Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1702 2023-10-31 17:30:23 |
2 format change Meta information modification 1702 2023-11-01 02:56:15 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Lichtor, T.; Tang, B.; Roy, E.J. Cytokine Gene Vaccine Therapy for Brain Tumor Treatment. Encyclopedia. Available online: https://encyclopedia.pub/entry/51007 (accessed on 17 May 2024).
Lichtor T, Tang B, Roy EJ. Cytokine Gene Vaccine Therapy for Brain Tumor Treatment. Encyclopedia. Available at: https://encyclopedia.pub/entry/51007. Accessed May 17, 2024.
Lichtor, Terry, Bingtao Tang, Edward J. Roy. "Cytokine Gene Vaccine Therapy for Brain Tumor Treatment" Encyclopedia, https://encyclopedia.pub/entry/51007 (accessed May 17, 2024).
Lichtor, T., Tang, B., & Roy, E.J. (2023, October 31). Cytokine Gene Vaccine Therapy for Brain Tumor Treatment. In Encyclopedia. https://encyclopedia.pub/entry/51007
Lichtor, Terry, et al. "Cytokine Gene Vaccine Therapy for Brain Tumor Treatment." Encyclopedia. Web. 31 October, 2023.
Cytokine Gene Vaccine Therapy for Brain Tumor Treatment
Edit

A glioma is a malignant brain tumor with a poor prognosis. Attempts at the surgical removal of the tumor are the first approach, but additional treatment strategies, including radiation therapy and systemic or local chemotherapy, are necessary. Furthermore, the treatments are often associated with significant adverse side effects. Normal and malignant cells generally have antigenic differences, and this is the rationale for clinical immunotherapeutic strategies. Cytokines such as IL-15 or IL-2, which stimulate an anti-tumor immune response, have been shown to have a particularly high potential for use in immunotherapy against various tumors.

immunotherapy IL-2 IL-15 brain tumors prolonged survival

1. Limitation of Current Brain Tumor Treatments

Some increase in the ability to diagnose and surgically treat primary brain tumors has been achieved, although the mortality and overall survival of patients with these tumors has not improved over many years [1]. The present standard treatment modalities, both surgery to remove the tumors and subsequent radiation therapy and chemotherapy, each have significant side effects. The few long-term survivors are inevitably left with cognitive deficits and other disabilities [2][3]. Gliomas are resistant to radiation and standard cytotoxic chemotherapies, making it difficult to treat these tumors [4][5]. Novel therapies are urgently needed.

2. Principles of Brain Tumor Immunology

Normal and malignant cells generally have antigenic differences, which are the basis for clinical immunotherapeutic strategies. Several different strategies have been attempted to enhance anti-tumor immune responses in mice and patients with intracerebral neoplasms. Immunization with dendritic cells that have been “fed” derivatives of tumor cells or transfected with tumor RNA can result in the development of immune responses against the tumor antigens expressed by malignant cells [6][7]. In patients, immunization with autologous dendritic cells, transfected with mRNA from malignant gliomas, has been found to elicit tumor-specific CD8+ cytotoxic T-lymphocyte (CTL) responses against the patient’s malignant cells [8]. Novel and more specific targets, such as glioma stem-like cells, have been shown to increase the success of dendritic cell immunotherapy [9]. Although the results of dendritic cell immunotherapy have demonstrated some good results in animal models, clinical trials have documented few benefits, found to be limited to a minority of treated patients [10].
Another strategy involves the preparation of a vaccine, prepared by the transfer of a cDNA expression library derived from tumor cells into an allogeneic mouse fibroblast cell line expressing a cytokine such as IL-2, which appears to have great potential for the development of an anti-tumor immune response, in the treatment of an intracerebral tumor, in mouse models [11]. Upon the transfer of the cDNA-expression library from the tumor cells into a highly immunogenic fibroblast cell line, genes specifying tumor antigens are expressed. The transferred DNA integrates spontaneously into the genome of the recipient cells and replicates as the cells divide. The transfected fibroblasts can be expanded to obtain quantities for repeated immunizations of the patient. This strategy has the capability of stimulating immunity to a broad array of tumor antigens that characterize the patient’s tumor. Only small amounts of DNA are required to prepare the vaccine, which can be obtained from tumor tissue, such that treatments can be initiated early in the development of the disease.
In many aggressive tumors, such as gliomas, progression is enhanced by local immunosuppression, associated with an increase in regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) [12]. The lack of response to treatment in glioma patients may be secondary to the immunosuppressive T cells that normally prevent an anti-tumor immune response [13]. Various cytokines, including interleukin-10 and transforming growth factor-β, have been implicated in the stimulation of Tregs. The targeting of immune checkpoints which regulate the immune system is emerging as a potent and viable cancer therapy [14]. Immunosuppressive mediators, such as IL-10, TGF-β and prostaglandin, can inhibit the function of the immune system and promote the growth of tumors [15]. Reversing the immunosuppressive tumor microenvironment is one of the keys to the success of tumor treatment.
There are several immunomodulatory cytokines, including IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21, which belong to the family of four α-helix bundle cytokines [16]. The development of IL-2 has been significant in the development of immunotherapy in cancer [17]. However, the use of IL-2 is limited by its toxicity risk and the expansion of regulatory T cells. The associated toxicity issues include hypertension, weight gain, hypothyroidism, cardiac arrhythmias and impaired renal function. To overcome these limitations and improve response rates, other T cell stimulatory agents, such as IL-15, have been in clinical development. IL-15 has been shown to have particularly high potential for use in immunotherapy against various tumors [18]. Furthermore, IL-15, unlike IL-2, does not contribute to the maintenance of regulatory T cells [19].
A variety of tumor vaccination strategies have been attempted, including the modification of neoplastic cells to stimulate anti-tumor immune responses. Immunization with tumor cells, modified to secrete immune-augmenting cytokines such as IL-2, IFN-γ and GM-CSF, has resulted in the development of generalized MHC-restricted anti-tumor immune responses in animal models [20][21][22][23][24][25][26][27][28]. Tumor regression has been documented in experimental animals receiving immunotherapy alone [22][23][27][28], which suggests that this treatment strategy may be effective.

3. Application of Oncolytic Viruses in Brain Tumor Therapy

Oncolytic virus is a type of virus, either engineered or in nature, which may selectively infect and lyse tumor cells while not affecting normal cells [29]. There are many oncolytic viruses, including herpes simplex virus (HSV), adenovirus, reovirus, poliovirus (PV), vaccinia virus (VV), myxoma virus (vMyx), measles virus, vesicular stomatitis virus (VSV) and Newcastle disease virus [30]. There are multiple potential mechanisms contributing to the selectivity of oncolytic viruses for tumor cells over normal cells. First, viruses can enter tumor cells by binding with certain receptors which are overexpressed on tumor cell surfaces. For instance, HSV binds with herpes virus entry mediators (HVEMs) or nectin-1, VV binds with glycosaminoglycans (GAGs) and VSV binds with low-density lipoprotein receptors (LDLRs) to enter host cells [30]. Second, some of the hyper-activated signaling pathways in tumor cells over normal cells may facilitate virus infection. Hyper-activation of AKT (serine/threonine kinase) is commonly found in most cancer cells, which is a requirement for vMyx infection [31][32]. EGFR activation, common in cancer cells, contributes to a productive infection by the attenuated vaccinia virus JX-594 [33]. Third, the deficiency of tumor cells to Type I interferon responses minimize the anti-viral immune responses, allowing oncolytic viruses to replicate [34][35]. Fourth, the dysfunction of tumor suppressor genes, such as p53, ataxia telangiectasia (ATM) and retinoblastoma protein (Rb), can potentially compromise cellular antiviral activity by accumulating genomic instability and blocking the apoptotic response [36], which contributes to the permissiveness of cancer cells.
Once oncolytic viruses infect tumor cells, they may contribute to the anti-tumor response through a direct cytotoxic effect on tumor cells and the consequent release of tumor-associated antigens, which could stimulate anti-tumor immune responses, turning a “cold” tumor into a “hot” tumor [37]. When the virus is engineered to express an immunostimulatory cytokine [38], it becomes a vector for local expression of potent immune-activating agents, attracting immune cells into the tumor microenvironment (TME) while limiting inflammation.
Many oncolytic viruses have already been tested in several preclinical and clinical trials. T-VEC (also known as Talimogene laherparepvec or OncoVEXGM-CSF) was the first oncolytic virus approved for the treatment of advanced melanoma by the U.S. Food and Drug Administration (FDA) in 2015 [39]. T-VEC is an engineered oncolytic herpes simplex virus type 1 (HSV-1), whose neurovirulence factor ICP34.5 is replaced by the gene of human granulocyte–macrophage colony-stimulating factor (hGM-CSF) and whose viral ICP47 gene is deleted [40], to prevent neuronal involvement [41] and enhance anti-tumor efficacy. A OPTiM phase III clinical trial showed great efficacy of T-VEC in targeting patients with early metastatic melanoma (stage IIIB/C-IVM1a) [42]. It also showed enhanced anti-tumor activity when T-VEC was combined with pembrolizumab (anti-programmed death-ligand 1 antibody; PD-1 blockade) in a phase II clinical trial [43]. Furthermore, G47∆, a triple-mutated third-generation oncolytic HSV-1 [44], has a high safety profile and high anti-tumor efficacy (with a 1-year survival rate of 92.3% versus 15%) when targeting human glioblastoma in a phase II clinical trial [45].
Poliovirus is another potential candidate for oncolytic virotherapy. The recombinant nonpathogenic polio–rhinovirus chimera (PVSRIPO) is a neuro-attenuated recombinant poliovirus (Sabin vaccine strains), whose internal ribosomal entry site (IRES) was replaced with human rhinovirus type 2 (HRV2) [46]. The result from a phase I clinical trial, where 61 patients with recurrent World Health Organization (WHO) grade IV malignant gliomas were intratumorally infused with PVSRIPO, confirmed the safety of PVSRIPO when used in the brain, and the patients showed a significantly higher survival rate at 24 and 36 months after virus infusion [47]. Studies also showed that PVSRIPO has the potential to have a therapeutic effect on breast cancer, prostate cancer [48] and neuroblastoma [49].
Poxvirus, a group of large, enveloped DNA viruses associated with diseases that generate poxes in the skin, can also be a good choice for oncolytic virotherapy, since the entire replication of poxvirus happens in viral factories within the cytoplasm of infected cells, with no integration of viral DNA into the host genome, which is safe for host cells [50]. Poxviruses can take multiple large foreign genes into their genomes [51] which supports the feasibility of further arming poxviruses (e.g., adding genes of tumor antigen or immune-enhancing cytokines to poxviruses). The vvDD vaccinia virus is a new strain of poxvirus, which was attenuated by the double deletion of the thymidine kinase and the vaccinia growth factor. A preclinical study showed great anti-tumor efficacy when mice bearing MC38 colon cancer or ID8 ovarian cancer were treated with IL15-armed vvDD vaccinia virus. In addition, when combined with a PD-1 blockade, IL15-armed vvDD vaccinia virus led to dramatic tumor regression [52]. It has been reported that IL15-armed myxoma virus (another poxvirus) cured 83% of mice bearing orthotopic gliomas, when combined with adoptive T cell therapy, rapamycin and celecoxib [53].
Despite the promising results, some concerns still need attention when using oncolytic viruses. Safety issues remain a major concern for oncolytic virotherapy. For example, vvDD vaccinia virus, which has undergone two phase I clinical trials and has been found to be safe in humans [54][55], can still be fatal for hosts if it accidentally enters the cerebral lateral ventricle [56]. Therefore, it is essential to study the safety profile of the oncolytic virus in detail before moving to clinical trials. Another concern is the development of the anti-viral immune responses mediated by neutralizing antibodies [57] and immune cells, such as macrophages [58] and natural killer [59] (NK) cells.

References

  1. Ries, L.A.G.; Kosary, C.L.; Hankey, B.F.; Miller, B.A.; Edwards, B.K. (Eds.) SEER Cancer Statistics Review, 1973–1995; National Cancer Institute: Bethesda, MD, USA, 1988.
  2. Imperato, J.P.; Paleologos, N.A.; Vick, N.A. Effects of treatment on long-term survivors with malignant astrocytomas. Ann. Neurol. 1990, 28, 818–822.
  3. Heimans, J.J.; Taphoorn, M.J. Impact of brain tumor treatment on quality of life. J. Neurol. 2002, 249, 955–960.
  4. Belanich, M.; Randall, T.; Pastor, M.A.; Kibitel, J.T.; Alas, L.G.; Dolan, M.E.; Schold, S.C.; Gander, M., Jr.; Lejeune, F.J.; Li, B.F.; et al. Intracellular localization and intercellular heterogeneity of the human DNA repair protein O(6)-methylguanine-DNA methyltransferase. Cancer Chemother. Pharmacol. 1996, 37, 547–555.
  5. Hotta, T.; Saito, Y.; Fujita, H.; Mikami, T.; Kurisu, K.; Kiya, K.; Uozumi, T.; Isowa, G.; Ishizaki, K.; Ikenaga, M. O6-alkylguanine-DNA alkyltransferase activity of human malignant glioma and its clinical implications. J. Neurooncol. 1994, 21, 135–140.
  6. Insug, O.; Ku, G.; Ertl, H.C.J.; Blaszczyk-Thurin, M. A dendritic cell vaccine induces protective immunity to intracranial growth of glioma. Anticancer. Res. 2002, 22, 613–622.
  7. Rosenberg, S.A.; Yang, J.C.; Restifo, N.P. Cancer immunotherapy: Moving beyond current vaccines. Nat. Med. 2004, 10, 909–915.
  8. Kobayashi, T.; Yamanaka, R.; Homma, J.; Tsuchiya, N.; Yajima, N.; Yoshida, S.; Tanaka, R. Tumor mRNA-loaded dendritic cells elicit tumor-specific CD8+ cytotoxic T cells in patients with malignant glioma. Cancer Immunol. Immunother. 2003, 52, 632–637.
  9. Finocchiaro, G.; Pellegatta, S. Immunotherapy with dendritic cells loaded with glioblastoma stem cells: From preclinical to clinical studies. Cancer Immunol. Immunother. 2016, 65, 101–109.
  10. Reardon, D.A.; Gilbert, M.R.; Wick, W.; Liau, L. Immunotherapy for neuro-oncology: The critical rationale for combinatorial therapy. Neuro Oncol. 2015, 17 (Suppl. S7), vii32–vii40.
  11. Lichtor, T.; Glick, R.P.; Feldman, L.A.; Osawa, G.; Hardman, J.; O-Sullivan, I.; Cohen, E.P. Enhanced immunity to intracerebral breast cancer in mice immunized with a cDNA-based vaccine enriched for immunotherapeutic cells. J. Immunother. 2008, 31, 18–27.
  12. Chang, A.L.; Miska, J.; Wainwright, D.A.; Dey, M.; Rivetta, C.V.; Yu, D.; Kanojia, D.; Pituch, K.C.; Qiao, J.; Pytel, P.; et al. CCL2 Produced by the glioma microenvironment is essential for the recruitment of regulatory T cells and myeloid-derived suppressor cells. Cancer Res. 2016, 76, 5671–5682.
  13. Ooi, Y.C.; Tran, P.; Ung, N.; Thill, K.; Trang, A.; Fong, B.M.; Nagasawa, D.T.; Lim, M.; Yang, I. The role of regulatory T-cells in glioma immunology. Clin. Neurol. Neurosurg. 2014, 119, 125–132.
  14. Lan, Y.; Zhang, D.; Xu, C.; Hance, K.W.; Marelli, B.; Qi, J.; Huakui, Y.; Qin, G.; Sircar, A.; Hernandez, V.M.; et al. Enhanced preclinical antitumor activity of M7824, a bifunctional fusion protein simultaneously targeting PD-Li and TGF-β. Sci. Tansl. Med. 2018, 10, eaan5488.
  15. Mostofa, A.G.M.; Punganuru, S.R.; Madala, H.R.; AL-Obaide, M.; Srivenugopal, K.S. The process and regulatory components of inflammation in brain oncogenesis. Biomolecules 2017, 7, 34.
  16. Steel, J.C.; Waldmann, T.A.; Morris, J.C. Interleukin-15 biology and its therapeutic implications in cancer. Trends Permacol. Sci. 2012, 33, 35–41.
  17. Wrangle, J.M.; Patterson, A.; Johnson, C.B.; Neitzke, D.J.; Mehrotra, S.; Denlinger, C.E.; Paulos, C.M.; Li, Z.; Cole, D.J.; Rubinstein, M.P. IL-2 and beyond in cancer immunotherapy. J. Interferon Cytokine Res. 2018, 38, 45–68.
  18. Cheever, M.A. Twelve immunotherapy drugs that could cure cancers. Immunol. Rev. 2008, 222, 357–368.
  19. Berger, C.; Berger, M.; Hackman, R.C.; Gough, M.; Elliott, C.; Jensen, M.C.; Riddell, S.R. Safety and immunologic effects of IL-15 administration in nonhuman primates. Blood 2009, 114, 2417–2426.
  20. Gansbacher, B.; Bannerji, R.; Daniels, B.; Zier, K.; Cronin, K.; Gilboa, E. Retroviral vector-mediated gamma-interferon gene transfer into tumor cells generates potent and long-lasting antitumor immunity. Cancer Res. 1990, 50, 7820–7825.
  21. Colombo, M.P.; Ferrari, G.; Stoppacciaro, A.; Parenza, M.; Rodolfo, M.; Mavilio, F.; Parmiani, G. Granulocyte colony-stimulating factor gene transfer suppressed tumorigenicity of a murine adenocarcinoma in vivo. J. Exp. Med. 1991, 173, 889–897.
  22. Golumbek, P.T.; Lazenby, A.J.; Levitsky, H.I.; Jaffee, L.M.; Karasuyama, H.; Baker, M.; Pardoll, D.M. Treatment of established renal cancer by tumor cells engineered to secrete interleukin-4. Science 1991, 254, 713–716.
  23. Mullen, C.A.; Coale, M.M.; Levy, A.T.; Stetler-Stevenson, W.G.; Liotta, L.A.; Brandt, S.; Blaese, R.M. Fibrosarcoma cells transduced with the IL-6 gene exhibit reduced tumorigenicity, increased immunogenicity, and decreased metastatic potential. Cancer Res. 1992, 52, 6020–6024.
  24. Dranoff, G.; Jaffee, E.; Lazenby, A.; Golumbek, P.; Levitsky, H.; Brose, K.; Jackson, V.; Hamada, H.; Pardoll, D.; Mulligan, R.C. Vaccination with irradiated tumor cells engineered to secrete murine granulocyte-macrophage colony-stimulating factor stimulates potent, specific and long-lasting anti-tumor immunity. Proc. Nat. Acad. Sci. USA 1993, 90, 3539–3543.
  25. Connor, J.; Bannerji, R.; Saito, S.; Heston, W.; Fair, W.; Gilboa, E. Regression of bladder tumors in mice treated with interleukin 2 gene-modified tumor cells. J. Exp. Med. 1993, 177, 1127–1134.
  26. Cavallo, F.; Pierro, F.D.; Giovarelli, M.; Gulino, A.; Vacca, A.; Stoppacciaro, A.; Forni, M.; Modesti, A.; Forni, G. Protective and curative potential of vaccination with interleukin-2-gene-transfected cells from a spontaneous mouse mammary adenocarcinoma. Cancer Res. 1993, 53, 5067–5070.
  27. Tahara, H.; Zeh, H.J.; Storkus, W.J.; Pappo, I.; Watkins, S.C.; Gubler, U.; Wolf, S.F.; Robbins, P.D.; Lotze, M.T. Fibroblasts genetically engineered to secrete interleukin 12 can suppress tumor growth and induce antitumor immunity to a murine melanoma in vivo. Cancer Res. 1994, 54, 182–189.
  28. Yu, J.S.; Wei, M.X.; Chiocca, E.A.; Martuza, R.L.; Tepper, R.I. Treatment of glioma by engineered interleukin 4-secreting cells. Cancer Res. 1993, 53, 3125–3128.
  29. Bommareddy, P.K.; Shettigar, M.; Kaufman, H.L. Integrating oncolytic viruses in combination cancer immunotherapy. Nat. Rev. Immunol. 2018, 18, 498–513.
  30. Jayawardena, N.; Poirier, J.T.; Burga, L.N.; Bostina, M. Virus-Receptor Interactions and Virus Neutralization: Insights for Oncolytic Virus Development. Oncolytic Virotherapy 2020, 9, 1–15.
  31. Wang, G.; Barrett, J.W.; Stanford, M.; Werden, S.J.; Johnston, J.B.; Gao, X.; Sun, M.; Cheng, J.Q.; McFadden, G. Infection of human cancer cells with myxoma virus requires Akt activation via interaction with a viral ankyrin-repeat host range factor. Proc. Natl. Acad. Sci. USA 2006, 103, 4640–4645.
  32. Werden, S.J.; McFadden, G. The role of cell signaling in poxvirus tropism: The case of the M-T5 host range protein of myxoma virus. Biochim. Biophys. Acta BBA—Proteins Proteom. 2008, 1784, 228–237.
  33. Parato, K.A.; Breitbach, C.J.; Le Boeuf, F.; Wang, J.; Storbeck, C.; Ilkow, C.; Diallo, J.S.; Falls, T.; Burns, J.; Garcia, V.; et al. The oncolytic poxvirus JX-594 selectively replicates in and destroys cancer cells driven by genetic pathways commonly activated in cancers. Mol. Ther. J. Am. Soc. Gene Ther. 2012, 20, 749–758.
  34. Bartee, E.; Mohamed, M.R.; Lopez, M.C.; Baker, H.V.; McFadden, G. The Addition of Tumor Necrosis Factor plus Beta Interferon Induces a Novel Synergistic Antiviral State against Poxviruses in Primary Human Fibroblasts. J. Virol. 2009, 83, 498–511.
  35. Bartee, E.; McFadden, G. Human cancer cells have specifically lost the ability to induce the synergistic state caused by tumor necrosis factor plus interferon-beta. Cytokine 2009, 47, 199–205.
  36. Kim, M.; Williamson, C.T.; Prudhomme, J.; Bebb, D.G.; Riabowol, K.; Lee, P.W.; Lees-Miller, S.P.; Mori, Y.; Rahman, M.M.; McFadden, G.; et al. The viral tropism of two distinct oncolytic viruses, reovirus and myxoma virus, is modulated by cellular tumor suppressor gene status. Oncogene 2010, 29, 3990–3996.
  37. Rahman, M.M.; McFadden, G. Oncolytic Virotherapy with Myxoma Virus. J. Clin. Med. 2020, 9, 171.
  38. Guo, Z.S.; Lu, B.; Guo, Z.; Giehl, E.; Feist, M.; Dai, E.; Liu, W.; Storkus, W.J.; He, Y.; Liu, Z.; et al. Vaccinia virus-mediated cancer immunotherapy: Cancer vaccines and oncolytics. J. Immunother. Cancer 2019, 7, 6.
  39. Johnson, D.B.; Puzanov, I.; Kelley, M.C. Talimogene laherparepvec (T-VEC) for the treatment of advanced melanoma. Immunotherapy 2015, 7, 611–619.
  40. Bommareddy, P.K.; Patel, A.; Hossain, S.; Kaufman, H.L. Talimogene Laherparepvec (T-VEC) and Other Oncolytic Viruses for the Treatment of Melanoma. Am. J. Clin. Dermatol. 2017, 18, 1–15.
  41. Liu, B.L.; Robinson, M.; Han, Z.Q.; Branston, R.H.; English, C.; Reay, P.; McGrath, Y.; Thomas, S.K.; Thornton, M.; Bullock, P.; et al. ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumour properties. Gene Ther. 2003, 10, 292–303.
  42. Franke, V.; Berger, D.M.S.; Klop, W.M.C.; van der Hiel, B.; van de Wiel, B.A.; Ter Meulen, S.; Wouters, M.W.J.M.; van Houdt, W.J.; van Akkooi, A.C.J. High response rates for T-VEC in early metastatic melanoma (stage IIIB/C-IVM1a). Int. J. Cancer 2019, 145, 974–978.
  43. Kelly, C.M.; Antonescu, C.R.; Bowler, T.; Munhoz, R.; Chi, P.; Dickson, M.A.; Gounder, M.M.; Keohan, M.L.; Movva, S.; Dholakia, R.; et al. Objective Response Rate Among Patients with Locally Advanced or Metastatic Sarcoma Treated with Talimogene Laherparepvec in Combination with Pembrolizumab: A Phase 2 Clinical Trial. JAMA Oncol. 2020, 6, 402–408.
  44. Todo, T.; Martuza, R.L.; Rabkin, S.D.; Johnson, P.A. Oncolytic herpes simplex virus vector with enhanced MHC class I presentation and tumor cell killing. Proc. Natl. Acad. Sci. USA 2001, 98, 6396–6401.
  45. Todo, T. ATIM-14. Results of Phase II clinical trial of oncolytic herpes virus G47Δ in patients with glioblastoma. Neuro-Oncol. 2019, 21, vi4.
  46. Gromeier, M.; Alexander, L.; Wimmer, E. Internal ribosomal entry site substitution eliminates neurovirulence in intergeneric poliovirus recombinants. Proc. Natl. Acad. Sci. USA 1996, 93, 2370–2375.
  47. Desjardins, A.; Gromeier, M.; Herndon, J.E., 2nd; Beaubier, N.; Bolognesi, D.P.; Friedman, A.H.; Friedman, H.S.; McSherry, F.; Muscat, A.M.; Nair, S.; et al. Recurrent Glioblastoma Treated with Recombinant Poliovirus. N. Engl. J. Med. 2018, 379, 150–161.
  48. Holl EK Brown, M.C.; Boczkowski, D.; McNamara, M.A.; George, D.J.; Bigner, D.D.; Gromeier, M.; Nair, S.K. Recombinant oncolytic poliovirus, PVSRIPO, has potent cytotoxic and innate inflammatory effects, mediating therapy in human breast and prostate cancer xenograft models. Oncotarget 2016, 7, 79828–79841.
  49. Toyoda, H.; Yin, J.; Mueller, S.; Wimmer, E.; Cello, J. Oncolytic treatment and cure of neuroblastoma by a novel attenuated poliovirus in a novel poliovirus-susceptible animal model. Cancer Res. 2007, 67, 2857–2864.
  50. Fields, B.N.; Knipe, D.M.; Howley, P.M. Fields Virology 2013; Wolters Kluwer Health/Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2013.
  51. Smith, G.L.; Moss, B. Infectious poxvirus vectors have capacity for at least 25,000 base pairs of foreign DNA. Gene 1983, 25, 21–28.
  52. Kowalsky, S.J.; Liu, Z.; Feist, M.; Berkey, S.E.; Ma, C.; Ravindranathan, R.; Dai, E.; Roy, E.J.; Guo, Z.S.; Bartlett, D.L. Superagonist IL-15-armed oncolytic virus elicits potent antitumor immunity and therapy that are enhanced by PD-1 blockade. Mol. Ther. 2018, 26, 2476–2486.
  53. Tang, B.; Guo, Z.S.; Bartlett, D.L.; Yan, D.Z.; Schane, C.P.; Thomas, D.L.; Liu, J.; McFadden, G.; Shisler, J.L.; Roy, E.J. Synergistic combination of oncolytic virotherapy and immunotherapy for glioma. Clin. Cancer Res. 2020, 26, 2216–2230.
  54. Zeh, H.J.; Downs-Canner, S.; McCart, J.A.; Guo, Z.S.; Rao, U.N.; Ramalingam, L.; Thorne, S.H.; Jones, H.L.; Kalinski, P.; Wieckowski, E.; et al. First-in-man study of western reserve strain oncolytic vaccinia virus: Safety, systemic spread, and antitumor activity. Mol. Ther. 2015, 23, 202–214.
  55. Downs-Canner, S.; Guo, Z.S.; Ravindranathan, R.; Breitbach, C.J.; O’Malley, M.E.; Jones, H.L.; Moon, A.; McCart, J.A.; Shuai, Y.; Zeh, H.L.; et al. Phase 1 Study of Intravenous Oncolytic Poxvirus (vvDD) in patients with advanced solid cancers. Mol. Ther. 2016, 24, 1492–1501.
  56. Tang, B.; Guo, Z.S.; Bartlett, D.L.; Liu, J.; McFadden, G.; Shisler, J.L.; Roy, E.J. A cautionary note on the selectivity of oncolytic poxviruses. Oncolytic Virotherapy 2019, 8, 3–8.
  57. Niemann, J.; Woller, N.; Brooks, J.; Fleischmann-Mundt, B.; Martin, N.T.; Kloos, A.; Knocke, S.; Ernst, A.M.; Manns, M.P.; Kubicka, S.; et al. Molecular retargeting of antibodies converts immune defense against oncolytic viruses into cancer immunotherapy. Nat. Commun. 2019, 10, 3236.
  58. Fulci, G.; Dmitrieva, N.; Gianni, D.; Fontana, E.J.; Pan, X.; Lu, Y.; Kaufman, C.S.; Kaur, B.; Lawler, S.E.; Lee, R.J.; et al. Depletion of peripheral macrophages and brain microglia increases brain tumor titers of oncolytic viruses. Cancer Res. 2007, 67, 9398–9406.
  59. Alvarez-Breckenridge, C.A.; Yu, J.; Price, R.; Wojton, J.; Pradarelli, J.; Mao, H.; Wei, M.; Wang, Y.; He, S.; Hardcastle, J.; et al. NK cells impede glioblastoma virotherapy through NKp30 and NKp46 natural cytotoxicity receptors. Nat. Med. 2012, 18, 1827–1834.
More
Information
Subjects: Surgery
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 138
Revisions: 2 times (View History)
Update Date: 01 Nov 2023
1000/1000