018, Lang and colleagues20 published landmark results from a Phase I, dose-escalation, biologic-end-point study investigating Delta-24-RGD oncolytic virus. Participants were separated into 2 groups, with group A receiving a single intratumoral injection of the virus into biopsy-confirmed recurrent tumor and group B undergoing intratumoral injection through an implanted catheter followed by en bloc resection days postimplantation to evaluate posttreatment specimens. The study demonstrated quite promising clinical results, with 20% of group A patients surviving more than 3 years posttreatment and 12% of patients demonstrating greater than 95% enhancing tumor reduction with associated more than 3 years of progression-free survival. Analysis of group B specimens postresection demonstrated direct virus-induced oncolysis with tumor infiltration by CD8 cells. Subsequent analyses of cell lines derived from these patients showed induction of immunogenic cell death after virus insertion into tumor cells. Overall, this Phase I study provided promising results demonstrating increased long-term survival in patients with recurrent high-grade gliomas due to the direct oncolytic effects of DNX-2401 adenovirus.20
A promising study recently published in Neuro-Oncology Advances found potentiating effects of the Adv Delta24-RGD on the response of a murine GBM model to anti-PD1 therapy overcoming tumor-induced immune suppression via significant recruitment of dendritic cells resulting in a robust antitumor response and survival benefit, suggesting the potential benefit of combination therapy.21,22 Other mechanisms of action affect the function of T cells, specifically decreasing tumor-infiltrating T regulatory (Treg) cells and increasing interferon-gamma producing CD8 T cells. In addition, the oncolytic AdCMVdelta24 virus can augment systemic tumor antigen specific T cells and reprogram Treg cells to a stimulatory rather than immunosuppressive state.23
Reduced expression in immortalized cells/Dickkopf-3 (REIC/Dkk-3) is a tumor suppressor and therapeutic gene in many human cancers, including malignant glioma with promising results with adenovirus oncolytic therapy.24 An adenovirus REIC vector was developed to increase REIC/Dkk-3 expression (Ad-SGE-REIC), which is currently undergoing a Phase I/IIa clinical trial for treatment of recurrent malignant glioma.25
Not only can the Adv vector be used to stimulate the antitumor immune response, but it also has possible applications to enhance intraoperative discernment of tumor tissue from normal brain. In 2015, Yano and colleagues26 reported the successful use of a green fluorescent protein expressing adenovirus OBP-401 to label GBM cells to allow fluorescence guided surgery techniques to resect the murine GBM with nearly undetectable residual macroscopic tumor in the surgical bed.
Herpes Simplex Virus Type-1
Genetically engineered oncolytic Herpes Simplex Virus type 1 (oHSV), in particular, has been the focus of extensive preclinical and clinical research, offering several advantages as a therapeutic vector.14 It is an enveloped icosahedral virus with double-stranded linear DNA that belongs to the Herpesviridae family. It is intrinsically neurotropic and does not integrate into the host cell DNA, making it an ideal vector for targeting primary brain tumors.27,28 The deletion of essential genes required for replication in normal cells in combination with replacement of nonessential genes with foreign DNA can provide therapeutic advantages.14,28 In addition, engineered oHSVs remain sensitive to antivirals, which contributes to its safety profile in the event of unanticipated adverse reactions.
The introduction of inactivating mutations in the γ134.5 neurovirulence gene, an essential gene for viral replication in normal cells in the central nervous system, has been extensively used in oncolytic viral models.29,30 In response to herpes simplex virus (HSV)-1 infection, normal cells activate the double-stranded RNA–dependent protein kinase R (PKR) system. This leads to phosphorylation of eukaryotic initiation factor (eIF) 2α inducing translational arrest and resulting in severe impairment of viral protein synthesis.29 Infected cell protein 34.5 (ICP34.5), the product of γ134.5, reverses this process and is thus essential for successful viral replication in the central nervous system. Deletion of γ134.5 results in conditional viral replication within tumor cells that have low intrinsic PKR activity, such as human glioma.5,29,30 This prevents productive infection in normal cells in the brain through PKR-mediated translational arrest while still maintaining oncolytic activity against glioma cells, which have defective signaling pathways and/or activating RAS mutations that suppress antiviral responses.5,29,30 Clinical trials of γ134.5-deleted oHSV G207 (Table 1) have demonstrated safety with evidence of efficacy in both adults and children (Table 2).14,31–37 Markert and colleagues32 conducted a phase I trial on 21 adult patients and demonstrated safety at doses up to 3 × 109 pfu with 9 patients showing evidence of neuropathologic or radiographic response. A follow-up phase 1b trial on 6 patients with recurrent GBM receiving 2 doses of G207 totaling 1.15 × 109 pfu, with 13% of this total dose injected before tumor resection via a catheter placed stereotactically into enhancing portion of the tumor, also demonstrated safety and confirmed viral replication.34 A third study demonstrated safety of vG207 in combination a single 5 Gy radiation dose in 9 adults with recurrent high-grade gliomas to provide in vivo synergistic viral replication based on preclinical data.33 A clinical trial in pediatric supratentorial HGG trial is now complete and demonstrated safety of a controlled-rate infusion of intratumoral G207 up to 1 × 108 pfu (maximum planned dose) alone and combined with 5 Gy of radiation. Radiographic, neuropathologic, and/or clinical responses were seen in 11 of 12 patients. Matched pretreatment and posttreatment tissue in several patients demonstrated marked increase in tumor-infiltrating lymphocyte months after treatment with G207 (data not yet published).31 A first-in-human trial assessing the safety of G207 alone and combined with 5 Gy of radiation in malignant cerebellar tumors, including malignant gliomas, is currently ongoing.37
Table 1.
Virus | GBM Type | Study Title | Phase | Biological | n | Duration | NCT Number and Reference | Status |
---|---|---|---|---|---|---|---|---|
Adenovirus | Recurrent | DNX-2401 (Formerly Known as Delta-24-RGD-4C) for Recurrent Malignant Gliomas | Phase I | DNX-2401 | 37 | February 2009–February 2015 | NCT00805376 20 | Completed |
Recurrent | Safety Study of Replication-competent Adenovirus (Delta-24-RGD) in Patients With Recurrent Glioblastoma | Phase I-II | DNX-2401 | 20 | June 2010–December 2014 | NCT01582516 | Completed | |
Recurrent | Virus DNX2401 and Temozolomide in Recurrent Glioblastoma | Phase I | DNX2401 | 31 | September 2013–March 2017 | NCT01956734 | Completed | |
Recurrent | DNX-2401 With Interferon Gamma (IFN-γ) for Recurrent Glioblastoma or Gliosarcoma Brain Tumors | Phase I | DNX-2401 | 37 | September 11, 2014–March 15, 2018 | NCT02197169 | Completed | |
Recurrent | Combination Adenovirus + Pembrolizumab to Trigger Immune Virus Effects | Phase II | DNX-2401 | 49 | June 2016–June 2021 | NCT02798406 | Active, not recruiting | |
Recurrent | DNX-2440 Oncolytic Adenovirus for Recurrent Glioblastoma | Phase I | DNX-2440 | 24 | October 16, 2018–October 16, 2022 | NCT03714334 | Recruiting | |
Recurrent | Oncolytic Adenovirus DNX-2401 in Treating Patients With Recurrent High-Grade Glioma | Phase I | DNX-2401 | 36 | February 12, 2019–May 31, 2022 | NCT03896568 | Recruiting | |
Herpes | Recurrent | Conditionally replicating herpes simplex virus mutant, G207 for the treatment of malignant glioma | Phase I | G207 | 21 | February 1998–May 1999 | NCT00036699 32 | Completed |
Recurrent | Phase Ib trial of mutant herpes simplex virus G207 inoculated pre-and post-tumor resection for recurrent GBM | Phase Ib | G207 | 6 | January 2002–August 2003 | NCT00028158 34 | Completed | |
Recurrent | G207 Followed by Radiation Therapy in Malignant Glioma | Phase I | G207 | 9 | May 2005–December 2008 | NCT00157703 33 | Completed | |
Recurrent | Oncolytic HSV-1716 in Treating Younger Patients With Refractory or Recurrent High-Grade Glioma That Can Be Removed By Surgery | Phase I | HSV-1716 | 2 | December 2013–May 2016 | NCT02031965 | Terminated | |
Recurrent | Genetically Engineered HSV-1 Phase 1 Study for the Treatment of Recurrent Malignant Glioma | Phase I | M032 (NSC 733972) | 15 of 26 | September 2014–September 2023 | NCT02062827 | Recruiting | |
Recurrent | HSV G207 Alone or With a Single Radiation Dose in Children With Progressive or Recurrent Supratentorial Brain Tumors | Phase I | G207 | 12 | May 2016–April 2021 | NCT02457845 | Active, not recruiting | |
Recurrent | A Study of the Treatment of Recurrent Malignant Glioma With rQNestin34.5v.2 | Phase I | rQNestin34.5v.2 | 108 | July 18, 2017–July 2022 | NCT03152318 | Recruiting | |
Recurrent | HSV G207 in Children With Recurrent or Refractory Cerebellar Brain Tumors | Phase I | G207 | 15 | September 12, 2019–September 1, 2024 | NCT03911388 | Recruiting | |
Recurrent | Trial of C134 in Patients With Recurrent GBM | Phase I | C134 | 24 | September 23, 2019–September 2024 | NCT03657576 | Active, not recruiting | |
Recurrent | HSV G207 With a Single Radiation Dose in Children With Recurrent High-Grade Glioma | Phase II | G207 | 30 | October 1, 2020–October 1, 2024 | NCT04482933 | Not yet recruiting | |
Measles | Recurrent | Viral Therapy in Treating Patients With Recurrent Glioblastoma Multiforme | Phase I | MV-CEA | 23 | October 23, 2006–November 30, 2019 | NCT00390299 | Completed, results not published yet |
NDV | Recurrent | New Castle Disease Virus (NDV) in Glioblastoma Multiforme (GBM), Sarcoma and Neuroblastoma | Phase I-II | HUJ | 0 | July 2011–July 2011 | NCT01174537 | Withdrawn |
Polio | Recurrent | PVSRIPO for Recurrent Glioblastoma (GBM) | Phase I | PVSRIPO | 61 | April 25, 2012–June 2021 | NCT01491893 75 | Active, not recruiting |
Recurrent | PVSRIPO in Recurrent Malignant Glioma | Phase II | PVSRIPO | 122 | June 1, 2017–December 2023 | NCT02986178 | Active, not recruiting | |
Recurrent | Phase 1b Study PVSRIPO for Recurrent Malignant Glioma in Children | Phase I | PVSRIPO | 12 | December 5, 2017–July 1, 2021 | NCT03043391 | Recruiting | |
Parvovirus | Recurrent | Parvovirus H-1 (ParvOryx) in Patients With Progressive Primary or Recurrent Glioblastoma Multiforme | Phase I-IIa | H-1PV | 18 | September 2011–May 2015 | NCT01301430 | Completed |
Reovirus | Recurrent | A Phase I Trial of Intratumoral Administration of Reovirus in Patients With Histologically Confirmed Recurrent Malignant Gliomas | Phase I | Reolysin | 12 | June 2002–July 2005 | N/A89 | Completed |
Recurrent | Safety and Efficacy Study of REOLYSIN® in the Treatment of Recurrent Malignant Gliomas | Phase I | Reolysin | 18 | July 2006–June 2010 | NCT00528684 90 | Completed | |
Recurrent | Wild-Type Reovirus in Combination With Sargramostim in Treating Younger Patients With High-Grade Relapsed or Refractory Brain Tumors | Phase I | Reolysin | 6 | June 21, 2015–January 1, 2025 | NCT02444546 | Active, not recruiting | |
Vaccinia | Recurrent | Safety and Efficacy of the Oncolytic Virus Armed for Local Chemotherapy, TG6002/5-FC, in Recurrent Glioblastoma Patients | Phase I-II | TG6002 | 78 | October 12, 2017–September 2021 | NCT03294486 | Recruiting |
Abbreviations: HSV, herpes simplex virus; MV-CEA, measles virus carcinoembryonic antigen.
Citations are included only for clinical trials with published results.
Table 2.
Viral Vector | Mechanism/Pathway Involved | Effect(s) on Tumor Cell |
---|---|---|
Adenovirus | ||
REIC/Dkk-3 + cRGD | Activation caspase-9; reduced expression B-catenin | Decreased proliferation rate |
Antisense MMP-9 | Downregulation of MMP-9 activity | Impaired tumor invasiveness |
DNX-2401 + pembrolizumab | Increased epitope presentation to CD8+ Tcells | Induced antiglioma immune response |
AAV8 and AAV9 +IFN-B | Increase in tumor-associated microglia | Improved tumor sensitivity to chemoradiation; improved median survival |
dsAAV2 | Downregulation of TGF-B | Suppressed tumor growth; reduced tumor immunosuppressive effects |
Herpes Virus | ||
G47Δ | Deletion of the γ134.5and α47 genes and a disabling lacZ insertion within ICP6; Murine angiostatin insertion | Gain of function mutation leading to increased MHC class I expression in infected cells this resulting in enhanced viral replication |
HSVtk + Flt3L | Release of HMGB1 | Phagocytosis of tumor; activation of immune response |
HSV-M032 | Deletion in both copies of γ134.5 gene; Insertion of Human IL-12 | Selective glioma cell replication and expression of IL-12 in infected glioma cells resulting in enhanced immune response and tumor cell lysis |
HSV-G207 | Deletion in both copies of γ134.5 gene and disabling lacZ insertion in UL39 | Selective glioma cell replication |
HSV-C134 | Deletion in both copies of γ134.5 gene, expression of the HCMVTRS1 gene product | Selective and enhanced glioma cell replication |
rQNestin34.5v.2 | Deletion in γ134.5 gene and UL39; ICP-34.5 under control of synthetic nestin promoter | Selective and enhanced glioma cell replication |
Lentivirus | ||
Sh-SirT1 | Downregulation SirT1 | Increased tumor sensitivity to radiotherapy |
Sh-TLX | Downregulation TLX; expression of TET3 | Impaired tumor growth and tumorigenicity of stem cells |
GAS1 + PTEN | Decreased AKT and ERK 1/2 expression | Impaired tumor growth |
Paramoxyvirus | ||
Measles (MV-CEA) | Attenuated strain modified to express the carcinoembryonic antigen gene | Designed to track viral gene expression in vivo via serum analysis to optimize dosing and administration schedule without resorting to histologic tissue analysis |
Measles (MV-NIS) | Attenuated strain modified to express human thyroidal sodium iodide symporter (NIS) gene | NIS can act as a reporter gene that enables the non-invasive tracking of viral localization, spread, gene expression and replication over time. NIS may also be used as a therapeutic transgene by allowing intracellular uptake of isotopes, such as131 [I] (radiovirotherapy) |
Picornavirus | ||
Poliovirus (PVSRIPO) | Enhanced immune cell infiltration; reduction of TIM-3 expression | Promote immune response and tumor inflammation |
Retrovirus | ||
Toca 511 | Increased delivery of 5-FC to tumor | Increased tumor sensitivity to radiotherapy |
Placing ICP34.5 or its human ortholog GADD34 under nestin promoter control (rQNestin34.5 and NG34) resulted in enhanced selectivity and efficacy compared with control virus in preclinical models.38,39 Nestin encodes for the intermediate filament, which is a protein expressed during neuronal embryogenesis but not in the adult brain and it has been shown to be upregulated in malignant glioma, resulting in selective production of ICP34.5.38,40 An ongoing Phase I clinical trial is currently ongoing to test the safety of these viral constructs (see Table 1). Another approach uses oHSV G47Δ constructed by deleting the α47 gene, responsible for inhibiting the transporter associated with antigen presentation, from γ34.5-deficient HSV-1 vectors; leading to increased MHC class I expression in infected human cells and enhanced viral replication. Ongoing phase I-IIa clinical trials in Japan are assessing the safety and efficacy of G47Δ for the treatment of GBM.41,42 Interim analysis of these showed that the 1-year survival rate of 13 patients was 92.3%.42
Pathophysiological hypoxia is a hallmark of high-grade gliomas. It fosters the glioma stemlike cell (GSC) phenotype and has been linked to tumor development, invasiveness, and resistance to chemotherapy and radiation. Although GSCs demonstrated no inherent resistance to oHSV, hypoxia may limit the oncolytic effect of some oHSVs.43–46 To improve replication in such hostile environments without increasing neurovirulence, chimeric HSV C134 was developed to express the human cytomegalovirus (HCMV) PKR-evasion gene.43,47 C134 is able to evade PKR-mediated protein shutoff and maintain late viral protein synthesis to significantly enhance virus replication, including in hypoxic conditions.43 There is an ongoing clinical trial assessing the safety and therapeutic benefit of C134.48
In addition to direct oncolytic effects, oHSV can elicit a robust antitumor immune response.1 Viruses with insertion of proinflammatory cytokine genes have been described, such as IL-12, which results in intratumoral production of IL-12 during viral replication to enhance targeted immune destruction.13 IL-12 has potent antitumor properties that enhance the cytolytic activity of natural killer cells and cytotoxic T cells.49 It also promotes the development of TH-1 immune response, potentially eliciting a more durable antitumor effect.49 Treatment with oHSV models producing IL-12 in combination with immune checkpoints (CTLA-4 and PD-1) have also shown promising results.50 There are several completed and ongoing trials assessing the safety and therapeutic benefit of second-generation oHSVs (eg, IL-12 producing oHSV M032) in adults.13,14
Measles Virus
Measles virus (MV) is a single-stranded, negative-sense, enveloped RNA virus within the Morbillivirus genera of the Paramyxoviridae family. MV expresses a glycoprotein hemagglutinin protein H that has a high affinity for CD46 receptors shown to be overexpressed in GBM cells.51,52 The MV Edmonston strain (MV-Edm), a well-known attenuated strain used to vaccinate humans against MV, has been further modified to express the carcinoembryonic antigen gene (MV-CEA).53
Phuong and colleagues54 were the first to show that intravenous MV-CEA resulted in significantly prolonged survival and regression of in vivo glioblastoma tumor in mice bearing subcutaneous and orthotopic U87 tumors MV-CEA treated mice had no neurologic or clinical toxicity, which sparked further investigation. In subsequent studies, MV specificity for GBM was increased by developing retargeted oncolytic measles strains that invade via different receptors: epidermal growth factor receptor (MV-EGFR), EGF receptor variant III (MV-EGFRvIII), and IL-13Rα2 receptor.55–58 Additional studies demonstrated that MV immunovirotherapy against GBMs can be enhanced with either adjuvant radiation therapy or anti-PD-1 antibody therapy.59,60 Recombinant oncolytic MV (MV-NIS) is another example that was designed to express human thyroidal sodium iodide symporter (NIS) gene. NIS can act as a reporter gene via radiotracers and can also be used as a therapeutic transgene via radiovirotherapy, by allowing intracellular uptake 131[I] potentially enhancing the therapeutic efficacy.61
A phase 1 clinical trial treated 23 measles immune patients who were candidates for gross total or subtotal tumor resection of recurrent GBM with intracranial injection of MV-CEA.62 One group received a total dose of MV-CEA ranging from 105 to 2 × 107 TCID50 via injection into the resection cavity. The second group of patients received one intratumoral MV-CEA injection and subsequently underwent tumor resection 5 days following this first intratumoral injection–time for projected maximum viral replication to be achieved– with a second MV-CEA injection into the resection cavity before closure. Resected tumor specimens were analyzed with in situ hybridization and immunohistochemistry.63
Poliovirus
Poliovirus is a positive-sense, single-stranded RNA encapsulated virus belonging to the Picornaviridae family known for its neurotoxic effects.64 The prototype oncolytic poliovirus developed by Gromeier and colleagues,65 PVS-RIPO, is the live attenuated poliovirus type 1 (Sabin) with its internal ribosome entry site (IRES) replaced by that of human rhinovirus type 2 (HRV2). Although this polio-rhinovirus chimera was found to possess neuronal incompetence, in vitro studies demonstrated its ability to infect and reduce glioma cell viability and trigger cytolysis of GBM primary cultures.66–71 In subsequent animal studies, PVSRIPO was able to arrest tumor growth in both murine GBM flank tumor models and improve survival after intracranial virus administration in mice.66,72 In addition, its efficacy was found to be correlated with CD155 expression, known to be overexpressed in human GBM.73,74
Indeed, its moderate success in preclinical models paved the way for a phase II clinical trial involving inoculation of 61 patients with recurrent GBM with PVS-RIPO. The results were published in a landmark article in 2018, which not only corroborated safety of intratumoral viral administration in humans but demonstrated an increase in patient survival rate from 4% to 21% at 36 months when compared with historical control groups.75,76 Three other clinical trials on PVSRIPO are currently ongoing assessing safety in children and combination therapy with lomustine (CCNU) and pembrolizumab.76–78 Because clinical and radiographic responses were observed after the first cycle of chemotherapy administered for tumor progression in patients receiving PVSRIPO infusion, a second follow-up randomized trial of PVSRIPO alone or in combination with single-cycle CCNU in patients with recurrent World Health Organization grade IV malignant glioma is ongoing to further assess the potential of combination therapy CCNU.76
Reovirus
Another human virus that has shown oncolytic ability is the Respiratory Enteric Orphan virus or Reovirus, a segmented nonenveloped double-stranded RNA virus composed of 3 size groups. This naturally occurring virus, which is commonly isolated in the respiratory and gastrointestinal tracts of humans but causes mild to no symptoms, preferentially targets the activated RAS pathway.79 The numerous downstream effectors induced by the RAS/RalGEF/p38 pathway in particular, have been implicated in promoting the reovirus life cycle and leading to cell death.80–84
Animal studies in severe combined immunodeficient (SCID) mice containing subcutaneous MG cell lines U251 N and intracerebral cell lines U251 N and U87lacZ showed a reduction in tumor burden after infection with serotype 3 (strain Dearing) live virus.85,86 Lethality was also demonstrated in vitro in 83% of 24 established malignant glioma cell lines. The susceptibility of cells to reovirus may in part be attributed to the various ways reovirus circumvents cell defense mechanisms. For example, when 3-dimensional cultures of stem cell-like cells (GSC) from grade IV gliomas (glioblastoma) expressing junction adhesion molecule-A (JAM-A) were infected by the wild-type (wt) variant and the JAM-A independent jin-1 reovirus variant, viral entry and protein synthesis were similar.87 JAM-A is typically used by wt reovirus for cell entry and level of expression is correlated with infectivity. These results suggest that reovirus may use alternative entry pathways for infectivity that avoid the JAM-A adhesion route. Interestingly, reovirus has been found to also upregulate PD-L1 expression lending credence to its use as part of a multifaceted tumor killing strategy with the use of PD-1/PD-L1 inhibitors.88
The first clinical trial using reovirus in recurrent malignant glioma demonstrated that intratumoral injection was safe.89 Although the trial’s purpose was not to show efficacy, 6 patients lived more than 6 months, 3 patients lived more than 1 year, and 1 continued to survive at 54 months. A subsequent study using convection-enhanced delivery also confirmed safety and noticed improved survival >2 years in select patients.90 Intravenous administration of reovirus has also been evaluated in preclinical studies with promising results.91
ZOONOTIC ONCOLYTIC VIRUS MODELS
Newcastle Disease Virus
Newcastle disease virus (NDV) is a chicken pathogen with selective oncolytic properties applicable to various types of human cancer.92 Molecularly, NDV is an avian paramyxovirus with a negative-stranded RNA genome.17 Although the tumor-suppressive abilities of NDV have been extensively demonstrated through in vivo models and clinical trials, the exact mechanism is not fully understood. It is theorized that NDV achieves oncolysis via activation of a Ras pathway in addition to inducing secretion of tumor necrosis factor alpha (TNF-alpha) by mononuclear cells resulting in an enhanced antitumor immune response.17 More recent studies suggest that the Ras-related C3 botulism toxin substrate 1 (Rac1) pathway may be the target of NDV.92 Rac1 is involved in proliferation signaling by regulating gene transcription and G1 cell cycle progression. In GBM, Rac1 is therefore a crucial contributor to cell survival. NDV interactions with Rac1 are believed to induce cell cycle arrest along with degradation of the actin cytoskeleton and ultimately cell death.92 Murine models have shown increased long-term survival after NDV injection due to cytotoxic T-cell infiltration.16 However, this long-term survival benefit was not seen in immunodeficient murine models with depleted CD8 cells, stressing the importance of an intact host immune system for maximal benefit.16 Type I interferon (IFN) expression in GBM cells also greatly impacts the effectiveness of NDV given the role of IFN in promoting an antiviral state and decreasing viral replication.93 Nonetheless, recombinant NDV expression of an IFN antagonistic protein can overcome this protective role of IFN in GBM cells.93
NDV delivery to GBM cells can be targeted via mesenchymal stem cells (MSCs). This technique takes advantage of the natural ability of MSCs to target sites of injury and inflammation, including tumors.94 Higher rates of apoptosis were demonstrated in glioma cells when MSCs were used as the vector for NDV delivery as compared with direct NDV infection with similar virus titers. Moreover, TNF-related apoptosis-inducing ligand (TRAIL) has been identified as a key mediator in the antitumor effects of these hybrid MSCs due to synergy between TRAIL and NDV in the induction of apoptosis.94 NDV can also potentiate the effects of TMZ. Bai and colleagues95 found that when combined with TMZ, NDV inhibits AKT and activates AMPK, ultimately resulting in enhanced antitumor effects of TMZ and extended survival in a murine model. Clinical trials have demonstrated therapeutic efficacy and safety of autologous NDV-modified cellular vaccines or oncolytic effects in clinical trials but larger clinical trials are necessary to confirm efficacy.96 In a phase I/II clinical trial, Freeman and colleagues97 showed that the toxicity of NDV strain (HUJ, lentogenic) was minimal and a maximal tolerated dose was not achieved when administered intravenously to 14 patients with GBM using intrapatient dose escalation (1–11 billion infectious units) followed by 3 cycles of 55 billion infectious units with 1 patient achieving a complete response, and the others developed progressive disease.
Rodent Parvovirus
Certain members of the Parvoviridae family, a group of nonenveloped icosahedral single-stranded DNA viruses, can selectively kill malignant glioma cells while sparing normal cells in preclinical studies. These include rodent oncolytic viruses such as the Minute Virus of Mice and the more extensively studied rat parvovirus H-1PV.98 Intratumoral and intravenous injection of H-1PV into 12 immunodeficient rats containing the U87 human glioma cell line resulted in prolonged survival and decreased tumor burden compared with controls.99 The efficacy was in part due to a secondary viremia that resulted from progeny particles after initial tumor infection and boosted infection of remaining tumor cells. The lethality of H-1PV also extends to malignant gliomas resistant to death ligands such as TRAIL and DNA-damaging agents such as cisplatin.100 The virus triggers accumulation of lysosomal cathepsins and downregulating cathepsin inhibitors. The orientation of certain variable regions of the capsid protein of H-1PV has also been tied to its infectivity.101
Studies in short-term and low-passage cultures of human grade IV and gliosarcoma cell lines also showed increased susceptibility to H-1PV at low multiplicities of infection (MOI; 1–5 infectious units per cell).102 These cell cultures more closely parallel clinically diseased cells than do cells from long-term in vitro cell cultures. Intranasal application of H-1PV has also been shown to prolong survival in immunodeficient rats containing U87 human glioma cells versus controls. A Phase I/IIa trial of H-1PV in 18 patients demonstrated no dose-limited toxicity and widespread distribution after intratumoral and intravenous injection.103,104
Other Viral Vectors
Several other potential viral vectors have been described, but have not been assessed in clinical trials for GBM. Pseudorabies virus (PRV) and the Seneca Valley Virus (SVV), 2 viruses in which pigs are the natural host, have shown potential as oncolytic targets. However, intravenous infusion of PRV did not result in uptake within intracranial glioma cells.105,106 SVV improved survival in mice bearing GBM as well as medulloblastoma and retinoblastoma models, which led to phase 1 clinical trials in adults and children with neuroendocrine tumors, which demonstrated safety, but no clear antitumor responses, and all patients rapidly developed anti-SVV antibodies and cleared the virus.107,108 Vesicular Stomatitis Virus (VSV) and Sindbis Virus (SIN) are mosquito-borne viruses that have also shown oncolytic potential. Chimeric VSV-lymphocytic choriomeningitis virus, and VSV-Chikungunya virus mutants with replacement of the VSV glycoprotein have demonstrated tumor lysis with decreased toxicity to normal cells in glioma and intracranial melanoma mouse models.109,110 SIN has tropism for neural cells and can cause encephalitis in mice.17 Tropism for tumor cells is believed to be related to the high affinity laminin receptor, which is overexpressed in many tumors.111 SIN can be a vector for introduction of hyperfusogenic membrane glycoproteins that lead to formation of syncytia and apoptosis.112 Myxoma virus and Vaccinia virus (VV), within the Poxviridae family are the most promising candidates for malignant glioma virotherapy because they are highly immunogenic and capable of creating antitumor immunity.113–116
NONONCOLYTIC VIRAL VECTORS FOR GENE THERAPY OR TARGETED DRUG DELIVERY
Gene therapy has emerged as a potential treatment for malignant gliomas, whereby a vector introduces tumor suppressing or growth regulating genes into malignant cells. Multiple approaches are used for gene therapy including suicide gene, oncolytic gene, and tumor suppressor gene therapies.117
Viruses are a prime candidate for the introduction of gene therapies. They create a potent cytotoxic effect and are easily modified to facilitate genetic engineering.19 Current approaches are attempting to target proteins commonly mutated or upregulated in GBM, including EGFR, PTEN, IDH-1, and p53.118 The most common viral vectors include neurotropic retrovirus and adenoviruses. Retroviral vectors were among the first studied, and the first trial began in 1992 with a retroviral HSV-thymidine kinase (HSV-tk) with ganciclovir. HSV-tk acts as a suicide gene and converts the prodrug ganciclovir into its active form to inhibit cell division and DNA replication. The efficacy of this treatment was limited to small tumor sizes given its poor transfection efficiency.119
Adenoviral vectors have been used in clinical trials. An early study of an adenoviral vector with wt p53 gene (Ad-p53) showed efficacious transfection of tumor cells with minimal toxicity; however, similar to retroviral vectors, Ad-p53 demonstrated poor ability to penetrate tumor tissue widely.120 Sandmair and colleagues121 demonstrated increased survival time in patients receiving ganciclovir with adenovirus-delivered HSV-tk as compared with retrovirus delivery, again demonstrating poor retroviral transfection and tumor penetrance. In addition, adenovirus and HSV vectors have been used to introduce CD, which convert the prodrug 5-fluorocytosine into 5-flurouracil, inducing apoptosis.122 A phase I study in patients with recurrent glioma with aglatimagene besadenovec (AdV-tk), which adenoviral vector engineered to express the HSV thymidine kinase (HSV-tk) gene in conjunction with a synthetic anti-herpetic prodrug acyclic guanosine analogue administration demonstrated a safe dose range with 3 of 13 patients surviving more than 24 months.123 A subsequent phase I trial in children treated with AdV-tk as adjuvant to surgery and radiation for pediatric malignant glioma and recurrent ependymoma also showed safety and potential efficacy.124
Lentiviral vectors have been used to introduce small-hairpin RNA (shRNA) to silence sirtuin 1 expression in GBM, which results in increased radiosensitivity with resultant increased tumor death.125 Similarly, lentiviral delivery of human orphan nuclear receptor tailless (TLX) shRNA resulted in tumor growth inhibition and decreased tumorigenicity.126,127
CHALLENGES, LIMITATIONS, AND FUTURE DIRECTIONS
Although clinical trials have been completed or are ongoing for several oncolytic viruses, only a few have moved beyond a Phase I clinical trial.16 Finding the ideal balance to achieve safety but also virulence to maximize efficacy remains a significant challenge.
Moreover, viral delivery remains a significant challenge, as most clinical trials have focused on intratumoral delivery. Recent trials have used stereotactic techniques to place a localized catheter into the tumor to use for administration of virus.31,33,35,37,128 This requires a neurosurgical procedure and may limit additional doses. Thus, innovative routes of administration need to be devised, such as systemic, intrathecal, intracavitary, and intraventricular delivery. However, the challenges of systemic delivery are considerable due to the blood-brain-barrier and virus neutralizing antibodies, and the safety of these routes needs to be confirmed.16
Although the clinical results of several oncolytic viruses have been promising including HSV, poliovirus, and adenovirus, these studies have all been in recurrent, often heavily pretreated patients. Thus, it will be important to test immunovirotherapy in upfront regimens. Furthermore, future studies are needed to combine oncolytic viruses with other potentially synergistic approaches to maximize oncolysis an antitumor immune response such as immune checkpoint inhibitors, CAR-T therapy enhanced with bispecific T-cell engagers (BiTE), vaccines, and other immunotherapies.14,50,129–131 For example, Saha and colleagues129 demonstrated durable responses in an orthotopic GBM model by combining anti-PD-1 and anti-CTLA-4 antibodies with oHSV expressing IL-12. An alternate approach to systemic delivery of checkpoint inhibitors is by using oncolytic viruses carrying genetic material to express the immune checkpoint inhibitors locally.103,132,133 In addition, CAR-T-cell therapy with bicistronic constructs can convert gliomas who have difficult-to-target surface topology to more familiar, targetable topology or help trigger enhanced immune responses with targeted, localized CD3 expression to facilitate local immunomodulation.130
SUMMARY
Immunovirotherapy has shown significant promise as a targeted therapy for malignant gliomas, and attempts to address several of the challenges often encountered in treatment, such as ability to treat unresectable lesions or addressing challenges encountered hypoxia, anti-inflammatory effects, and consequences of intratumoral and intertumoral heterogeneity in treatment. However, barriers related to therapeutic delivery, viral entry and replication, and immunosuppressed patients must be overcome. Strategies such as arming viral vectors with enhancements (therapeutic transgenes, checkpoint inhibition, host antiviral immune response, improved and selective replication) and combining viruses with synergistic agents must continue to be developed and tested in the clinics so that the great therapeutic potential of oncolytic immunovirotherapy can be realized.
KEY POINTS.
Immunovirotherapy has emerged as a promising targeted approach for treatment of GBM and other malignant gliomas.
There are multiple viral prototypes for targeted oncolytic virotherapy and targeted drug delivery in various stages of clinical development with promising results.
Herpes Simplex Virus type 1 offers numerous advantages as an oncolytic virus with several genetic enhancements currently being tested in clinical trials in adults and children.
CLINICS CARE POINTS.
To date, no oncolytic virus has been approved by the FDA for the treatment of malignant glioma and all remain investigational treatments.
Multiple ongoing clinical trials are currently enrolling participants, most of them available for patients with recurrent malignant gliomas.
Oncolytic viral models engineered to alter/modulate various cellular and inflammatory pathways leading to selective replication in tumor cells, enhanced immune response, impaired tumor angiogenesis, amongst others.
Multiple non-oncolytic viral vectors have been studied as gene therapy vectors in glioma; these varied approaches include increasing radiosensitivity via gene silencing and induction of tumor cell apoptosis in conjunction with various prodrug administrations.
Talimogene laherparepvec (T-VEC) is the first US Food and Drug Administration (FDA)-approved oncolytic virus; and is currently indicated for advanced melanoma. T-VEC is an oHSV that and expresses human granulocyte macrophage colony-stimulating factor (GM-CSF) to active the immune system and has specific genetic deletions that result in improved capacity for MHC presentation.
ACKNOWLEDGMENT
The manuscript was supported in part by National Cancer Institute grants R01CA222903, R01CA217179 and a grant from Gateway for Cancer Research (all JMM -Last Author). And supported in part by the National Institutes of Health (Training Grant R25 TW009337) funded by the Fogarty International Center, National Institutes of Health Office of the Director, and National Institute of Mental Health. (DEO - First author).
Footnotes
DISCLOSURES
Dr J.M. Markert holds equity (<8%) in Aettis, Inc. (a company that holds stocks of oncolytic virus); Treovir, Inc (25%), a company holding intellectual property and funding clinical trials of oncolytic virus for pediatric brain tumors. A company that Dr J.M. Markert formerly held equity in (<8%) Catherex, Inc., was purchased in a structured buyout. Dr J.M. Markert has served as a consultant for Imugene. He also holds a fraction of the IP associated with oncolytic virus C134, which is licensed by Mustang Biotech.
REFERENCES
- 1.Louis David N, Hiroko O, Wiestler Otmar D, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 2007; 114(2):97–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Ostrom Quinn T, Gino C, Gittleman H, et al. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2012–2016. Neuro Oncol 2019; 21(Supplement_5):v1–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Mackay A, Anna B, Carvalho D, et al. Integrated molecular meta-analysis of 1,000 pediatric high-grade and diffuse intrinsic pontine glioma. Cancer Cell 2017;32(4):520–37.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Wen Patrick Y, Santosh K. Malignant gliomas in adults. N Engl J Med 2008;359(5):492–507. [DOI] [PubMed] [Google Scholar]
- 5.Andreansky SS, He B, Gillespie GY, et al. The application of genetically engineered herpes simplex viruses to the treatment of experimental brain tumors. Proc Natl Acad Sci U S A 1996;93(21): 11313–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Roger S, Mason Warren P, van den Bent Martin J, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005;352(10):987–96. [DOI] [PubMed] [Google Scholar]
- 7.Omuro A, DeAngelis Lisa M. Glioblastoma and other malignant gliomas: a clinical review. JAMA 2013;310(17):1842–50. [DOI] [PubMed] [Google Scholar]
- 8.Cohen KJ, Pollack Ian F, Zhou T, et al. Temozolomide in the treatment of high-grade gliomas in children: a report from the Children’s Oncology Group. Neuro Oncol 2011;13(3):317–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Cheng L, Wu Q, Guryanova Olga A, et al. Elevated invasive potential of glioblastoma stem cells. Biochem Biophys Res Commun 2011;406(4):643–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Hirokazu S, Yoshikawa K, Makoto I, et al. Pathological features of highly invasive glioma stem cells in a mouse xenograft model. Brain Tumor Pathol 2014;31(2):77–84. [DOI] [PubMed] [Google Scholar]
- 11.Aboody Karen S, Brown A, Rainov Nikolai G, et al. Neural stem cells display extensive tropism for pathology in adult brain: evidence from intracranial gliomas. Proc Natl Acad Sci U S A 2000;97(23): 12846–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Bernstock Joshua D, Mooney James H, Ilyas A, et al. Molecular and cellular intratumoral heterogeneity in primary glioblastoma: clinical and translational implications. J Neurosurg 2019;1–9. 10.3171/2019.5.JNS19364. [DOI] [PubMed] [Google Scholar]
- 13.Patel Daxa M, Foreman Paul M, Burt NL, et al. Design of a phase I clinical trial to evaluate M032, a genetically engineered HSV-1 Expressing IL-12, in patients with recurrent/progressive glioblastoma multiforme, anaplastic astrocytoma, or gliosarcoma. Hum Gene Ther Clin Dev 2016; 27(2):69–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Totsch Stacie K, Charles S, Kang KD, et al. Oncolytic herpes simplex virus immunotherapy for brain tumors: current pitfalls and emerging strategies to overcome therapeutic resistance. Oncogene 2019;38(34):6159–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Foreman Paul M, Friedman Gregory K, Cassady Kevin A, et al. Oncolytic virotherapy for the treatment of malignant glioma. Neurotherapeutics 2017;14(2):333–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Martikainen M, Magnus E. Virus-based immunotherapy of glioblastoma. Cancers (Basel) 2019; 11(2). 10.3390/cancers11020186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Guido W, Koray O, van den Pol Anthony N. Oncolytic virus therapy of glioblastoma multiforme – concepts and candidates. Cancer J 2012;18(1): 69–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Zhang Q, Zhang J, Tian Y, et al. Efficacy of a novel double-controlled oncolytic adenovirus driven by the Ki67 core promoter and armed with IL-15 against glioblastoma cells. Cell Biosci 2020;10. 10.1186/s13578-020-00485-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Manikandan C, Kaushik A, Sen D. Viral vector: potential therapeutic for glioblastoma multiforme. Cancer Gene Ther 2020;27(5):270–9. [DOI] [PubMed] [Google Scholar]
- 20.Lang Frederick F, Conrad C, Gomez-Manzano C, et al. Phase I Study of DNX-2401 (Delta-24-RGD) oncolytic adenovirus: replication and immunotherapeutic effects in recurrent malignant glioma. J Clin Oncol 2018;36(14):1419–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Kim Julius W, Jason M, Young Jacob S, et al. A comparative study of replication-incompetent and -competent adenoviral therapy-mediated immune response in a murine glioma model. Mol Ther Oncolytics 2017;5:97–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Zineb B, Cor B, John C, et al. Low-dose oncolytic adenovirus therapy overcomes tumor-induced immune suppression and sensitizes intracranial gliomas to anti-PD-1 therapy. Neurooncol Adv 2020; 2(1). 10.1093/noajnl/vdaa011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Qiao J, Dey M, Chang Alan L, et al. Intratumoral oncolytic adenoviral treatment modulates the glioma microenvironment and facilitates systemic tumor-antigen-specific T cell therapy. Oncoimmunology 2015;4(8). 10.1080/2162402X.2015.1022302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Oka T, Kazuhiko K, Shimazu Y, et al. A super gene expression system enhances the anti-glioma effects of adenovirus-mediated REIC/Dkk-3 gene therapy. Sci Rep 2016;6:33319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kazuhiko K, Fujii K, Shimazu Y, et al. Study protocol of a Phase I/IIa clinical trial of Ad-SGE-REIC for treatment of recurrent malignant glioma. Future Oncol 2020;16(6):151–9. [DOI] [PubMed] [Google Scholar]
- 26.Yano S, Miwa S, Kishimoto H, et al. Experimental curative fluorescence-guided surgery of highly invasive glioblastoma multiforme selectively labeled with a killer-reporter adenovirus. Mol Ther 2015;23(7):1182–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Friedman Gregory K, Pressey Joseph G, Reddy Alyssa T, et al. Herpes simplex virus oncolytic therapy for pediatric malignancies. Mol Ther 2009; 17(7):1125–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Shah Amish C, Dale B, Yancey GG, et al. Oncolytic viruses: clinical applications as vectors for the treatment of malignant gliomas. J Neurooncol 2003;65(3):203–26. [DOI] [PubMed] [Google Scholar]
- 29.Wilcox Douglas R, Richard L. The herpes simplex virus neurovirulence factor γ34.5: revealing virus–host interactions. PLoS Pathog 2016;12(3). 10.1371/journal.ppat.1005449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Toshihiro M, Rabkin Samuel D, Yazaki T, et al. Attenuated multi–mutated herpes simplex virus–1 for the treatment of malignant gliomas. Nat Med 1995;1(9):938–43. [DOI] [PubMed] [Google Scholar]
- 31.Waters Alicia M, Johnston James M, Reddy Alyssa T, et al. Rationale and design of a Phase 1 clinical trial to evaluate HSV G207 alone or with a single radiation dose in children with progressive or recurrent malignant supratentorial brain tumors. Hum Gene Ther Clin Dev 2017;28(1):7–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Markert JM, Medlock MD, Rabkin SD, 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(10):867–74. [DOI] [PubMed] [Google Scholar]
- 33.Markert James M, Razdan Shantanu N, Hui-Chien K, et al. A phase 1 trial of oncolytic HSV-1, G207, given in combination with radiation for recurrent GBM demonstrates safety and radiographic responses. Mol Ther 2014;22(5):1048–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Markert James M, Liechty Peter G, Wang W, et al. Phase Ib trial of mutant herpes simplex virus G207 inoculated pre-and post-tumor resection for recurrent GBM. Mol Ther 2009;17(1):199–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Bernstock Joshua D, Wright Z, Bag Asim K, et al. Stereotactic placement of intratumoral catheters for continuous infusion delivery of herpes simplex virus -1 G207 in pediatric malignant supratentorial brain tumors. World Neurosurg 2019;122:e1592–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Bernstock Joshua D, Nunzio V, Rong Li, et al. A novel in situ multiplex immunofluorescence panel for the assessment of tumor immunopathology and response to virotherapy in pediatric glioblastoma reveals a role for checkpoint protein inhibition. Oncoimmunology 2019;8(12):e1678921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bernstock Joshua D, Bag Asim K, Fiveash J, et al. Design and rationale for first-in-human phase 1 immunovirotherapy clinical trial of oncolytic HSV G207 to treat malignant pediatric cerebellar brain tumors. Hum Gene Ther 2020;31(19–20):1132–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Kambara H, Okano H, Chiocca E, et al. An oncolytic HSV-1 mutant expressing ICP34.5 under control of a nestin promoter increases survival of animals even when symptomatic from a brain tumor. Cancer Res 2005;65(7):2832–9. [DOI] [PubMed] [Google Scholar]
- 39.Nakashima H, Nguyen T, Kasai K, et al. Toxicity and efficacy of a Novel GADD34-expressing Oncolytic HSV-1 for the treatment of experimental glioblastoma. Clin Cancer Res 2018;24(11):2574–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Dahlstrand J, Collins VP, Lendahl U. Expression of the class VI intermediate filament nestin in human central nervous system tumors. Cancer Res 1992; 52(19):5334–41. [PubMed] [Google Scholar]
- 41.Taguchi S, Fukuhara H, Todo T. Oncolytic virus therapy in Japan: progress in clinical trials and future perspectives. Jpn J Clin Oncol 2019;49(3): 201–9. [DOI] [PubMed] [Google Scholar]
- 42.Tomoki T ATIM-14. results of phase II clinical trial of oncolytic herpes virus G47Δ in patients with glioblastoma. Neuro Oncol 2019;21(Suppl 6):vi4. [Google Scholar]
- 43.Friedman GK, Nan L, Haas MC, et al. γ134.5-deleted HSV-1-expressing human cytomegalovirus IRS1 gene kills human glioblastoma cells as efficiently as wild-type HSV-1 in normoxia or hypoxia. Gene Ther 2015;22(4):348–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Jensen Randy L. Brain tumor hypoxia: tumorigenesis, angiogenesis, imaging, pseudoprogression, and as a therapeutic target. J Neurooncol 2009; 92(3):317–35. [DOI] [PubMed] [Google Scholar]
- 45.Friedman Gregory K, Langford Catherine P, Coleman Jennifer M, et al. Engineered herpes simplex viruses efficiently infect and kill CD1331 human glioma xenograft cells that express CD111. J Neurooncol 2009;95(2):199–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Friedman Gregory K, Haas Marilyn C, Kelly VM, et al. Hypoxia moderates γ134.5-deleted herpes simplex virus oncolytic activity in human glioma xenoline primary cultures. Transl Oncol 2012;5(3):200–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Cassady Kevin A. Human cytomegalovirus TRS1 and IRS1 gene products block the double-stranded-RNA-activated host protein shutoff response induced by herpes simplex virus type 1 infection. J Virol 2005;79(14):8707–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Markert James M. Trial of C134 in patients with recurrent GBM - Full Text View - ClinicalTrials.gov. Available at: https://clinicaltrials.gov/ct2/show/NCT03657576. Accessed September 17, 2020.
- 49.Parker JN, Gillespie GY, Love CE, et al. Engineered herpes simplex virus expressing IL-12 in the treatment of experimental murine brain tumors. Proc Natl Acad Sci U S A 2000;97(5): 2208–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Saha D, Martuza Robert L, Rabkin Samuel D. Macrophage polarization contributes to glioblastoma eradication by combination immunovirotherapy and immune checkpoint blockade. Cancer Cell 2017;32(2):253–67.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Dörig Ruth E, Anne M, Chopra A, et al. The human CD46 molecule is a receptor for measles virus (Edmonston strain). Cell 1993;75(2):295–305. [DOI] [PubMed] [Google Scholar]
- 52.MaenpAa A, Sami J, Hakulinen J, et al. Expression of complement membrane regulators membrane cofactor protein (CD46), decay accelerating factor (CD55), and protectin (CD59) in human malignant gliomas. Am J Pathol 1996;148(4):14. [PMC free article] [PubMed] [Google Scholar]
- 53.Peng Kah-Whye, Facteau S, Wegman T, et al. Non-invasive in vivo monitoring of trackable viruses expressing soluble marker peptides. Nat Med 2002; 8(5):527–31. [DOI] [PubMed] [Google Scholar]
- 54.Phuong LK, Allen C, Peng KW, et al. Use of a vaccine strain of measles virus genetically engineered to produce carcinoembryonic antigen as a novel therapeutic agent against glioblastoma multiforme. Cancer research 2003;63(10):2462–9. [PubMed] [Google Scholar]
- 55.Allen C, Vongpunsawad S, Nakamura T, et al. Retargeted oncolytic measles strains entering via the EGFRvIII receptor maintain significant antitumor activity against gliomas with increased tumor specificity. Cancer Res 2006;66(24):11840–50. [DOI] [PubMed] [Google Scholar]
- 56.Allen C, Paraskevakou G, Iankov I, et al. Interleukin-13 displaying retargeted oncolytic measles virus strains have significant activity against gliomas with improved specificity. Mol Ther 2008; 16(9):1556–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Allen C, Opyrchal M, Aderca I, et al. Oncolytic measles virus strains have significant antitumor activity against glioma stem cells. Gene Ther 2013; 20(4):444–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Georgia P, Allen C, Nakamura T, et al. Epidermal growth factor receptor (EGFR)–retargeted measles virus strains effectively target EGFR- or EGFRvIII expressing gliomas. Mol Ther 2007; 15(4):677–86. [DOI] [PubMed] [Google Scholar]
- 59.Liu C, Sarkaria JN, Petell CA, et al. Combination of measles virus virotherapy and radiation therapy has synergistic activity in the treatment of glioblastoma multiforme. Clin Cancer Res 2007;13(23): 7155–65. [DOI] [PubMed] [Google Scholar]
- 60.Hardcastle J, Mills L, Malo Courtney S, et al. Immunovirotherapy with measles virus strains in combination with anti–PD-1 antibody blockade enhances antitumor activity in glioblastoma treatment. Neuro Oncol 2017;19(4):493–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Pavlos M, Dispenzieri A, Evanthia G. Clinical testing of engineered oncolytic measles virus strains in the treatment of cancer: An overview. Curr Opin Mol Ther 2009;11(1):43–53. [PMC free article] [PubMed] [Google Scholar]
- 62.Evanthia G Viral therapy in treating patients with recurrent glioblastoma multiforme - full text view - ClinicalTrials.gov. Available at: https://clinicaltrials.gov/ct2/show/NCT00390299. Accessed November 29, 2020.
- 63.Pavlos M, Mateusz O, Dispenzieri A, et al. Clinical trials with oncolytic measles virus: current status and future prospects. Curr Cancer Drug Targets 2018;18(2). 10.2174/1568009617666170222125035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Man Mohan M, Mehndiratta P, Pande R. Poliomyelitis: historical facts, epidemiology, and current challenges in eradication. Neurohospitalist 2014; 4(4):223–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Gromeier M, Alexander L, Wimmer E. Internal ribosomal entry site substitution eliminates neurovirulence in intergeneric poliovirus recombinants. Proc Natl Acad Sci U S A 1996;93(6):2370–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Gromeier M, Lachmann S, Rosenfeld MR, et al. Intergeneric poliovirus recombinants for the treatment of malignant glioma. Proc Natl Acad Sci U S A 2000;97(12):6803–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Merrill Melinda K, Dobrikova Elena Y, Matthias G. Cell-type-specific repression of internal ribosome entry site activity by double-stranded RNA-binding protein 76. J Virol 2006;80(7):3147–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Merrill Melinda K, Matthias G. The double-stranded RNA binding protein 76:NF45 heterodimer inhibits translation initiation at the rhinovirus type 2 internal ribosome entry site. J Virol 2006; 80(14):6936–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Dobrikova Elena Y, Goetz C, Walters Robert W, et al. Attenuation of neurovirulence, biodistribution, and shedding of a poliovirus:rhinovirus chimera after intrathalamic inoculation in Macaca fascicularis. J Virol 2012;86(5):2750–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Yang X, Chen E, Jiang H, et al. Evaluation of IRES-mediated, cell-type-specific cytotoxicity of poliovirus using a colorimetric cell proliferation assay. J Virol Methods 2009;155(1):44–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Merrill Melinda K, Bernhardt G, Sampson John H, et al. Poliovirus receptor CD155-targeted oncolysis of glioma. Neuro Oncol 2004;6(3):208–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Dobrikova Elena Y, Trevor B, Poiley-Nelson J, et al. Recombinant oncolytic poliovirus eliminates glioma in vivo without genetic adaptation to a pathogenic phenotype. Mol Ther 2008;16(11): 1865–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Vidyalakshmi C, Bryant Jeffrey D, Piao H, et al. Validation of an immunohistochemistry assay for detection of CD155, the poliovirus receptor, in malignant gliomas. Arch Pathol Lab Med 2017; 141(12):1697–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Goetz C, Dobrikova E, Mayya S, et al. Oncolytic poliovirus against malignant glioma. Future Virol 2011;6(9):1045–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Annick D, Matthias G, Herndon James E, et al. Recurrent glioblastoma treated with recombinant poliovirus. N Engl J Med 2018;379(2):150–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Istari Oncology, Inc. PVSRIPO for recurrent glioblastoma (GBM) - full text view–ClinicalTrials.gov. Available at: https://clinicaltrials.gov/ct2/show/NCT01491893. Accessed November 23, 2020.
- 77.Istari Oncology, Inc. A phase 2, open-label, single arm study evaluating the efficacy, safety and tolerability of PVSRIPO and the immune checkpoint inhibitor pembrolizumab in the treatment of patients with recurrent glioblastoma.- full text view–ClinicalTrials.gov. Available at: https://clinicaltrials.gov/ct2/show/NCT04479241. Accessed November 23, 2020.
- 78.Istari Oncology, Inc. Phase 1b Study PVSRIPO for recurrent malignant glioma in children–full text view–ClinicalTrials.gov. Available at: https://clinicaltrials.gov/ct2/show/NCT03043391. Accessed November 23, 2020.
- 79.Prior Ian A, Hood Fiona E, Hartley James L. The frequency of ras mutations in cancer. Cancer Res 2020;80(14):2969–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Gong J, Mita Monica M. Activated ras signaling pathways and reovirus oncolysis: an update on the mechanism of preferential reovirus replication in cancer cells. Front Oncol 2014;4:167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Norman Kara L, Lee Patrick WK. Reovirus as a novel oncolytic agent. J Clin Invest 2000;105(8): 1035–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Norman Kara L, Lee Patrick WK. Not all viruses are bad guys: the case for reovirus in cancer therapy. Drug Discov Today 2005;10(12):847–55. [DOI] [PubMed] [Google Scholar]
- 83.Smakman N, van der Bilt JDW, van den Wollenberg DJM, et al. Immunosuppression promotes reovirus therapy of colorectal liver metastases. Cancer Gene Ther 2006;13(8):815–8. [DOI] [PubMed] [Google Scholar]
- 84.Strong JE, Coffey MC, Tang D, et al. The molecular basis of viral oncolysis: usurpation of the Ras signaling pathway by reovirus. EMBO J 1998; 17(12):3351–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Wilcox ME, Yang W, Senger D, et al. Reovirus as an oncolytic agent against experimental human malignant gliomas. J Natl Cancer Inst 2001;93(12): 903–12. [DOI] [PubMed] [Google Scholar]
- 86.Radhashree M, Ghalib Mohammad H, Goel S. Reovirus: a targeted therapeutic – progress and potential. Mol Cancer Res 2012;10(12). 10.1158/1541-7786.MCR-12-0157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.van den Hengel SK, Balvers RK, Dautzenberg IJC, et al. Heterogeneous reovirus susceptibility in human glioblastoma stem-like cell cultures. Cancer Gene Ther 2013;20(9):507–13. [DOI] [PubMed] [Google Scholar]
- 88.Samson A, Scott Karen J, Taggart D, et al. Intravenous delivery of oncolytic reovirus to brain tumor patients immunologically primes for subsequent checkpoint blockade. Sci Transl Med 2018;10(422). 10.1126/scitranslmed.aam7577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Forsyth P, Gloria R, George D, et al. A Phase I trial of intratumoral administration of reovirus in patients with histologically confirmed recurrent malignant gliomas. Mol Ther 2008;16(3):627–32. [DOI] [PubMed] [Google Scholar]
- 90.Kicielinski Kimberly P, Chiocca EA, Yu John S, et al. Phase 1 clinical trial of intratumoral reovirus infusion for the treatment of recurrent malignant gliomas in adults. Mol Ther 2014;22(5):1056–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Romit C, Tran H, Giovanni S, et al. The oncolytic virus, pelareorep, as a novel anticancer agent: a review. Invest New Drugs 2015;33(3):761–74. [DOI] [PubMed] [Google Scholar]
- 92.Jafri Malin A, Mustafa Z, Aini I. Newcastle disease virus interaction in targeted therapy against proliferation and invasion pathways of glioblastoma multiforme. Biomed Res Int 2014;2014. 10.1155/2014/386470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.García-Romero N, Palacín-Aliana I, Esteban-Rubio S, et al. Newcastle disease virus (NDV) oncolytic activity in human glioma tumors is dependent on CDKN2A-Type I IFN gene cluster codeletion. Cells 2020;9(6). 10.3390/cells9061405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Gila K, Jiang W, Shimon S, et al. Mesenchymal stem cells enhance the oncolytic effect of Newcastle disease virus in glioma cells and glioma stem cells via the secretion of TRAIL. Stem Cell Res Ther 2016;7. 10.1186/s13287-016-0414-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Bai Y, Chen Y, Hong X, et al. Newcastle disease virus enhances the growth-inhibiting and proapoptotic effects of temozolomide on glioblastoma cells in vitro and in vivo. Sci Rep 2018;8. 10.1038/s41598-018-29929-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Shi J, Sun P, Zhang Y, et al. The antitumor effects of Newcastle disease virus on glioma. Biocell 2019; 43(3):119–28. [Google Scholar]
- 97.Freeman Arnold I, Zichria ZR, Gomori John M, et al. Phase I/II Trial of Intravenous NDV-HUJ oncolytic virus in recurrent glioblastoma multiforme. Mol Ther 2006;13(1):221–8. [DOI] [PubMed] [Google Scholar]
- 98.Paglino Justin C, Koray O, van den Pol Anthony N. LuIII parvovirus selectively and efficiently targets, replicates in, and kills human glioma cells. J Virol 2012;86(13):7280–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Geletneky K, Kiprianova I, Ayache A, et al. Regression of advanced rat and human gliomas by local or systemic treatment with oncolytic parvovirus H-1 in rat models. Neuro Oncol 2010;12(8):804–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Piazza MD, Carmen M, Geletneky K, et al. Cytosolic activation of cathepsins mediates parvovirus H-1-induced killing of cisplatin and TRAIL-resistant glioma cells. J Virol 2007;81(8):4186–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Cho I-R, Kaowinn S, Song J, et al. VP2 capsid domain of the H-1 parvovirus determines susceptibility of human cancer cells to H-1 viral infection. Cancer Gene Ther 2015;22(5):271–7. [DOI] [PubMed] [Google Scholar]
- 102.Calle Marta Herrero y, Cornelis Jan J, Herold-Mende C, et al. Parvovirus H-1 infection of human glioma cells leads to complete viral replication and efficient cell killing. Int J Cancer 2004;109(1): 76–84. [DOI] [PubMed] [Google Scholar]
- 103.Geletneky Karsten, H Johannes, Rommelaere J, 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] [PMC free article] [PubMed] [Google Scholar]
- 104.Geletneky K, Hajda J, Angelova Assia L, et al. Oncolytic H-1 parvovirus shows safety and signs of immunogenic activity in a first phase I/IIa Glioblastoma Trial. Mol Ther 2017;25(12):2620–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Guido W, Tattersall P, Pol Anthony N. Targeting human glioblastoma cells: comparison of nine viruses with oncolytic potential. J Virol 2005;79(10): 6005–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Koray O, Guido W, Piepmeier Joseph M, et al. Systemic vesicular stomatitis virus selectively destroys multifocal glioma and metastatic carcinoma in brain. J Neurosci 2008;28(8):1882–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Rudin Charles M, Poirier John T, Senzer Neil N, et al. Phase I clinical study of seneca valley virus (SVV-001), a replication-competent picornavirus, in advanced solid tumors with neuroendocrine features. Clin Cancer Res 2011;17(4):888–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Burke MJ, Charlotte A, Weigel Brenda J, et al. Phase I trial of seneca valley virus (NTX-010) in children with relapsed/refractory solid tumors: a report of the Children’s Oncology Group. Pediatr Blood Cancer 2015;62(5):743–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Muik Alexander, Inna Kneiske, Werbizki Marina, et al. Pseudotyping vesicular stomatitis virus with lymphocytic choriomeningitis virus glycoproteins enhances infectivity for glioma cells and minimizes neurotropism. J Virol 2011;85(11):5679–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Zhang X, Mao G, van den Pol Anthony N. Chikungunya-vesicular stomatitis chimeric virus targets and eliminates brain tumors. Virology 2018;522: 244–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Wang KS, Kuhn RJ, Strauss EG, et al. High-affinity laminin receptor is a receptor for Sindbis virus in mammalian cells. J Virol 1992;66(8):4992–5001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Zhang J, Frolov I, Russell Stephen J. Gene therapy for malignant glioma using Sindbis vectors expressing a fusogenic membrane glycoprotein. J Gene Med 2004;6(10):1082–91. [DOI] [PubMed] [Google Scholar]
- 113.Villa Nancy Y, Wasserfall Clive H, Meacham Amy M, et al. Myxoma virus suppresses proliferation of activated T lymphocytes yet permits oncolytic virus transfer to cancer cells. Blood 2015; 125(24):3778–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Torres-Domínguez Lino E, McF Grant. Poxvirus oncolytic virotherapy. Expert Opin Biol Ther 2019; 19(6):561–73. [DOI] [PubMed] [Google Scholar]
- 115.Lun XQ, Jang J-H, Tang N, et al. Efficacy of systemically administered oncolytic vaccinia virotherapy for malignant gliomas is enhanced by combination therapy with rapamycin or cyclophosphamide. Clin Cancer Res 2009;15(8):2777–88. [DOI] [PubMed] [Google Scholar]
- 116.Lun X, Yang W, Tommy A, et al. Myxoma virus is a novel oncolytic virus with significant antitumor activity against experimental human gliomas. Cancer Res 2005;65(21):9982–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Hidehiro O, Smith Christian A, Rutka James T. Gene therapy for malignant glioma. Mol Cell Ther 2014;2:21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Breanne C, Lee JS, Alexander-Bryant Angela A. Vectors for glioblastoma gene therapy: viral & non-viral delivery strategies. Nanomaterials (Basel) 2019;9(1). 10.3390/nano9010105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Rainov NG. A phase III clinical evaluation of herpes simplex virus type 1 thymidine kinase and ganciclovir gene therapy as an adjuvant to surgical resection and radiation in adults with previously untreated glioblastoma multiforme. Hum Gene Ther 2000;11(17):2389–401. [DOI] [PubMed] [Google Scholar]
- 120.Lang Frederick F, Bruner Janet M, Fuller Gregory N, et al. Phase I trial of adenovirus-mediated p53 gene therapy for recurrent glioma: biological and clinical results. J Clin Oncol 2003; 21(13):2508–18. [DOI] [PubMed] [Google Scholar]
- 121.Sandmair AM, Loimas S, Puranen P, et al. Thymi-dine kinase gene therapy for human malignant glioma, using replication-deficient retroviruses or adenoviruses. Hum Gene Ther 2000;11(16): 2197–205. [DOI] [PubMed] [Google Scholar]
- 122.Kazuhiko K, Tamiya T, Ono Y, et al. Apoptosis induction with 5-fluorocytosine/cytosine deaminase gene therapy for human malignant glioma cells mediated by adenovirus. J Neurooncol 2004; 66(1–2):117–27. [DOI] [PubMed] [Google Scholar]
- 123.Trask TW, Trask RP, Aguilar-Cordova E, et al. Phase I study of adenoviral delivery of the HSV-tk gene and ganciclovir administration in patients with current malignant brain tumors. Mol Ther 2000;1(2): 195–203. [DOI] [PubMed] [Google Scholar]
- 124.Kieran Mark W, Goumnerova L, Peter M, et al. Phase I study of gene-mediated cytotoxic immunotherapy with AdV-tk as adjuvant to surgery and radiation for pediatric malignant glioma and recurrent ependymoma. Neuro Oncol 2019;21(4):537–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Chang C-J, Hsu C-C, Ming-Chi Y, et al. Enhanced radiosensitivity and radiation-induced apoptosis in glioma CD133-positive cells by knockdown of SirT1 expression. Biochem Biophys Res Commun 2009;380(2):236–42. [DOI] [PubMed] [Google Scholar]
- 126.Cui Q, Yang S, Ye P, et al. Downregulation of TLX induces TET3 expression and inhibits glioblastoma stem cell self-renewal and tumorigenesis. Nat Commun 2016;7:10637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Guffey MB, Parker JN, Luckett WS, et al. Engineered herpes simplex virus expressing bacterial cytosine deaminase for experimental therapy of brain tumors. Cancer Gene Ther 2007;14(1):45–56. [DOI] [PubMed] [Google Scholar]
- 128.Markert James M. A Phase 1 Study of M032 (NSC 733972), a Genetically Engineered HSV-1 Expressing IL-12, in Patients with recurrent/progressive glioblastoma multiforme, Anaplastic Astrocytoma, or Gliosarcoma. - full text view–ClinicalTrials.gov. Available at: https://clinicaltrials.gov/ct2/show/NCT02062827. Accessed November 23, 2020. [DOI] [PMC free article] [PubMed]
- 129.Saha D, Martuza Robert L, Rabkin Samuel D. Oncolytic herpes simplex virus immunovirotherapy in combination with immune checkpoint blockade to treat glioblastoma. Immunotherapy 2018;10(9): 779–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Choi Bryan D, Yu X, Castano Ana P, et al. CAR-T cells secreting BiTEs circumvent antigen escape without detectable toxicity. Nat Biotechnol 2019; 37(9):1049–58. [DOI] [PubMed] [Google Scholar]
- 131.Eleonora P, De Maria Ruggero, Haas TL. Identification of targets to redirect CAR T cells in glioblastoma and colorectal cancer: an arduous venture. Front Immunol 2020;11:565631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Senior M Checkpoint inhibitors go viral. Nat Biotechnol 2019;37(1):12–7. [DOI] [PubMed] [Google Scholar]
- 133.Yin Y, Boesteanu Alina C, Binder Zev A, et al. Checkpoint blockade reverses anergy in IL-13Rα2 humanized scFv-based CAR T cells to treat murine and canine gliomas. Mol Ther Oncolytics 2018;11:20–38. [DOI] [PMC free article] [PubMed] [Google Scholar]