214 results on '"Vile RG"'
Search Results
2. Severe and multifaceted systemic immunosuppression caused by experimental cancers of the central nervous system requires release of non-steroid soluble mediators
- Author
-
Ayasoufi, K, primary, Pfaller, CK, additional, Evgin, L, additional, Khadka, RH, additional, Tritz, ZP, additional, Goddery, EN, additional, Fain, CE, additional, Yokanovich, LT, additional, Himes, BT, additional, Jin, F, additional, Zheng, J, additional, Schuelke, MR, additional, Hansen, MJ, additional, Tung, W, additional, Pease, LR, additional, Vile, RG, additional, and Johnson, AJ, additional
- Published
- 2020
- Full Text
- View/download PDF
3. Rapid adenoviral transduction of freshly resected tumour explants with therapeutically useful genes provides a rationale for genetic immunotherapy for colorectal cancer
- Author
-
Diaz, RM, Todryk, S, Chong, H, Hart, IR, Sikora, K, Dorudi, S, and Vile, RG
- Published
- 1998
- Full Text
- View/download PDF
4. Sticky and smelly issues: lessons on tumour cell and leucocyte trafficking, gene and immunotherapy of cancer
- Author
-
Alexandroff, AB, McIntyre, CA, Porter, JC, Zeuthen, J, Vile, RG, and Taub, DD
- Published
- 1998
- Full Text
- View/download PDF
5. Tumour cell expression of B7 costimulatory molecules and interleukin-12 or granulocyte–macrophage colony-stimulating factor induces a local antitumour response and may generate systemic protective immunity
- Author
-
Chong, H, Todryk, S, Hutchinson, G, Hart, IR, and Vile, RG
- Published
- 1998
- Full Text
- View/download PDF
6. Diverse immunotherapies can effectively treat syngeneic brainstem tumors in the absence of overt toxicity
- Author
-
Schuelke, MR, Wongthida, P, Thompson, J, Kottke, T, Driscoll, CB, Huff, AL, Shim, KG, Coffey, M, Pulido, J, Evgin, L, and Vile, RG
- Abstract
Background: Immunotherapy has shown remarkable clinical promise in the treatment of various types of cancers. However, clinical benefits derive from a highly inflammatory mechanism of action. This presents unique challenges for use in pediatric brainstem tumors including diffuse intrinsic pontine glioma (DIPG), since treatment-related inflammation could cause catastrophic toxicity. Therefore, the goal of this study was to investigate whether inflammatory, immune-based therapies are likely to be too dangerous to pursue for the treatment of pediatric brainstem tumors. Methods: To complement previous immunotherapy studies using patient-derived xenografts in immunodeficient mice, we developed fully immunocompetent models of immunotherapy using transplantable, syngeneic tumors. These four models – HSVtk/GCV suicide gene immunotherapy, oncolytic viroimmunotherapy, adoptive T cell transfer, and CAR T cell therapy – have been optimized to treat tumors outside of the CNS and induce a broad spectrum of inflammatory profiles, maximizing the chances of observing brainstem toxicity. Results: All four models achieved anti-tumor efficacy in the absence of toxicity, with the exception of recombinant vaccinia virus expressing GMCSF, which demonstrated inflammatory toxicity. Histology, imaging, and flow cytometry confirmed the presence of brainstem inflammation in all models. Where used, the addition of immune checkpoint blockade did not introduce toxicity. Conclusions: It remains imperative to regard the brainstem with caution for immunotherapeutic intervention. Nonetheless, we show that further careful development of immunotherapies for pediatric brainstem tumors is warranted to harness the potential potency of anti-tumor immune responses, despite their possible toxicity within this anatomically sensitive location.
- Published
- 2019
7. Potentiating Oncolytic Virus-Induced Immune-Mediated Tumor Cell Killing Using Histone Deacetylase Inhibition
- Author
-
Jennings, VA, Scott, GB, Rose, AMS, Scott, KJ, Migneco, G, Keller, B, Reilly, K, Donnelly, O, Peach, H, Dewar, D, Harrington, KJ, Pandha, H, Samson, A, Vile, RG, AA, M, and Errington-Mais, F
- Subjects
Skin Neoplasms ,Cell Survival ,Genetic Vectors ,Herpesvirus 1, Human ,Antineoplastic Agents, Immunological ,valproic acid ,Antigens, Neoplasm ,Humans ,Simplexvirus ,histone deacetylase inhibitor ,Melanoma ,oncolytic virus ,Oncolytic Virotherapy ,Biological Products ,Granulocyte-Macrophage Colony-Stimulating Factor ,Dendritic Cells ,herpes simplex virus ,HSV,cancer immunotherapy ,Histone Deacetylase Inhibitors ,Killer Cells, Natural ,Oncolytic Viruses ,Interferon Type I ,MCF-7 Cells ,Original Article ,Drug Therapy, Combination ,VPA ,T-Lymphocytes, Cytotoxic - Abstract
A clinical oncolytic herpes simplex virus (HSV) encoding granulocyte-macrophage colony-stimulating factor (GM-CSF), talimogene laherparepvec, causes regression of injected and non-injected melanoma lesions in patients and is now licensed for clinical use in advanced melanoma. To date, limited data are available regarding the mechanisms of human anti-tumor immune priming, an improved understanding of which could inform the development of future combination strategies with improved efficacy. This study addressed direct oncolysis and innate and adaptive human immune-mediated effects of a closely related HSV encoding GM-CSF (HSVGM-CSF) alone and in combination with histone deacetylase inhibition. We found that HSVGM-CSF supported activation of anti-melanoma immunity via monocyte-mediated type I interferon production, which activates NK cells, and viral maturation of immature dendritic cells (iDCs) into potent antigen-presenting cells for cytotoxic T lymphocyte (CTL) priming. Addition of the histone deacetylase inhibitor valproic acid (VPA) to HSVGM-CSF treatment of tumor cells increased viral replication, viral GM-CSF production, and oncolysis and augmented the development of anti-tumor immunity. Mechanistically, VPA increased expression of activating ligands for NK cell recognition and induced expression of tumor-associated antigens, supporting innate NK cell killing and CTL priming. These data support the clinical combination of talimogene laherparepvec with histone deacetylase inhibition to enhance oncolysis and anti-tumor immunity., Jennings et al., demonstrate that oncolytic HSVGM-CSF activates NK cells and matures immature dendritic cells to promote cytotoxic T lymphocyte (CTL) priming. Moreover, they report that histone deacetylase inhibition can augment innate and adaptive anti-tumor immune responses through increased expression of NK cell-activating ligands and tumor-associated antigens.
- Published
- 2019
8. Progress in clinical oncolytic virus-based therapy for hepatocellular carcinoma
- Author
-
Jebar, AH, Errington-Mais, F, Vile, RG, Selby, PJ, Melcher, AA, and Griffin, S
- Abstract
Hepatocellular carcinoma (HCC) carries a dismal prognosis, with advanced disease being resistant to both radiotherapy and conventional cytotoxic drugs, whilst anti-angiogenic drugs are marginally efficacious. Oncolytic viruses (OVs) offer the promise of selective cancer therapy through direct and immune-mediated mechanisms. The premise of OVs lies in their preferential genomic replication, protein expression and productive infection of malignant cells. Numerous OVs are being tested in preclinical models of HCC, with good evidence of direct and immune-mediated antitumour efficacy. Efforts to enhance the performance of these agents have concentrated on engineering OV cellular specificity, immune evasion, enhancing anti-tumour potency and improving delivery. The lead agent in HCC clinical trials, JX-594, a recombinant Wyeth strain vaccinia virus, has demonstrated evidence for significant benefit and earned orphan drug status. Thus, JX-594 appears to be transcending the barrier between novel laboratory science and credible clinical therapy. Relatively few other OVs have entered clinical testing, a hurdle that must be overcome if significant progress is to be made in this field. This review summarizes the preclinical and clinical experience of OV therapy in the difficult-to-treat area of HCC.
- Published
- 2015
9. Oncolytic vaccinia virus as a vector for therapeutic sodium iodide symporter gene therapy in prostate cancer
- Author
-
Mansfield, DC, Kyula, JN, Rosenfelder, N, Chao-Chu, J, Kramer-Marek, G, Khan, AA, Roulstone, V, McLaughlin, M, Melcher, AA, Vile, RG, Pandha, HS, Khoo, V, Harrington, KJ, Mansfield, DC, Kyula, JN, Rosenfelder, N, Chao-Chu, J, Kramer-Marek, G, Khan, AA, Roulstone, V, McLaughlin, M, Melcher, AA, Vile, RG, Pandha, HS, Khoo, V, and Harrington, KJ
- Abstract
Oncolytic strains of vaccinia virus are currently in clinical development with clear evidence of safety and promising signs of efficacy. Addition of therapeutic genes to the viral genome may increase the therapeutic efficacy of vaccinia. We evaluated the therapeutic potential of vaccinia virus expressing the sodium iodide symporter (NIS) in prostate cancer models, combining oncolysis, external beam radiotherapy and NIS-mediated radioiodide therapy. The NIS-expressing vaccinia virus (VV-NIS), GLV-1h153, was tested in in vitro analyzes of viral cell killing, combination with radiotherapy, NIS expression, cellular radioiodide uptake and apoptotic cell death in PC3, DU145, LNCaP and WPMY-1 human prostate cell lines. In vivo experiments were carried out in PC3 xenografts in CD1 nude mice to assess NIS expression and tumor radioiodide uptake. In addition, the therapeutic benefit of radioiodide treatment in combination with viral oncolysis and external beam radiotherapy was measured. In vitro viral cell killing of prostate cancers was dose- and time-dependent and was through apoptotic mechanisms. Importantly, combined virus therapy and iodizing radiation did not adversely affect oncolysis. NIS gene expression in infected cells was functional and mediated uptake of radioiodide both in vitro and in vivo. Therapy experiments with both xenograft and immunocompetent Transgenic Adenocarcinoma of the Mouse Prostate (TRAMP) mouse models showed that the addition of radioiodide to VV-NIS-infected tumors was more effective than each single-agent therapy, restricting tumor growth and increasing survival. In conclusion, VV-NIS is effective in prostate cancer models. This treatment modality would be an attractive complement to existing clinical radiotherapy practice.
- Published
- 2016
10. Expression of B7 co-stimulatory molecules by B16 melanoma results in a natural killer cell-dependent local anti-tumour response, but induces T-cell-dependent systemic immunity only against B7-expressing tumours
- Author
-
Chong, H, primary, Hutchinson, G, additional, Hart, IR, additional, and Vile, RG, additional
- Published
- 1998
- Full Text
- View/download PDF
11. Dendritic cells and T cells deliver oncolytic reovirus for tumour killing despite pre-existing anti-viral immunity.
- Author
-
Ilett, EJ, Prestwich, RJ, Kottke, T., Errington, F., Thompson, JM, Harrington, KJ, Pandha, HS, Coffey, M., Selby, PJ, Vile, RG, and Melcher, AA
- Subjects
TUMORS ,DENDRITIC cells ,T cells ,THERAPEUTICS ,IMMUNE response ,ANTIVIRAL agents - Abstract
Reovirus is a naturally occurring oncolytic virus currently in early clinical trials. However, the rapid induction of neutralizing antibodies represents a major obstacle to successful systemic delivery. This study addresses, for the first time, the ability of cellular carriers in the form of T cells and dendritic cells (DC) to protect reovirus from systemic neutralization. In addition, the ability of these cellular carriers to manipulate the subsequent balance of anti-viral versus anti-tumour immune response is explored. Reovirus, either neat or loaded onto DC or T cells, was delivered intravenously into reovirus-naive or reovirus-immune C57Bl/6 mice bearing lymph node B16tk melanoma metastases. Three and 10 days after treatment, reovirus delivery, carrier cell trafficking, metastatic clearance and priming of anti-tumour/anti-viral immunity were assessed. In naive mice, reovirus delivered either neat or through cell carriage was detectable in the tumour-draining lymph nodes 3 days after treatment, though complete clearance of metastases was only obtained when the virus was delivered on T cells or mature DC (mDC); neat reovirus or loaded immature DC (iDC) gave only partial early tumour clearance. Furthermore, only T cells carrying reovirus generated anti-tumour immune responses and long-term tumour clearance; reovirus-loaded DC, in contrast, generated only an anti-viral immune response. In reovirus-immune mice, however, the results were different. Neat reovirus was completely ineffective as a therapy, whereas mDC—though not iDC—as well as T cells, effectively delivered reovirus to melanoma in vivo for therapy and anti-tumour immune priming. Moreover, mDC were more effective than T cells over a range of viral loads. These data show that systemically administered neat reovirus is not optimal for therapy, and that DC may be an appropriate vehicle for carriage of significant levels of reovirus to tumours. The pre-existing immune status against the virus is critical in determining the balance between anti-viral and anti-tumour immunity elicited when reovirus is delivered by cell carriage, and the viral dose and mode of delivery, as well as the immune status of patients, may profoundly affect the success of any clinical anti-tumour viral therapy. These findings are therefore of direct translational relevance for the future design of clinical trials.Gene Therapy (2009) 16, 689–699; doi:10.1038/gt.2009.29; published online 12 March 2009 [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
12. Targeted radiosensitisation by pegylated liposome-encapsulated 3', 5'-O-dipalmitoyl 5-iodo-2'-deoxyuridine in a head and neck cancer xenograft model.
- Author
-
Harrington, KJ, Syrigos, KN, Uster, PS, Zetter, A, Lewanski, CR, Gullick, WJ, Vile, RG, Stewart, JSW, Harrington, K J, Syrigos, K N, Uster, P S, Lewanski, C R, Gullick, W J, Vile, R G, and Stewart, J S W
- Subjects
TOXICOLOGY ,LIPOSOMES ,HEAD & neck cancer ,XENOGRAFTS ,IMMUNOHISTOCHEMISTRY ,ONCOLOGY - Abstract
5-Iodo-2'-deoxyuridine (IUdR) is an effective radiosensitiser but its clinical development has been limited by toxicity. Prolonged intravenous infusions of IUdR are necessary for optimal tumour uptake but cause dose-limiting myelosuppression. The lack of selective tumour uptake can lead to radiosensitisation of adjacent normal tissues and enhanced local radiation toxicity. Liposomal IUdR delivery offers selective targeting of tumour tissues and avoidance of local and systemic toxicity. In these studies, we report the development of a pegylated liposome containing a lipophilic IUdR derivative (3', 5'-O-dipalmitoyl-5-iodo-2'-deoxyuridine) for use in a head and neck cancer xenograft model. Initial studies confirmed the ability of IUdR to sensitise two head and neck cancer cell lines to single fractions of radiotherapy (SFRT) and this effect was seen to correlate with the thymidine replacement index in KB cells. In vivo delivery of single doses of either unencapsulated IUdR or pegylated liposomal IUdR (PLIUdR) to nude mice bearing KB xenograft tumours did not enhance the effect of SFRT delivered 16 h later. When PLIUdR was delivered by a protracted administration schedule to a dose of 48 mg kg(-1) over 7 days, it enhanced the effect of both 4.5 Gy SFRT and fractionated radiotherapy. PLIUdR was at least as effective as unencapsulated IUdR delivered by multiple intravenous injections or continuous subcutaneous infusion. Immunohistochemistry with a specific anti-IUdR monoclonal antibody confirmed greater levels of tumour staining in tumours from animals treated with PLIUdR compared with those treated with unencapsulated IUdR. [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
13. Intentional killing of normal nelanocytes by gene transfer generates curative anti melanoma immunity without obligate autoimmunity
- Author
-
Daniels, G., Sanchez-Perez, L., Kottke, T., Diaz, Rm, Thompson, J., Lai, M., Michael Gough, Karim, M., Bushell, A., Chong, H., Melcher, A., Harrington, K., and Vile, Rg
14. Immunotherapeutic potential of oncolytic virotherapy.
- Author
-
Prestwich RJ, Harrington KJ, Vile RG, and Melcher AA
- Subjects
- *
GRANULOCYTE-macrophage colony-stimulating factor , *LIVER tumors , *VIRUSES , *ONCOLYTIC virotherapy , *IMMUNOTHERAPY - Published
- 2008
- Full Text
- View/download PDF
15. Malignant melanoma in the 21st century: the emerging molecular landscape.
- Author
-
Sekulic A, Haluska P Jr, Miller AJ, Genebriera De Lamo J, Ejadi S, Pulido JS, Salomao DR, Thorland EC, Vile RG, Swanson DL, Pockaj BA, Laman SD, Pittelkow MR, Markovic SN, Melanoma Study Group of Mayo Clinic Cancer Center, Sekulic, Aleksandar, Haluska, Paul Jr, Miller, Arlo J, Genebriera De Lamo, Josep, and Ejadi, Samuel
- Abstract
Malignant melanoma presents a substantial clinical challenge. Current diagnostic methods are limited in their ability to diagnose early disease and accurately predict individual risk of disease progression and outcome. The lack of adequate approaches to properly define disease subgroups precludes rational treatment design and selection. Better tools are urgently needed to provide more accurate and personalized melanoma patient management. Recent progress in the understanding of the molecular aberrations that underlie melanoma oncogenesis will likely advance the diagnosis, prognosis, and treatment of melanoma. The emerging pattern of molecular complexity in melanoma tumors mirrors the clinical diversity of the disease and highlights the notion that melanoma, like other cancers, is not a single disease but a heterogeneous group of disorders that arise from complex molecular changes. Understanding of molecular aberrations involving important cellular processes, such as cellular signaling networks, cell cycle regulation, and cell death, will be essential for better diagnosis, accurate assessment of prognosis, and rational design of effective therapeutics. Defining an individual patient's unique tumor characteristics may lead to personalized prediction of outcomes and selection of therapy. We review the emerging molecular landscape of melanoma and its implications for better management of patients with melanoma. [ABSTRACT FROM AUTHOR]
- Published
- 2008
16. Intranasal Prime-Boost with Spike Vectors Generates Antibody and T-Cell Responses at the Site of SARS-CoV-2 Infection.
- Author
-
Metko M, Tonne J, Veliz Rios A, Thompson J, Mudrick H, Masopust D, Diaz RM, Barry MA, and Vile RG
- Abstract
Background: Long-lived, re-activatable immunity to SARS-CoV-2 and its emerging variants will rely on T cells recognizing conserved regions of viral proteins across strains. Heterologous prime-boost regimens can elicit elevated levels of circulating CD8+ T cells that provide a reservoir of first responders upon viral infection. Although most vaccines are currently delivered intramuscularly (IM), the initial site of infection is the nasal cavity., Methods: Here, we tested the hypothesis that a heterologous prime and boost vaccine regimen delivered intranasally (IN) will generate improved immune responses locally at the site of virus infection compared to intramuscular vaccine/booster regimens., Results: In a transgenic human ACE2 murine model, both a Spike-expressing single-cycle adenovirus (SC-Ad) and an IFNß safety-enhanced replication-competent Vesicular Stomatitis Virus (VSV) platform generated anti-Spike antibody and T-cell responses that diminished with age. Although SC-Ad-Spike boosted a prime with VSV-Spike-mIFNß, SC-Ad-Spike alone induced maximal levels of IgG, IgA, and CD8+ T-cell responses., Conclusions: There were significant differences in T-cell responses in spleens compared to lungs, and the intranasal boost was significantly superior to the intramuscular boost in generating sentinel immune effectors at the site of the virus encounter in the lungs. These data show that serious consideration should be given to intranasal boosting with anti-SARS-CoV-2 vaccines.
- Published
- 2024
- Full Text
- View/download PDF
17. Smoldering oncolysis by foamy virus carrying CD19 as a CAR target escapes CAR T detection by genomic modification.
- Author
-
Tonne JM, Budzik K, Carrasco TF, Ebbert L, Thompson J, Nace R, Kendall B, Diaz RM, Russell SJ, and Vile RG
- Abstract
Chimeric antigen receptor (CAR) T cells have had limited success against solid tumors. Here, we used an oncolytic foamy virus (oFV) to display a model CAR target antigen (CD19) on tumors in combination with anti-CD19 CAR T cells. We generated oFV-Δ bel2 and oFV- bel2 vectors to test the efficiency and stability of viral/CD19 spread. While both viruses conferred equal CAR T killing in vitro , the oFV-Δ bel2 virus acquired G-to-A mutations, whereas oFV- bel2 virus had genome deletions. In subcutaneous tumor models in vivo , CAR T cells led to a significant decrease in oFV-specific bioluminescence, confirming clearance of oFV-infected tumor cells. However, the most effective therapy was with high-dose oFV in the absence of CAR T cells, indicating that CAR T clearance of oFV was detrimental. Moreover, in tumors that escaped CAR T cell treatment, resurgent virus contained deletions within the oFV-CD19 transgene, allowing the virus to escape CAR T elimination. Therefore, oFV represents a slow smoldering type of oncolytic virus, whose chronic spread through tumors generates anti-tumor therapy, which is abolished by CAR T therapy. These results suggest that further development of this oncolytic platform, with additional immunotherapeutic arming, may allow for an effective combination of chronic oncolysis., Competing Interests: R.V. was a recipient of a research grant from Vyriad Inc., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
18. Direct early growth response-1 knockdown decreases melanoma viability independent of mitogen-activated extracellular signal-related kinase inhibition.
- Author
-
Miley DR, Andrews-Pfannkoch CM, Pulido JS, Erickson SA, Vile RG, Fautsch MP, Marmorstein AD, and Dalvin LA
- Subjects
- Humans, Mitogens, Dimethyl Sulfoxide, Extracellular Signal-Regulated MAP Kinases metabolism, Protein Kinase Inhibitors pharmacology, Mitogen-Activated Protein Kinase Kinases, RNA, Small Interfering, Cell Line, Tumor, Proto-Oncogene Proteins B-raf genetics, Melanoma drug therapy, Melanoma genetics, Melanoma metabolism, Skin Neoplasms drug therapy, Skin Neoplasms genetics, Skin Neoplasms pathology
- Abstract
To investigate downstream molecular changes caused by mitogen-activated protein kinase (MEK) inhibitor treatment and further explore the impact of direct knockdown of early growth response-1 ( EGR1 ) in melanoma cell culture. RNA-sequencing (RNA-Seq) was performed to determine gene expression changes with MEK inhibitor treatment. Treatment with MEK inhibitor (trametinib) was then assessed in two cutaneous (MEL888, MEL624) and one conjunctival (YUARGE 13-3064) melanoma cell line. Direct knockdown of EGR1 was accomplished using lentiviral vectors containing shRNA. Cell viability was measured using PrestoBlueHS Cell Viability Reagent. Total RNA and protein were assessed by qPCR and SimpleWestern. RNA-Seq demonstrated a profound reduction in EGR1 with MEK inhibitor treatment, prompting further study of melanoma cell lines. Following trametinib treatment of melanoma cells, viability was reduced in both cutaneous (MEL888 26%, P < 0.01; MEL624 27%, P < 0.001) and conjunctival (YUARGE 13-3064 33%, P < 0.01) melanoma compared with DMSO control, with confirmed EGR1 knockdown to 0.04-, 0.01-, and 0.16-fold DMSO-treated levels (all P < 0.05) in MEL888, MEL624, and YUARGE 13-3064, respectively. Targeted EGR1 knockdown using shRNA reduced viability in both cutaneous (MEL624 78%, P = 0.05) and conjunctival melanoma (YUARGE-13-3064 67%, P = 0.02). RNA-Sequencing in MEK inhibitor-treated cells identified EGR1 as a candidate effector molecule of interest. In a malignant melanoma cell population, MEK inhibition reduced viability in both cutaneous and conjunctival melanoma with a profound downstream reduction in EGR1 expression. Targeted knockdown of EGR1 reduced both cutaneous and conjunctival melanoma cell viability independent of MEK inhibition, suggesting a key role for EGR1 in melanoma pathobiology., (Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
19. A phase I oncolytic virus trial with vesicular stomatitis virus expressing human interferon beta and tyrosinase related protein 1 administered intratumorally and intravenously in uveal melanoma: safety, efficacy, and T cell responses.
- Author
-
Smith KER, Peng KW, Pulido JS, Weisbrod AJ, Strand CA, Allred JB, Newsom AN, Zhang L, Packiriswamy N, Kottke T, Tonne JM, Moore M, Montane HN, Kottschade LA, McWilliams RR, Dudek AZ, Yan Y, Dimou A, Markovic SN, Federspiel MJ, Vile RG, Dronca RS, and Block MS
- Subjects
- Animals, Humans, Interferon-beta metabolism, Melanoma-Specific Antigens, Monophenol Monooxygenase metabolism, T-Lymphocytes metabolism, Vesicular stomatitis Indiana virus, Oncolytic Virotherapy adverse effects, Oncolytic Viruses genetics, Vesicular Stomatitis
- Abstract
Introduction: Metastatic uveal melanoma (MUM) has a poor prognosis and treatment options are limited. These patients do not typically experience durable responses to immune checkpoint inhibitors (ICIs). Oncolytic viruses (OV) represent a novel approach to immunotherapy for patients with MUM., Methods: We developed an OV with a Vesicular Stomatitis Virus (VSV) vector modified to express interferon-beta (IFN-β) and Tyrosinase Related Protein 1 (TYRP1) (VSV-IFNβ-TYRP1), and conducted a Phase 1 clinical trial with a 3 + 3 design in patients with MUM. VSV-IFNβ-TYRP1 was injected into a liver metastasis, then administered on the same day as a single intravenous (IV) infusion. The primary objective was safety. Efficacy was a secondary objective., Results: 12 patients with previously treated MUM were enrolled. Median follow up was 19.1 months. 4 dose levels (DLs) were evaluated. One patient at DL4 experienced dose limiting toxicities (DLTs), including decreased platelet count (grade 3), increased aspartate aminotransferase (AST), and cytokine release syndrome (CRS). 4 patients had stable disease (SD) and 8 patients had progressive disease (PD). Interferon gamma (IFNγ) ELIspot data showed that more patients developed a T cell response to virus encoded TYRP1 at higher DLs, and a subset of patients also had a response to other melanoma antigens, including gp100, suggesting epitope spreading. 3 of the patients who responded to additional melanoma antigens were next treated with ICIs, and 2 of these patients experienced durable responses., Discussion: Our study found that VSV-IFNβ -TYRP1 can be safely administered via intratumoral (IT) and IV routes in a previously treated population of patients with MUM. Although there were no clear objective radiographic responses to VSV-IFNβ-TYRP1, dose-dependent immunogenicity to TYRP1 and other melanoma antigens was seen., Competing Interests: KP: Imanis Life Sciences stock; Vyriad stock. RM: Zentalis pharmaceuticals consulting. AD: Guardant health advisory board; TP therapeutics advisory board; Novartis clinical trial support; Sorrento therapeutics clinical trial support; Syntrix therapeutics clinical trial support; AnHeart Therapeutics clinical trial support; Merck clinical trial support; Intellisphere LLC honoraria. NP: Imanis life sciences LLCprincipal scientist. LK: Immunocore consulting; Novartis consulting. MS: Bristol-Myers Squibb grant/contract; Sorrento therapeutics grant/contract; other intellectual property. RV: Oncolytics Biotech, Canada scientific advisory board; Greenfire/MGFB scientific advisory board. RD: Bristol Myers Squibb funding; Elsai consulting; Elsevier advisory board; EMD Serono Inc consulting; Genzyme Corporation case series discussion; Immunovaccine Technologies consulting; Natera advisory board; Regeneron Pharmaceuticals consultant; Sanofi Genzyme advisory board. MB: Alkermes grant/contract; Bristol-Meyers Squibb grant/contract; Genentech grant/contract; Immune Design grant/contract; Marker Therapeutics grant/ contract; Merck grant/contract; nFerence grant/contract; Pharmacyclics grant/contract; TILT Biotherapeutics grant/contract; Transgene grant/contract; Viewpoint MolecularTargeting grant/contract. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission., (Copyright © 2023 Smith, Peng, Pulido, Weisbrod, Strand, Allred, Newsom, Zhang, Packiriswamy, Kottke, Tonne, Moore, Montane, Kottschade, McWilliams, Dudek, Yan, Dimou, Markovic, Federspiel, Vile, Dronca and Block.)
- Published
- 2023
- Full Text
- View/download PDF
20. Anti-PD-1 and Extended Half-life IL2 Synergize for Treatment of Murine Glioblastoma Independent of Host MHC Class I Expression.
- Author
-
Tritz ZP, Ayasoufi K, Wolf DM, Owens CA, Malo CS, Himes BT, Fain CE, Goddery EN, Yokanovich LT, Jin F, Hansen MJ, Parney IF, Wang C, Moynihan KD, Irvine DJ, Wittrup KD, Diaz Marcano RM, Vile RG, and Johnson AJ
- Subjects
- Mice, Animals, Interleukin-2 pharmacology, Interleukin-2 therapeutic use, Half-Life, Mice, Inbred C57BL, Cell Line, Tumor, Glioblastoma, Glioma pathology
- Abstract
Glioblastoma (GBM) is the most common malignant brain tumor in adults, responsible for approximately 225,000 deaths per year. Despite preclinical successes, most interventions have failed to extend patient survival by more than a few months. Treatment with anti-programmed cell death protein 1 (anti-PD-1) immune checkpoint blockade (ICB) monotherapy has been beneficial for malignant tumors such as melanoma and lung cancers but has yet to be effectively employed in GBM. This study aimed to determine whether supplementing anti-PD-1 ICB with engineered extended half-life IL2, a potent lymphoproliferative cytokine, could improve outcomes. This combination therapy, subsequently referred to as enhanced checkpoint blockade (ECB), delivered intraperitoneally, reliably cures approximately 50% of C57BL/6 mice bearing orthotopic GL261 gliomas and extends median survival of the treated cohort. In the CT2A model, characterized as being resistant to CBI, ECB caused a decrease in CT2A tumor volume in half of measured animals similar to what was observed in GL261-bearing mice, promoting a trending survival increase. ECB generates robust immunologic responses, features of which include secondary lymphoid organ enlargement and increased activation status of both CD4 and CD8 T cells. This immunity is durable, with long-term ECB survivors able to resist GL261 rechallenge. Through employment of depletion strategies, ECB's efficacy was shown to be independent of host MHC class I-restricted antigen presentation but reliant on CD4 T cells. These results demonstrate ECB is efficacious against the GL261 glioma model through an MHC class I-independent mechanism and supporting further investigation into IL2-supplemented ICB therapies for tumors of the central nervous system., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
21. Current Status and Challenges of Oncolytic Virotherapy for the Treatment of Glioblastoma.
- Author
-
Webb MJ, Sener U, and Vile RG
- Abstract
Despite decades of research and numerous clinical trials, the prognosis of patients diagnosed with glioblastoma (GBM) remains dire with median observed survival at 8 months. There is a critical need for novel treatments for GBM, which is the most common malignant primary brain tumor. Major advances in cancer therapeutics such as immune checkpoint inhibitors and chimeric antigen receptor (CAR) T-cell therapy have not yet led to improved outcomes for GBM. Conventional therapy of surgery followed by chemoradiation with or without tumor treating fields remains the standard of care. One of the many approaches to GBM therapy currently being explored is viral therapies. These typically work by selectively lysing target neoplastic cells, called oncolysis, or by the targeted delivery of a therapeutic transgene via a viral vector. In this review, we discuss the underlying mechanisms of action and describe both recent and current human clinical trials using these viruses with an emphasis on promising viral therapeutics that may ultimately break the field's current stagnant paradigm.
- Published
- 2023
- Full Text
- View/download PDF
22. Oncolytic viruses: Narcissistic or altruistic arsonists?
- Author
-
Kendall L and Vile RG
- Published
- 2023
- Full Text
- View/download PDF
23. Phase I trial of sargramostim/pelareorep therapy in pediatric patients with recurrent or refractory high-grade brain tumors.
- Author
-
Schuelke MR, Gundelach JH, Coffey M, West E, Scott K, Johnson DR, Samson A, Melcher A, Vile RG, and Bram RJ
- Abstract
Background: Brain tumors are the leading cause of cancer death for pediatric patients. Pelareorep, an immunomodulatory oncolytic reovirus, has intravenous efficacy in preclinical glioma models when preconditioned with GM-CSF (sargramostim). We report a phase I trial with the primary goal of evaluating the safety of sargramostim/pelareorep in pediatric patients with recurrent or refractory high-grade brain tumors and a secondary goal of characterizing immunologic responses., Methods: The trial was open to pediatric patients with recurrent or refractory high-grade brain tumors (3 + 3 cohort design). Each cycle included 3 days of subcutaneous sargramostim followed by 2 days of intravenous pelareorep. Laboratory studies and imaging were acquired upon recruitment and periodically thereafter., Results: Six patients participated, including three glioblastoma, two diffuse intrinsic pontine glioma, and one medulloblastoma. Two pelareorep dose levels of 3 × 10
8 and 5 × 108 tissue culture infectious dose 50 (TCID50 ) were assessed. One patient experienced a dose limiting toxicity of persistent hyponatremia. Common low-grade (1 or 2) adverse events included transient fatigue, hypocalcemia, fever, flu-like symptoms, thrombocytopenia, and leukopenia. High-grade (3 or 4) adverse events included neutropenia, lymphopenia, leukopenia, hypophosphatemia, depressed level of consciousness, and confusion. All patients progressed on therapy after a median of 32.5 days and died a median of 108 days after recruitment. Imaging at progression did not show evidence of pseudoprogression or inflammation. Correlative assays revealed transient but consistent changes in immune cells across patients., Conclusions: Sargramostim/pelareorep was administered to pediatric patients with recurrent or refractory high-grade brain tumors. Hyponatremia was the only dose limiting toxicity (DLT), though maximum tolerated dose (MTD) was not determined., (© The Author(s) 2022. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.)- Published
- 2022
- Full Text
- View/download PDF
24. Neoadjuvant Intravenous Oncolytic Vaccinia Virus Therapy Promotes Anticancer Immunity in Patients.
- Author
-
Samson A, West EJ, Carmichael J, Scott KJ, Turnbull S, Kuszlewicz B, Dave RV, Peckham-Cooper A, Tidswell E, Kingston J, Johnpulle M, da Silva B, Jennings VA, Bendjama K, Stojkowitz N, Lusky M, Prasad KR, Toogood GJ, Auer R, Bell J, Twelves CJ, Harrington KJ, Vile RG, Pandha H, Errington-Mais F, Ralph C, Newton DJ, Anthoney A, Melcher AA, and Collinson F
- Subjects
- Humans, Leukocytes, Mononuclear, Neoadjuvant Therapy, Vaccinia virus genetics, Liver Neoplasms therapy, Oncolytic Virotherapy methods, Oncolytic Viruses genetics
- Abstract
Improving the chances of curing patients with cancer who have had surgery to remove metastatic sites of disease is a priority area for cancer research. Pexa-Vec (Pexastimogene Devacirepvec; JX-594, TG6006) is a principally immunotherapeutic oncolytic virus that has reached late-phase clinical trials. We report the results of a single-center, nonrandomized biological end point study (trial registration: EudraCT number 2012-000704-15), which builds on the success of the presurgical intravenous delivery of oncolytic viruses to tumors. Nine patients with either colorectal cancer liver metastases or metastatic melanoma were treated with a single intravenous infusion of Pexa-Vec ahead of planned surgical resection of the metastases. Grade 3 and 4 Pexa-Vec-associated side effects were lymphopaenia and neutropaenia. Pexa-Vec was peripherally carried in plasma and was not associated with peripheral blood mononuclear cells. Upon surgical resection, Pexa-Vec was found in the majority of analyzed tumors. Pexa-Vec therapy associated with IFNα secretion, chemokine induction, and resulted in transient innate and long-lived adaptive anticancer immunity. In the 2 patients with significant and complete tumor necrosis, a reduction in the peripheral T-cell receptor diversity was observed at the time of surgery. These results support the development of presurgical oncolytic vaccinia virus-based therapies to stimulate anticancer immunity and increase the chances to cure patients with cancer., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
25. Oncolytic virus-mediated expansion of dual-specific CAR T cells improves efficacy against solid tumors in mice.
- Author
-
Evgin L, Kottke T, Tonne J, Thompson J, Huff AL, van Vloten J, Moore M, Michael J, Driscoll C, Pulido J, Swanson E, Kennedy R, Coffey M, Loghmani H, Sanchez-Perez L, Olivier G, Harrington K, Pandha H, Melcher A, Diaz RM, and Vile RG
- Subjects
- Animals, Immunotherapy, Adoptive, Mice, Receptors, Antigen, T-Cell, T-Lymphocytes, Tumor Microenvironment, Xenograft Model Antitumor Assays, Glioma therapy, Melanoma therapy, Oncolytic Virotherapy, Oncolytic Viruses physiology, Receptors, Chimeric Antigen
- Abstract
Oncolytic viruses (OVs) encoding a variety of transgenes have been evaluated as therapeutic tools to increase the efficacy of chimeric antigen receptor (CAR)-modified T cells in the solid tumor microenvironment (TME). Here, using systemically delivered OVs and CAR T cells in immunocompetent mouse models, we have defined a mechanism by which OVs can potentiate CAR T cell efficacy against solid tumor models of melanoma and glioma. We show that stimulation of the native T cell receptor (TCR) with viral or virally encoded epitopes gives rise to enhanced proliferation, CAR-directed antitumor function, and distinct memory phenotypes. In vivo expansion of dual-specific (DS) CAR T cells was leveraged by in vitro preloading with oncolytic vesicular stomatitis virus (VSV) or reovirus, allowing for a further in vivo expansion and reactivation of T cells by homologous boosting. This treatment led to prolonged survival of mice with subcutaneous melanoma and intracranial glioma tumors. Human CD19 CAR T cells could also be expanded in vitro with TCR reactivity against viral or virally encoded antigens and was associated with greater CAR-directed cytokine production. Our data highlight the utility of combining OV and CAR T cell therapy and show that stimulation of the native TCR can be exploited to enhance CAR T cell activity and efficacy in mice.
- Published
- 2022
- Full Text
- View/download PDF
26. The spike protein of SARS-CoV-2 induces heme oxygenase-1: Pathophysiologic implications.
- Author
-
Singh RD, Barry MA, Croatt AJ, Ackerman AW, Grande JP, Diaz RM, Vile RG, Agarwal A, and Nath KA
- Subjects
- Animals, COVID-19 virology, Cell Line, Chlorocebus aethiops, HEK293 Cells, Host-Pathogen Interactions drug effects, Host-Pathogen Interactions physiology, Humans, Protein Binding drug effects, Protein Binding physiology, Quercetin pharmacology, Signal Transduction drug effects, Signal Transduction physiology, Up-Regulation drug effects, Up-Regulation physiology, Vero Cells, Virus Internalization drug effects, COVID-19 metabolism, Heme Oxygenase-1 metabolism, SARS-CoV-2 pathogenicity, Spike Glycoprotein, Coronavirus metabolism
- Abstract
Background: Acute kidney injury (AKI) is both a consequence and determinant of outcomes in COVID-19. The kidney is one of the major organs infected by the causative virus, SARS-CoV-2. Viral entry into cells requires the viral spike protein, and both the virus and its spike protein appear in the urine of COVID-19 patients with AKI. We examined the effects of transfecting the viral spike protein of SARS-CoV-2 in kidney cell lines., Methods: HEK293, HEK293-ACE2
+ (stably overexpressing ACE2), and Vero E6 cells having endogenous ACE2 were transfected with SARS-CoV-2 spike or control plasmid. Assessment of gene and protein expression, and syncytia formation was performed, and the effects of quercetin on syncytia formation examined., Findings: Spike transfection in HEK293-ACE2+ cells caused syncytia formation, cellular sloughing, and focal denudation of the cell monolayer; transfection in Vero E6 cells also caused syncytia formation. Spike expression upregulated potentially nephrotoxic genes (TNF-α, MCP-1, and ICAM1). Spike upregulated the cytoprotective gene HO-1 and relevant signaling pathways (p-Akt, p-STAT3, and p-p38). Quercetin, an HO-1 inducer, reduced syncytia formation and spike protein expression., Interpretation: The major conclusions of the study are: 1) Spike protein expression in kidney cells provides a relevant model for the study of maladaptive and adaptive responses germane to AKI in COVID-19; 2) such spike protein expression upregulates HO-1; and 3) quercetin, an HO-1 inducer, may provide a clinically relevant/feasible protective strategy in AKI occurring in the setting of COVID-19., Funding: R01-DK119167 (KAN), R01-AI100911 (JPG), P30-DK079337; R01-DK059600 (AA)., (Copyright © 2021 Elsevier B.V. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
27. Systems neuroimmunology: a review of multiomics methodologies to characterize neuroimmunological interactions in spinal and cranial diseases.
- Author
-
Zamanian C, Bhandarkar AR, Monie DD, Moinuddin FM, Vile RG, Quiñones-Hinojosa A, and Bydon M
- Subjects
- Humans, Machine Learning, Metabolomics methods, Transcriptome, Glioblastoma, Proteomics methods
- Abstract
Neuroimmunology plays a critical role in our understanding of the pathophysiological processes that underlie a variety of diseases treated by neurosurgeons, including degenerative disc disease (DDD), glioblastoma (GBM), aneurysmal subarachnoid hemorrhage (aSAH), and others. Compared with traditional methods in neuroimmunology, which study one pathway or gene at a time, emerging multiomics methodologies allow for holistic interrogation of multiple immune-signaling pathways to test hypotheses and the effects of therapeutics at a systems level. In this review, the authors summarize key concepts for gathering and analyzing multiomics data so that neurosurgeons can contribute to the emerging field of systems neuroimmunology. Additionally, they describe 3 use cases, based on original research published by their group and others, that utilize transcriptomic, metabolomic, and proteomic analyses to study immune-signaling pathways in DDD, aSAH, and GBM. Through these use cases, techniques for performing machine learning and network-based analyses to generate new clinical insights from multiomics data are shared. The authors hope that neurosurgeons might use this review as a summary of common tools and principles in systems immunology to better engage in creating the immunotherapies of tomorrow.
- Published
- 2022
- Full Text
- View/download PDF
28. Inactivation of Replication-Competent Vesicular Stomatitis Virus as SARS-CoV-2 Surrogate on Common Surfaces by Disinfectants.
- Author
-
Pope ZC, Kottke TJ, Shah A, Vile RG, and Rizza SA
- Subjects
- Animals, Humans, SARS-CoV-2, Vesicular stomatitis Indiana virus, COVID-19, Disinfectants pharmacology, Vesicular Stomatitis
- Abstract
Surface disinfection is part of a larger mitigation strategy to prevent the spread of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) virus causing coronavirus disease-2019 (COVID-19). Research evaluating the time, nature, and extent of surface disinfection of replication-competent viruses is needed. We evaluated the efficacy of two disinfectants against a replication-competent SARS-CoV-2 surrogate on three common public surfaces. Vesicular stomatitis virus expressing green fluorescent protein (VSV-GFP) was our replication-competent SARS-CoV-2 surrogate. Disinfection occurred using Super Sani-Cloth Germicidal Disposable Wipes and Oxivir Tb spray per manufacturer instructions to test the efficacy at reducing the presence, viability, and later replication of VSV-GFP on stainless steel, laminate wood, and porcelain surfaces using standardized methods after recovery and toxicity testing. During the main trials, we placed 100 µL spots of VSV-GFP at viral titers of 10
8 , 107 , and 106 PFU/mL on each surface prior to disinfection. Trials were completed in triplicate and post-disinfection measurements on each surface were compared to the measurements of non-disinfected surfaces. Disinfectants were considered efficacious when ≥3-log10 reduction in the number of infectious VSV-GFP virus units was observed on a given surface during all trials. Both disinfectants produced a ≥3.23-log10 reduction in infectious VSV-GFP virus unit numbers, with all trials showing no viable, replication-competent VSV-GFP present on any tested surface. The two disinfectants eliminated the presence, viability, and later replication of VSV-GFP, our SARS-CoV-2 surrogate, on all surfaces. This information suggests that, if following manufacturer instructions, overcleaning surfaces with multiple disinfectant solutions may be unnecessary.- Published
- 2021
- Full Text
- View/download PDF
29. APOBEC and Cancer Viroimmunotherapy: Thinking the Unthinkable.
- Author
-
Vile RG, Melcher A, Pandha H, Harrington KJ, and Pulido JS
- Subjects
- Cytidine Deaminase genetics, Cytidine Deaminase metabolism, Genomics, Humans, Mutagenesis, Mutation, Neoplasms genetics, Neoplasms therapy
- Abstract
The apolipoprotein B mRNA editing enzyme catalytic polypeptide (APOBEC) family protects against infection by degrading incoming viral genomes through cytosine deamination. Here, we review how the potential to unleash these potent DNA mutagens comes at a price as APOBEC DNA mutagenesis can contribute to development of multiple types of cancer. In addition, because viral infection induces its expression, APOBEC is seen as the enemy of oncolytic virotherapy through mutation of the viral genome and by generating virotherapy-resistant tumors. Therefore, overall APOBEC in cancer has received very poor press. However, we also speculate how there may be silver linings to the storm clouds (kataegis) associated with APOBEC activity. Thus, although mutagenic genomic chaos promotes emergence of ever more aggressive subclones, it also provides significant opportunity for cytotoxic and immune therapies. In particular, the superpower of cancer immunotherapy derives in part from mutation, wherein generation of tumor neoantigens-neoantigenesis-exposes tumor cells to functional T-cell repertoires, and susceptibility to immune checkpoint blockade. Moreover, APOBECs may be able to induce suprathreshold levels of cellular mutation leading to mitotic catastrophe and direct tumor cell killing. Finally, we discuss the possibility that linking predictable APOBEC-induced mutation with escape from specific frontline therapies could identify mutated molecules/pathways that can be targeted with small molecules and/or immunotherapies in a Trap and Ambush strategy. Together, these considerations lead to the counterintuitive hypothesis that, instead of attempting to expunge and excoriate APOBEC activity in cancer therapy, it might be exploited-and even, counterintuitively, encouraged., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
30. Modular network mechanism of CCN1-associated resistance to HSV-1-derived oncolytic immunovirotherapies for glioblastomas.
- Author
-
Monie DD, Correia C, Zhang C, Ung CY, Vile RG, and Li H
- Subjects
- Cell Line, Tumor, Humans, Tumor Microenvironment, Brain Neoplasms therapy, Cysteine-Rich Protein 61 metabolism, Glioblastoma therapy, Herpesvirus 1, Human, Oncolytic Virotherapy methods, Oncolytic Viruses
- Abstract
Glioblastomas (GBMs) are the most common and lethal primary brain malignancy in adults. Oncolytic virus (OV) immunotherapies selectively kill GBM cells in a manner that elicits antitumor immunity. Cellular communication network factor 1 (CCN1), a protein found in most GBM microenvironments, expression predicts resistance to OVs, particularly herpes simplex virus type 1 (HSV-1). This study aims to understand how extracellular CCN1 alters the GBM intracellular state to confer OV resistance. Protein-protein interaction network information flow analyses of LN229 human GBM transcriptomes identified 39 novel nodes and 12 binary edges dominating flow in CCN1
high cells versus controls. Virus response programs, notably against HSV-1, and cytokine-mediated signaling pathways are highly enriched. Our results suggest that CCN1high states exploit IDH1 and TP53, and increase dependency on RPL6, HUWE1, and COPS5. To validate, we reproduce our findings in 65 other GBM cell line (CCLE) and 174 clinical GBM patient sample (TCGA) datasets. We conclude through our generalized network modeling and system level analysis that CCN1 signals via several innate immune pathways in GBM to inhibit HSV-1 OVs before transduction. Interventions disrupting this network may overcome immunovirotherapy resistance.- Published
- 2021
- Full Text
- View/download PDF
31. Oncolytic virotherapy induced CSDE1 neo-antigenesis restricts VSV replication but can be targeted by immunotherapy.
- Author
-
Kottke T, Tonne J, Evgin L, Driscoll CB, van Vloten J, Jennings VA, Huff AL, Zell B, Thompson JM, Wongthida P, Pulido J, Schuelke MR, Samson A, Selby P, Ilett E, McNiven M, Roberts LR, Borad MJ, Pandha H, Harrington K, Melcher A, and Vile RG
- Subjects
- Animals, Cancer Vaccines administration & dosage, Cancer Vaccines immunology, Cell Line, Tumor, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Female, Humans, Immunotherapy methods, Interferon-beta metabolism, Mice, Inbred C57BL, Mutation, Oncolytic Viruses metabolism, Oncolytic Viruses physiology, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, T-Lymphocytes immunology, T-Lymphocytes metabolism, Vesicular stomatitis Indiana virus metabolism, Vesicular stomatitis Indiana virus physiology, Mice, DNA-Binding Proteins immunology, Interferon-beta immunology, Oncolytic Virotherapy methods, Oncolytic Viruses immunology, RNA-Binding Proteins immunology, Vesicular stomatitis Indiana virus immunology, Virus Replication immunology
- Abstract
In our clinical trials of oncolytic vesicular stomatitis virus expressing interferon beta (VSV-IFNβ), several patients achieved initial responses followed by aggressive relapse. We show here that VSV-IFNβ-escape tumors predictably express a point-mutated CSDE1
P5S form of the RNA-binding Cold Shock Domain-containing E1 protein, which promotes escape as an inhibitor of VSV replication by disrupting viral transcription. Given time, VSV-IFNβ evolves a compensatory mutation in the P/M Inter-Genic Region which rescues replication in CSDE1P5S cells. These data show that CSDE1 is a major cellular co-factor for VSV replication. However, CSDE1P5S also generates a neo-epitope recognized by non-tolerized T cells. We exploit this predictable neo-antigenesis to drive, and trap, tumors into an escape phenotype, which can be ambushed by vaccination against CSDE1P5S , preventing tumor escape. Combining frontline therapy with escape-targeting immunotherapy will be applicable across multiple therapies which drive tumor mutation/evolution and simultaneously generate novel, targetable immunopeptidomes associated with acquired treatment resistance.- Published
- 2021
- Full Text
- View/download PDF
32. Parking CAR T Cells in Tumours: Oncolytic Viruses as Valets or Vandals?
- Author
-
Evgin L and Vile RG
- Abstract
Oncolytic viruses (OVs) and adoptive T cell therapy (ACT) each possess direct tumour cytolytic capabilities, and their combination potentially seems like a match made in heaven to complement the strengths and weakness of each modality. While providing strong innate immune stimulation that can mobilize adaptive responses, the magnitude of anti-tumour T cell priming induced by OVs is often modest. Chimeric antigen receptor (CAR) modified T cells bypass conventional T cell education through introduction of a synthetic receptor; however, realization of their full therapeutic properties can be stunted by the heavily immune-suppressive nature of the tumour microenvironment (TME). Oncolytic viruses have thus been seen as a natural ally to overcome immunosuppressive mechanisms in the TME which limit CAR T cell infiltration and functionality. Engineering has further endowed viruses with the ability to express transgenes in situ to relieve T cell tumour-intrinsic resistance mechanisms and decorate the tumour with antigen to overcome antigen heterogeneity or loss. Despite this helpful remodeling of the tumour microenvironment, it has simultaneously become clear that not all virus induced effects are favourable for CAR T, begging the question whether viruses act as valets ushering CAR T into their active site, or vandals which cause chaos leading to both tumour and T cell death. Herein, we summarize recent studies combining these two therapeutic modalities and seek to place them within the broader context of viral T cell immunology which will help to overcome the current limitations of effective CAR T therapy to make the most of combinatorial strategies.
- Published
- 2021
- Full Text
- View/download PDF
33. Synthetic and systems biology principles in the design of programmable oncolytic virus immunotherapies for glioblastoma.
- Author
-
Monie DD, Bhandarkar AR, Parney IF, Correia C, Sarkaria JN, Vile RG, and Li H
- Subjects
- Humans, Immunotherapy, Systems Biology, Tumor Microenvironment, Glioblastoma genetics, Glioblastoma therapy, Oncolytic Virotherapy, Oncolytic Viruses genetics
- Abstract
Oncolytic viruses (OVs) are a class of immunotherapeutic agents with promising preclinical results for the treatment of glioblastoma (GBM) but have shown limited success in recent clinical trials. Advanced bioengineering principles from disciplines such as synthetic and systems biology are needed to overcome the current challenges faced in developing effective OV-based immunotherapies for GBMs, including off-target effects and poor clinical responses. Synthetic biology is an emerging field that focuses on the development of synthetic DNA constructs that encode networks of genes and proteins (synthetic genetic circuits) to perform novel functions, whereas systems biology is an analytical framework that enables the study of complex interactions between host pathways and these synthetic genetic circuits. In this review, the authors summarize synthetic and systems biology concepts for developing programmable, logic-based OVs to treat GBMs. Programmable OVs can increase selectivity for tumor cells and enhance the local immunological response using synthetic genetic circuits. The authors discuss key principles for developing programmable OV-based immunotherapies, including how to 1) select an appropriate chassis, a vector that carries a synthetic genetic circuit, and 2) design a synthetic genetic circuit that can be programmed to sense key signals in the GBM microenvironment and trigger release of a therapeutic payload. To illustrate these principles, some original laboratory data are included, highlighting the need for systems biology studies, as well as some preliminary network analyses in preparation for synthetic biology applications. Examples from the literature of state-of-the-art synthetic genetic circuits that can be packaged into leading candidate OV chassis are also surveyed and discussed.
- Published
- 2021
- Full Text
- View/download PDF
34. Ad-CD40L mobilizes CD4 T cells for the treatment of brainstem tumors.
- Author
-
Wongthida P, Schuelke MR, Driscoll CB, Kottke T, Thompson JM, Tonne J, Stone C, Huff AL, Wetmore C, Davies JA, Parker AL, Evgin L, and Vile RG
- Subjects
- Adenoviridae, Animals, CD4-Positive T-Lymphocytes, CD40 Ligand, Humans, Mice, Brain Stem Neoplasms therapy, Glioma therapy
- Abstract
Background: Diffuse midline glioma, formerly DIPG (diffuse intrinsic pontine glioma), is the deadliest pediatric brainstem tumor with median survival of less than one year. Here, we investigated (i) whether direct delivery of adenovirus-expressing cluster of differentiation (CD)40 ligand (Ad-CD40L) to brainstem tumors would induce immune-mediated tumor clearance and (ii) if so, whether therapy would be associated with a manageable toxicity due to immune-mediated inflammation in the brainstem., Methods: Syngeneic gliomas in the brainstems of immunocompetent mice were treated with Ad-CD40L and survival, toxicity, and immune profiles determined. A clinically translatable vector, whose replication would be tightly restricted to tumor cells, rAd-Δ24-CD40L, was tested in human patient-derived diffuse midline gliomas and immunocompetent models., Results: Expression of Ad-CD40L restricted to brainstem gliomas by pre-infection induced complete rejection, associated with immune cell infiltration, of which CD4+ T cells were critical for therapy. Direct intratumoral injection of Ad-CD40L into established brainstem tumors improved survival and induced some complete cures but with some acute toxicity. RNA-sequencing analysis showed that Ad-CD40L therapy induced neuroinflammatory immune responses associated with interleukin (IL)-6, IL-1β, and tumor necrosis factor α. Therefore, to generate a vector whose replication, and transgene expression, would be tightly restricted to tumor cells, we constructed rAd-Δ24-CD40L, the backbone of which has already entered clinical trials for diffuse midline gliomas. Direct intratumoral injection of rAd-Δ24-CD40L, with systemic blockade of IL-6 and IL-1β, generated significant numbers of cures with readily manageable toxicity., Conclusions: Virus-mediated delivery of CD40L has the potential to be effective in treating diffuse midline gliomas without obligatory neuroinflammation-associated toxicity., (© The Author(s) 2020. Published by Oxford University Press on behalf of the Society for Neuro-Oncology.)
- Published
- 2020
- Full Text
- View/download PDF
35. Brain cancer induces systemic immunosuppression through release of non-steroid soluble mediators.
- Author
-
Ayasoufi K, Pfaller CK, Evgin L, Khadka RH, Tritz ZP, Goddery EN, Fain CE, Yokanovich LT, Himes BT, Jin F, Zheng J, Schuelke MR, Hansen MJ, Tung W, Parney IF, Pease LR, Vile RG, and Johnson AJ
- Subjects
- Animals, Bone Marrow Cells immunology, CD4-Positive T-Lymphocytes immunology, Cell Proliferation, Disease Progression, Female, Genes, MHC Class II genetics, Glioblastoma immunology, Glioblastoma metabolism, Glioblastoma pathology, Glioma immunology, Glioma metabolism, Glioma pathology, Male, Melanoma, Experimental immunology, Melanoma, Experimental metabolism, Melanoma, Experimental pathology, Mice, Mice, Inbred C57BL, Parabiosis, Seizures chemically induced, Spleen immunology, Spleen pathology, Theilovirus, Thymus Gland pathology, Brain Neoplasms immunology, Brain Neoplasms metabolism, Immune Tolerance, Inflammation Mediators metabolism
- Abstract
Immunosuppression of unknown aetiology is a hallmark feature of glioblastoma and is characterized by decreased CD4 T-cell counts and downregulation of major histocompatibility complex class II expression on peripheral blood monocytes in patients. This immunosuppression is a critical barrier to the successful development of immunotherapies for glioblastoma. We recapitulated the immunosuppression observed in glioblastoma patients in the C57BL/6 mouse and investigated the aetiology of low CD4 T-cell counts. We determined that thymic involution was a hallmark feature of immunosuppression in three distinct models of brain cancer, including mice harbouring GL261 glioma, B16 melanoma, and in a spontaneous model of diffuse intrinsic pontine glioma. In addition to thymic involution, we determined that tumour growth in the brain induced significant splenic involution, reductions in peripheral T cells, reduced MHC II expression on blood leucocytes, and a modest increase in bone marrow resident CD4 T cells. Using parabiosis we report that thymic involution, declines in peripheral T-cell counts, and reduced major histocompatibility complex class II expression levels were mediated through circulating blood-derived factors. Conversely, T-cell sequestration in the bone marrow was not governed through circulating factors. Serum isolated from glioma-bearing mice potently inhibited proliferation and functions of T cells both in vitro and in vivo. Interestingly, the factor responsible for immunosuppression in serum is non-steroidal and of high molecular weight. Through further analysis of neurological disease models, we determined that the immunosuppression was not unique to cancer itself, but rather occurs in response to brain injury. Non-cancerous acute neurological insults also induced significant thymic involution and rendered serum immunosuppressive. Both thymic involution and serum-derived immunosuppression were reversible upon clearance of brain insults. These findings demonstrate that brain cancers cause multifaceted immunosuppression and pinpoint circulating factors as a target of intervention to restore immunity., (© The Author(s) (2020). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
36. Combining BRAF inhibition with oncolytic herpes simplex virus enhances the immune-mediated antitumor therapy of BRAF-mutant thyroid cancer.
- Author
-
Crespo-Rodriguez E, Bergerhoff K, Bozhanova G, Foo S, Patin EC, Whittock H, Buus R, Haider S, Muirhead G, Thway K, Newbold K, Coffin RS, Vile RG, Kim D, McLaughlin M, Melcher AA, Harrington KJ, and Pedersen M
- Subjects
- Animals, Cell Line, Tumor, Disease Models, Animal, Female, Herpesvirus 1, Human pathogenicity, Humans, Male, Mice, Thyroid Neoplasms pathology, Oncolytic Virotherapy methods, Proto-Oncogene Proteins B-raf metabolism, Thyroid Neoplasms genetics
- Abstract
Background: The aggressive clinical behavior of poorly differentiated and anaplastic thyroid cancers (PDTC and ATC) has proven challenging to treat, and survival beyond a few months from diagnosis is rare. Although 30%-60% of these tumors contain mutations in the BRAF gene, inhibitors designed specifically to target oncogenic BRAF have shown limited and only short-lasting therapeutic benefits as single agents, thus highlighting the need for improved treatment strategies, including novel combinations., Methods: Using a BRAF
V600E -driven mouse model of ATC, we investigated the therapeutic efficacy of the combination of BRAF inhibition and oncolytic herpes simplex virus (oHSV). Analyses of samples from tumor-bearing mice were performed to immunologically characterize the effects of different treatments. These immune data were used to inform the incorporation of immune checkpoint inhibitors into triple combination therapies., Results: We characterized the immune landscape in vivo following BRAF inhibitor treatment and detected only modest immune changes. We, therefore, hypothesized that the addition of oncolytic virotherapy to BRAF inhibition in thyroid cancer would create a more favorable tumor immune microenvironment, boost the inflammatory status of tumors and improve BRAF inhibitor therapy. First, we showed that thyroid cancer cells were susceptible to infection with oHSV and that this process was associated with activation of the immune tumor microenvironment in vivo. Next, we showed improved therapeutic responses when combining oHSV and BRAF inhibition in vivo, although no synergistic effects were seen in vitro, further confirming that the dominant effect of oHSV in this context was likely immune-mediated. Importantly, both gene and protein expression data revealed an increase in activation of T cells and natural killer (NK) cells in the tumor in combination-treated samples. The benefit of combination oHSV and BRAF inhibitor therapy was abrogated when T cells or NK cells were depleted in vivo. In addition, we showed upregulation of PD-L1 and CTLA-4 following combined treatment and demonstrated that blockade of the PD-1/PD-L1 axis or CTLA-4 further improved combination therapy., Conclusions: The combination of oHSV and BRAF inhibition significantly improved survival in a mouse model of ATC by enhancing immune-mediated antitumor effects, and triple combination therapies, including either PD-1 or CTLA-4 blockade, further improved therapy., Competing Interests: Competing interests: RC: employment and share ownership with Replimune. RV: research grant funding Oncolytics Biotech. AM: research grant funding from AstraZeneca/Medimmune, Bristol Myers Squibb and Oncolytics Biotech; advisory board membership, honoraria and/or speakers’ bureaus from Amgen, AstraZeneca, Bristol-Myers-Squibb, Merck-Serono, Turnstone Biologics. KH: research grant funding from AstraZeneca/Medimmune, Boehringer-Ingelheim, Merck Sharp Dohme and Replimune; advisory board membership, honoraria and/or speakers’ bureaus from Amgen, AstraZeneca, Bristol-Myers-Squibb, Boehringer-Ingelheim, Merck-Serono, Merck Sharp Dohme, Oncolys, Pfizer and Replimune., (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2020
- Full Text
- View/download PDF
37. APOBEC3B-mediated corruption of the tumor cell immunopeptidome induces heteroclitic neoepitopes for cancer immunotherapy.
- Author
-
Driscoll CB, Schuelke MR, Kottke T, Thompson JM, Wongthida P, Tonne JM, Huff AL, Miller A, Shim KG, Molan A, Wetmore C, Selby P, Samson A, Harrington K, Pandha H, Melcher A, Pulido JS, Harris R, Evgin L, and Vile RG
- Subjects
- Animals, Cell Line, Tumor, Cytidine Deaminase genetics, Cytidine Deaminase metabolism, Drug Resistance, Neoplasm, Epitopes immunology, Female, Humans, Killer Cells, Natural immunology, Melanoma immunology, Melanoma therapy, Melanoma, Experimental immunology, Melanoma, Experimental therapy, Mice, Inbred C57BL, Minor Histocompatibility Antigens genetics, Minor Histocompatibility Antigens metabolism, Mutation, Tumor Escape drug effects, Cancer Vaccines pharmacology, Cytidine Deaminase immunology, Immunotherapy methods, Minor Histocompatibility Antigens immunology, T-Lymphocytes immunology
- Abstract
APOBEC3B, an anti-viral cytidine deaminase which induces DNA mutations, has been implicated as a mediator of cancer evolution and therapeutic resistance. Mutational plasticity also drives generation of neoepitopes, which prime anti-tumor T cells. Here, we show that overexpression of APOBEC3B in tumors increases resistance to chemotherapy, but simultaneously heightens sensitivity to immune checkpoint blockade in a murine model of melanoma. However, in the vaccine setting, APOBEC3B-mediated mutations reproducibly generate heteroclitic neoepitopes in vaccine cells which activate de novo T cell responses. These cross react against parental, unmodified tumors and lead to a high rate of cures in both subcutaneous and intra-cranial tumor models. Heteroclitic Epitope Activated Therapy (HEAT) dispenses with the need to identify patient specific neoepitopes and tumor reactive T cells ex vivo. Thus, actively driving a high mutational load in tumor cell vaccines increases their immunogenicity to drive anti-tumor therapy in combination with immune checkpoint blockade.
- Published
- 2020
- Full Text
- View/download PDF
38. Generation of a Tumor-Specific Chemokine Gradient Using Oncolytic Vesicular Stomatitis Virus Encoding CXCL9.
- Author
-
Eckert EC, Nace RA, Tonne JM, Evgin L, Vile RG, and Russell SJ
- Abstract
Genetically modified vesicular stomatitis virus (VSV) is an attractive agent for cancer treatment due to rapid intratumoral replication and observed clinical responses. Although VSV selectively kills malignant cells and can boost antitumor immunity, limited induction of intratumoral immune infiltration remains a barrier to efficacy in some cancer models. Here we engineered the oncolytic VSV platform to encode the T cell chemokine CXCL9, which is known to mediate the recruitment of activated CD8
+ cytotoxic T cells and CD4+ T helper cells, and demonstrates conserved protein function between mice and humans. Chemotactic activity of the virally encoded chemokine was confirmed in vitro . Intratumoral concentration of CXCL9 was shown to increase after VSV therapy in three different cancer models, but to a much greater degree after VSV-CXCL9 therapy as compared with VSV control viruses. Despite a steep chemokine gradient from the tumor to the bloodstream, tumor trafficking of adoptively transferred and endogenous T cells was not measurably increased following VSV-CXCL9 therapy. Our results indicate that oncolytic VSV infection promotes release of CXCL9 in the tumor microenvironment, but further boosting of the functional chemokine gradient through virus engineering has little incremental impact on intratumoral immune cell infiltration in mouse and human tumor models., (© 2019 The Authors.)- Published
- 2019
- Full Text
- View/download PDF
39. Diverse immunotherapies can effectively treat syngeneic brainstem tumors in the absence of overt toxicity.
- Author
-
Schuelke MR, Wongthida P, Thompson J, Kottke T, Driscoll CB, Huff AL, Shim KG, Coffey M, Pulido J, Evgin L, and Vile RG
- Subjects
- Animals, Brain Stem Neoplasms genetics, Brain Stem Neoplasms immunology, Cell Line, Tumor, Diffuse Intrinsic Pontine Glioma immunology, Female, Genes, Transgenic, Suicide, Genetic Therapy adverse effects, Granulocyte-Macrophage Colony-Stimulating Factor genetics, Humans, Immunotherapy, Adoptive adverse effects, Mice, Oncolytic Virotherapy adverse effects, Treatment Outcome, Vaccinia virus genetics, Xenograft Model Antitumor Assays, Brain Stem Neoplasms therapy, Diffuse Intrinsic Pontine Glioma therapy, Genetic Therapy methods, Immunotherapy, Adoptive methods, Oncolytic Virotherapy methods, T-Lymphocytes transplantation
- Abstract
Background: Immunotherapy has shown remarkable clinical promise in the treatment of various types of cancers. However, clinical benefits derive from a highly inflammatory mechanism of action. This presents unique challenges for use in pediatric brainstem tumors including diffuse intrinsic pontine glioma (DIPG), since treatment-related inflammation could cause catastrophic toxicity. Therefore, the goal of this study was to investigate whether inflammatory, immune-based therapies are likely to be too dangerous to pursue for the treatment of pediatric brainstem tumors., Methods: To complement previous immunotherapy studies using patient-derived xenografts in immunodeficient mice, we developed fully immunocompetent models of immunotherapy using transplantable, syngeneic tumors. These four models - HSVtk/GCV suicide gene immunotherapy, oncolytic viroimmunotherapy, adoptive T cell transfer, and CAR T cell therapy - have been optimized to treat tumors outside of the CNS and induce a broad spectrum of inflammatory profiles, maximizing the chances of observing brainstem toxicity., Results: All four models achieved anti-tumor efficacy in the absence of toxicity, with the exception of recombinant vaccinia virus expressing GMCSF, which demonstrated inflammatory toxicity. Histology, imaging, and flow cytometry confirmed the presence of brainstem inflammation in all models. Where used, the addition of immune checkpoint blockade did not introduce toxicity., Conclusions: It remains imperative to regard the brainstem with caution for immunotherapeutic intervention. Nonetheless, we show that further careful development of immunotherapies for pediatric brainstem tumors is warranted to harness the potential potency of anti-tumor immune responses, despite their possible toxicity within this anatomically sensitive location.
- Published
- 2019
- Full Text
- View/download PDF
40. Potentiating Oncolytic Virus-Induced Immune-Mediated Tumor Cell Killing Using Histone Deacetylase Inhibition.
- Author
-
Jennings VA, Scott GB, Rose AMS, Scott KJ, Migneco G, Keller B, Reilly K, Donnelly O, Peach H, Dewar D, Harrington KJ, Pandha H, Samson A, Vile RG, Melcher AA, and Errington-Mais F
- Subjects
- Antigens, Neoplasm drug effects, Antigens, Neoplasm metabolism, Antineoplastic Agents, Immunological therapeutic use, Biological Products therapeutic use, Cell Survival genetics, Dendritic Cells immunology, Drug Therapy, Combination, Genetic Vectors, Granulocyte-Macrophage Colony-Stimulating Factor genetics, Herpesvirus 1, Human, Humans, Interferon Type I metabolism, Killer Cells, Natural immunology, MCF-7 Cells, Melanoma pathology, Oncolytic Viruses genetics, Simplexvirus genetics, Skin Neoplasms pathology, T-Lymphocytes, Cytotoxic immunology, Histone Deacetylase Inhibitors therapeutic use, Melanoma therapy, Oncolytic Virotherapy methods, Oncolytic Viruses metabolism, Simplexvirus immunology, Skin Neoplasms therapy, Valproic Acid therapeutic use
- Abstract
A clinical oncolytic herpes simplex virus (HSV) encoding granulocyte-macrophage colony-stimulating factor (GM-CSF), talimogene laherparepvec, causes regression of injected and non-injected melanoma lesions in patients and is now licensed for clinical use in advanced melanoma. To date, limited data are available regarding the mechanisms of human anti-tumor immune priming, an improved understanding of which could inform the development of future combination strategies with improved efficacy. This study addressed direct oncolysis and innate and adaptive human immune-mediated effects of a closely related HSV encoding GM-CSF (HSV
GM-CSF ) alone and in combination with histone deacetylase inhibition. We found that HSVGM-CSF supported activation of anti-melanoma immunity via monocyte-mediated type I interferon production, which activates NK cells, and viral maturation of immature dendritic cells (iDCs) into potent antigen-presenting cells for cytotoxic T lymphocyte (CTL) priming. Addition of the histone deacetylase inhibitor valproic acid (VPA) to HSVGM-CSF treatment of tumor cells increased viral replication, viral GM-CSF production, and oncolysis and augmented the development of anti-tumor immunity. Mechanistically, VPA increased expression of activating ligands for NK cell recognition and induced expression of tumor-associated antigens, supporting innate NK cell killing and CTL priming. These data support the clinical combination of talimogene laherparepvec with histone deacetylase inhibition to enhance oncolysis and anti-tumor immunity., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
41. Suboptimal T-cell Therapy Drives a Tumor Cell Mutator Phenotype That Promotes Escape from First-Line Treatment.
- Author
-
Evgin L, Huff AL, Kottke T, Thompson J, Molan AM, Driscoll CB, Schuelke M, Shim KG, Wongthida P, Ilett EJ, Smith KK, Harris RS, Coffey M, Pulido JS, Pandha H, Selby PJ, Harrington KJ, Melcher A, and Vile RG
- Subjects
- Animals, Antigens, Neoplasm genetics, Antigens, Neoplasm immunology, Cell Line, Tumor, Cytidine Deaminase genetics, Cytidine Deaminase immunology, Female, Ganciclovir therapeutic use, Mammalian orthoreovirus 3, Melanoma, Experimental genetics, Melanoma, Experimental immunology, Mice, Inbred C57BL, Mice, Transgenic, Minor Histocompatibility Antigens genetics, Minor Histocompatibility Antigens immunology, Mutation, Oncolytic Virotherapy, Tumor Escape, CD8-Positive T-Lymphocytes immunology, Immunotherapy, Melanoma, Experimental therapy
- Abstract
Antitumor T-cell responses raised by first-line therapies such as chemotherapy, radiation, tumor cell vaccines, and viroimmunotherapy tend to be weak, both quantitatively (low frequency) and qualitatively (low affinity). We show here that T cells that recognize tumor-associated antigens can directly kill tumor cells if used at high effector-to-target ratios. However, when these tumor-reactive T cells were present at suboptimal ratios, direct T-cell-mediated tumor cell killing was reduced and the ability of tumor cells to evolve away from a coapplied therapy (oncolytic or suicide gene therapy) was promoted. This T-cell-mediated increase in therapeutic resistance was associated with C to T transition mutations that are characteristic of APOBEC3 cytosine deaminase activity and was induced through a TNFα and protein kinase C-dependent pathway. Short hairpin RNA inhibition of endogenous APOBEC3 reduced rates of tumor escape from oncolytic virus or suicide gene therapy to those seen in the absence of antitumor T-cell coculture. Conversely, overexpression of human APOBEC3B in tumor cells enhanced escape from suicide gene therapy and oncolytic virus therapy both in vitro and in vivo Our data suggest that weak affinity or low frequency T-cell responses against tumor antigens may contribute to the ability of tumor cells to evolve away from first-line therapies. We conclude that immunotherapies need to be optimized as early as possible so that, if they do not kill the tumor completely, they do not promote treatment resistance., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
42. Antibody-Neutralized Reovirus Is Effective in Oncolytic Virotherapy.
- Author
-
Berkeley RA, Steele LP, Mulder AA, van den Wollenberg DJM, Kottke TJ, Thompson J, Coffey M, Hoeben RC, Vile RG, Melcher A, and Ilett EJ
- Subjects
- Animals, Cell Line, Chlorocebus aethiops, Female, Humans, Immunoglobulin A immunology, Immunoglobulin G immunology, Mice, Inbred C57BL, Receptors, IgG immunology, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Melanoma, Experimental therapy, Monocytes immunology, Oncolytic Virotherapy, Oncolytic Viruses, Reoviridae
- Abstract
Immunotherapy is showing promise for otherwise incurable cancers. Oncolytic viruses (OVs), developed as direct cytotoxic agents, mediate their antitumor effects via activation of the immune system. However, OVs also stimulate antiviral immune responses, including the induction of OV-neutralizing antibodies. Current dogma suggests that the presence of preexisting antiviral neutralizing antibodies in patients, or their development during viral therapy, is a barrier to systemic OV delivery, rendering repeat systemic treatments ineffective. However, we have found that human monocytes loaded with preformed reovirus-antibody complexes, in which the reovirus is fully neutralized, deliver functional replicative reovirus to tumor cells, resulting in tumor cell infection and lysis. This delivery mechanism is mediated, at least in part, by antibody receptors (in particular FcγRIII) that mediate uptake and internalization of the reovirus/antibody complexes by the monocytes. This finding has implications for oncolytic virotherapy and for the design of clinical OV treatment strategies. Cancer Immunol Res; 6(10); 1161-73. ©2018 AACR ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
43. Ad5 NULL -A20: A Tropism-Modified, αvβ6 Integrin-Selective Oncolytic Adenovirus for Epithelial Ovarian Cancer Therapies.
- Author
-
Uusi-Kerttula H, Davies JA, Thompson JM, Wongthida P, Evgin L, Shim KG, Bradshaw A, Baker AT, Rizkallah PJ, Jones R, Hanna L, Hudson E, Vile RG, Chester JD, and Parker AL
- Subjects
- Adenoviridae genetics, Animals, Carbohydrate Epimerases genetics, Carcinoma, Ovarian Epithelial pathology, Carcinoma, Ovarian Epithelial virology, Cell Line, Tumor, Coxsackie and Adenovirus Receptor-Like Membrane Protein genetics, Female, Genes, cdc genetics, Genetic Vectors genetics, Genetic Vectors pharmacology, Humans, Ketone Oxidoreductases genetics, Mice, Oncolytic Viruses genetics, Tissue Distribution, Transduction, Genetic, Tropism genetics, Antigens, Neoplasm genetics, Carcinoma, Ovarian Epithelial genetics, Carcinoma, Ovarian Epithelial therapy, Integrins genetics, Oncolytic Virotherapy
- Abstract
Purpose: Virotherapies are maturing in the clinical setting. Adenoviruses (Ad) are excellent vectors for the manipulability and tolerance of transgenes. Poor tumor selectivity, off-target sequestration, and immune inactivation hamper clinical efficacy. We sought to completely redesign Ad5 into a refined, tumor-selective virotherapy targeted to αvβ6 integrin, which is expressed in a range of aggressively transformed epithelial cancers but nondetectable in healthy tissues. Experimental Design: Ad5
NULL -A20 harbors mutations in each major capsid protein to preclude uptake via all native pathways. Tumor-tropism via αvβ6 targeting was achieved by genetic insertion of A20 peptide (NAVPNLRGDLQVLAQKVART) within the fiber knob protein. The vector's selectivity in vitro and in vivo was assessed. Results: The tropism-ablating triple mutation completely blocked all native cell entry pathways of Ad5NULL -A20 via coxsackie and adenovirus receptor (CAR), αvβ3/5 integrins, and coagulation factor 10 (FX). Ad5NULL -A20 efficiently and selectively transduced αvβ6+ cell lines and primary clinical ascites-derived EOC ex vivo , including in the presence of preexisting anti-Ad5 immunity. In vivo biodistribution of Ad5NULL -A20 following systemic delivery in non-tumor-bearing mice was significantly reduced in all off-target organs, including a remarkable 107 -fold reduced genome accumulation in the liver compared with Ad5. Tumor uptake, transgene expression, and efficacy were confirmed in a peritoneal SKOV3 xenograft model of human EOC, where oncolytic Ad5NULL -A20-treated animals demonstrated significantly improved survival compared with those treated with oncolytic Ad5. Conclusions: Oncolytic Ad5NULL -A20 virotherapies represent an excellent vector for local and systemic targeting of αvβ6-overexpressing cancers and exciting platforms for tumor-selective overexpression of therapeutic anticancer modalities, including immune checkpoint inhibitors. Clin Cancer Res; 24(17); 4215-24. ©2018 AACR ., (©2018 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
44. APOBEC3 Mediates Resistance to Oncolytic Viral Therapy.
- Author
-
Huff AL, Wongthida P, Kottke T, Thompson JM, Driscoll CB, Schuelke M, Shim KG, Harris RS, Molan A, Pulido JS, Selby PJ, Harrington KJ, Melcher A, Evgin L, and Vile RG
- Abstract
Tumor cells frequently evade applied therapies through the accumulation of genomic mutations and rapid evolution. In the case of oncolytic virotherapy, understanding the mechanisms by which cancer cells develop resistance to infection and lysis is critical to the development of more effective viral-based platforms. Here, we identify APOBEC3 as an important factor that restricts the potency of oncolytic vesicular stomatitis virus (VSV). We show that VSV infection of B16 murine melanoma cells upregulated APOBEC3 in an IFN-β-dependent manner, which was responsible for the evolution of virus-resistant cell populations and suggested that APOBEC3 expression promoted the acquisition of a virus-resistant phenotype. Knockdown of APOBEC3 in B16 cells diminished their capacity to develop resistance to VSV infection in vitro and enhanced the therapeutic effect of VSV in vivo . Similarly, overexpression of human APOBEC3B promoted the acquisition of resistance to oncolytic VSV both in vitro and in vivo . Finally, we demonstrate that APOBEC3B expression had a direct effect on the fitness of VSV, an RNA virus that has not previously been identified as restricted by APOBEC3B. This research identifies APOBEC3 enzymes as key players to target in order to improve the efficacy of viral or broader nucleic acid-based therapeutic platforms.
- Published
- 2018
- Full Text
- View/download PDF
45. White paper on microbial anti-cancer therapy and prevention.
- Author
-
Forbes NS, Coffin RS, Deng L, Evgin L, Fiering S, Giacalone M, Gravekamp C, Gulley JL, Gunn H, Hoffman RM, Kaur B, Liu K, Lyerly HK, Marciscano AE, Moradian E, Ruppel S, Saltzman DA, Tattersall PJ, Thorne S, Vile RG, Zhang HH, Zhou S, and McFadden G
- Subjects
- Animals, Biological Therapy standards, Biological Therapy trends, Cancer Vaccines genetics, Cancer Vaccines immunology, Clinical Studies as Topic, Combined Modality Therapy, Drug Evaluation, Preclinical, Genetic Engineering, Humans, Neoplasms etiology, Oncolytic Virotherapy, Treatment Outcome, Bacteria genetics, Biological Therapy methods, Genetic Vectors genetics, Neoplasms prevention & control, Neoplasms therapy, Viruses genetics
- Abstract
In this White Paper, we discuss the current state of microbial cancer therapy. This paper resulted from a meeting ('Microbial Based Cancer Therapy') at the US National Cancer Institute in the summer of 2017. Here, we define 'Microbial Therapy' to include both oncolytic viral therapy and bacterial anticancer therapy. Both of these fields exploit tumor-specific infectious microbes to treat cancer, have similar mechanisms of action, and are facing similar challenges to commercialization. We designed this paper to nucleate this growing field of microbial therapeutics and increase interactions between researchers in it and related fields. The authors of this paper include many primary researchers in this field. In this paper, we discuss the potential, status and opportunities for microbial therapy as well as strategies attempted to date and important questions that need to be addressed. The main areas that we think will have the greatest impact are immune stimulation, control of efficacy, control of delivery, and safety. There is much excitement about the potential of this field to treat currently intractable cancer. Much of the potential exists because these therapies utilize unique mechanisms of action, difficult to achieve with other biological or small molecule drugs. By better understanding and controlling these mechanisms, we will create new therapies that will become integral components of cancer care.
- Published
- 2018
- Full Text
- View/download PDF
46. The Immune System in Oncolytic Immunovirotherapy: Gospel, Schism and Heresy.
- Author
-
Vile RG
- Subjects
- Immune System, Immunotherapy, Oncolytic Virotherapy, Oncolytic Viruses
- Published
- 2018
- Full Text
- View/download PDF
47. Genetically modified lentiviruses that preserve microvascular function protect against late radiation damage in normal tissues.
- Author
-
Khan AA, Paget JT, McLaughlin M, Kyula JN, Wilkinson MJ, Pencavel T, Mansfield D, Roulstone V, Seth R, Halle M, Somaiah N, Boult JKR, Robinson SP, Pandha HS, Vile RG, Melcher AA, Harris PA, and Harrington KJ
- Subjects
- Animals, Cell Death, Connective Tissue Growth Factor metabolism, Endothelial Cells metabolism, Endothelial Cells radiation effects, Fibrosis, Genetic Therapy, HEK293 Cells, Humans, Magnetic Resonance Imaging, Male, Mitochondria metabolism, Mitochondria radiation effects, Phenotype, Rats, Inbred F344, Reproducibility of Results, Skin pathology, Superoxide Dismutase metabolism, Surgical Flaps blood supply, Transgenes, X-Rays, Lentivirus genetics, Microvessels pathology, Microvessels physiopathology, Radiation Injuries pathology, Radiation Injuries physiopathology
- Abstract
Improvements in cancer survival mean that long-term toxicities, which contribute to the morbidity of cancer survivorship, are being increasingly recognized. Late adverse effects (LAEs) in normal tissues after radiotherapy (RT) are characterized by vascular dysfunction and fibrosis causing volume loss and tissue contracture, for example, in the free flaps used for immediate breast reconstruction after mastectomy. We evaluated the efficacy of lentivirally delivered superoxide dismutase 2 (SOD2) overexpression and connective tissue growth factor (CTGF) knockdown by short hairpin RNA in reducing the severity of LAEs in an animal model of free flap LAEs. Vectors were delivered by intra-arterial injection, ex vivo, to target the vascular compartment. LVSOD2 and LVshCTGF monotherapy before irradiation resulted in preservation of flap volume or reduction in skin contracture, respectively. Flaps transduced with combination therapy experienced improvements in both volume loss and skin contracture. Both therapies reduced the fibrotic burden after irradiation. LAEs were associated with impaired vascular perfusion, loss of endothelial permeability, and stromal hypoxia, which were all reversed in the treatment model. Using a tumor recurrence model, we showed that SOD2 overexpression in normal tissues did not compromise the efficacy of RT against tumor cells but appeared to enhance it. LVSOD2 and LVshCTGF combination therapy by targeted, intravascular delivery reduced LAE severities in normal tissues without compromising the efficacy of RT and warrants translational evaluation as a free flap-targeted gene therapy., (Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2018
- Full Text
- View/download PDF
48. Intravenous delivery of oncolytic reovirus to brain tumor patients immunologically primes for subsequent checkpoint blockade.
- Author
-
Samson A, Scott KJ, Taggart D, West EJ, Wilson E, Nuovo GJ, Thomson S, Corns R, Mathew RK, Fuller MJ, Kottke TJ, Thompson JM, Ilett EJ, Cockle JV, van Hille P, Sivakumar G, Polson ES, Turnbull SJ, Appleton ES, Migneco G, Rose AS, Coffey MC, Beirne DA, Collinson FJ, Ralph C, Alan Anthoney D, Twelves CJ, Furness AJ, Quezada SA, Wurdak H, Errington-Mais F, Pandha H, Harrington KJ, Selby PJ, Vile RG, Griffin SD, Stead LF, Short SC, and Melcher AA
- Subjects
- Animals, Glioma therapy, Humans, Immunotherapy methods, Mice, Mice, Inbred C57BL, Programmed Cell Death 1 Receptor metabolism, Brain Neoplasms therapy, Oncolytic Viruses pathogenicity
- Abstract
Immune checkpoint inhibitors, including those targeting programmed cell death protein 1 (PD-1), are reshaping cancer therapeutic strategies. Evidence suggests, however, that tumor response and patient survival are determined by tumor programmed death ligand 1 (PD-L1) expression. We hypothesized that preconditioning of the tumor immune microenvironment using targeted, virus-mediated interferon (IFN) stimulation would up-regulate tumor PD-L1 protein expression and increase cytotoxic T cell infiltration, improving the efficacy of subsequent checkpoint blockade. Oncolytic viruses (OVs) represent a promising form of cancer immunotherapy. For brain tumors, almost all studies to date have used direct intralesional injection of OV, because of the largely untested belief that intravenous administration will not deliver virus to this site. We show, in a window-of-opportunity clinical study, that intravenous infusion of oncolytic human Orthoreovirus (referred to herein as reovirus) leads to infection of tumor cells subsequently resected as part of standard clinical care, both in high-grade glioma and in brain metastases, and increases cytotoxic T cell tumor infiltration relative to patients not treated with virus. We further show that reovirus up-regulates IFN-regulated gene expression, as well as the PD-1/PD-L1 axis in tumors, via an IFN-mediated mechanism. Finally, we show that addition of PD-1 blockade to reovirus enhances systemic therapy in a preclinical glioma model. These results support the development of combined systemic immunovirotherapy strategies for the treatment of both primary and secondary tumors in the brain., (Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2018
- Full Text
- View/download PDF
49. Inhibitory Receptors Induced by VSV Viroimmunotherapy Are Not Necessarily Targets for Improving Treatment Efficacy.
- Author
-
Shim KG, Zaidi S, Thompson J, Kottke T, Evgin L, Rajani KR, Schuelke M, Driscoll CB, Huff A, Pulido JS, and Vile RG
- Subjects
- Animals, Disease Models, Animal, Female, Hepatitis A Virus Cellular Receptor 2 antagonists & inhibitors, Hepatitis A Virus Cellular Receptor 2 metabolism, Immunologic Memory, Melanoma, Experimental genetics, Melanoma, Experimental immunology, Melanoma, Experimental pathology, Melanoma, Experimental therapy, Mice, Mortality, Neoplasm Metastasis, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor metabolism, Treatment Outcome, Adoptive Transfer, Antigens, Neoplasm genetics, Antigens, Neoplasm immunology, Genetic Vectors genetics, Immunotherapy, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, Vesicular stomatitis Indiana virus genetics
- Abstract
Systemic viroimmunotherapy activates endogenous innate and adaptive immune responses against both viral and tumor antigens. We have shown that therapy with vesicular stomatitis virus (VSV) engineered to express a tumor-associated antigen activates antigen-specific adoptively transferred T cells (adoptive cell therapy, ACT) in vivo to generate effective therapy. The overall goal of this study was to phenotypically characterize the immune response to VSV+ACT therapy and use the information gained to rationally improve combination therapy. We observed rapid expansion of blood CD8
+ effector cells acutely following VSV therapy with markedly high expression of the immune checkpoint molecules PD-1 and TIM-3. Using these data, we tested a treatment schedule incorporating mAb immune checkpoint inhibitors with VSV+ACT treatment. Unlike clinical scenarios, we delivered therapy at early time points following tumor establishment and treatment. Our goal was to potentiate the immune response generated by VSV therapy to achieve durable control of metastatic disease. Despite the high frequency of endogenous PD-1+ TIM-3+ CD8+ T cells following virus administration, antibody blockade did not improve survival. These findings provide highly significant information about response kinetics to viroimmunotherapy and juxtapose the clinical use of checkpoint inhibitors against chronically dysfunctional T cells and the acute T cell response to oncolytic viruses., (Copyright © 2017. Published by Elsevier Inc.)- Published
- 2017
- Full Text
- View/download PDF
50. Socializing Individualized T-Cell Cancer Immunotherapy.
- Author
-
Vile RG
- Subjects
- Humans, Neoplasms, Precision Medicine, Immunotherapy, T-Lymphocytes
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.