Combining Oncolytic Viruses and Small Molecule Therapeutics: Mutual Benefits
Abstract
:Simple Summary
Abstract
1. Introduction
2. Combinations Affecting Viral Propagation in Tumor Cells
2.1. JAK-STAT Signaling Inhibition
2.2. Inhibition of NF-kB Signaling
2.3. PI3K/AKT/mTOR Pathway Antagonists
2.4. Proteasome Inhibitors
2.5. Tankyrase Inhibition
2.6. Receptor Tyrosine Kinase Inhibitor
2.7. Histone Deacetylase Inhibitors (HDIs)
3. Combinations Enhancing Tumor Cell Death
3.1. ER Stress Inducers
3.2. Analogues of DNA Building Blocks
3.3. Antagonizing Inhibitors of Apoptosis (IAPs)
3.4. Microtubule Targeting Compounds
3.5. Topoisomerase Inhibitors
4. Combinations Improving the Antitumor Immune Response
4.1. Cyclophosphamide (CP)
4.2. Inhibitors of VEGF and PDGF Signaling
4.3. Transforming Growth Factor-β TGF-β Inhibition
4.4. Topoisomerase Inhibitors
4.5. Novel Compounds Targeting Adaptive Treatment Resistance of the Tumor
4.6. Checkpoint Inhibitors (CPIs)
4.7. Stimulator of Interferon Genes (STING)
5. Safety Considerations
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef] [Green Version]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
- Kaufman, H.L.; Kohlhapp, F.J.; Zloza, A. Oncolytic viruses: A new class of immunotherapy drugs. Nat. Rev. Drug Discov. 2016, 15, 660. [Google Scholar] [CrossRef] [Green Version]
- Maroun, J.; Muñoz-Alía, M.; Ammayappan, A.; Schulze, A.; Peng, K.W.; Russell, S. Designing and building oncolytic viruses. Future Virol. 2017, 12, 193–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Macedo, N.; Miller, D.M.; Haq, R.; Kaufman, H.L. Clinical landscape of oncolytic virus research in 2020. J. Immunother. Cancer 2020, 8. [Google Scholar] [CrossRef] [PubMed]
- 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, 1–21. [Google Scholar] [CrossRef]
- Rahman, M.M.; McFadden, G. Oncolytic Virotherapy with Myxoma Virus. J. Clin. Med. 2020, 9, 171. [Google Scholar] [CrossRef] [Green Version]
- Bretscher, C.; Marchini, A. H-1 parvovirus as a cancer-killing agent: Past, present, and future. Viruses 2019, 11, 562. [Google Scholar] [CrossRef] [Green Version]
- Müller, L.; Berkeley, R.; Barr, T.; Ilett, E.; Errington-Mais, F. Past, present and future of oncolytic reovirus. Cancers 2020, 12, 3219. [Google Scholar] [CrossRef] [PubMed]
- Pidelaserra-Martí, G.; Engeland, C.E. Mechanisms of measles virus oncolytic immunotherapy. Cytokine Growth Factor Rev. 2020, 56, 28–38. [Google Scholar] [CrossRef]
- Tayeb, S.; Zakay-Rones, Z.; Panet, A. Therapeutic potential of oncolytic Newcastle disease virus: A critical review. Oncolytic Virother. 2015, 4, 49–62. [Google Scholar] [CrossRef] [Green Version]
- Felt, S.A.; Grdzelishvili, V.Z. Ecent advances in vesicular stomatitis virus-based oncolytic virotherapy: A 5-year update. J. Gen. Virol. 2017, 98, 2895–2911. [Google Scholar] [CrossRef] [PubMed]
- Stojdl, D.F.; Lichty, B.; Knowles, S.; Marius, R.; Atkins, H.; Sonenberg, N.; Bell, J.C. Exploiting tumor-specific defects in the interferon pathway with a previously unknown oncolytic virus. Nat. Med. 2000, 6, 821–825. [Google Scholar] [CrossRef] [PubMed]
- Melzer, M.; Lopez-Martinez, A.; Altomonte, J. Oncolytic Vesicular Stomatitis Virus as a Viro-Immunotherapy: Defeating Cancer with a “Hammer” and “Anvil”. Biomedicines 2017, 5, 8. [Google Scholar] [CrossRef]
- Kirn, D.; Martuza, R.L.; Zwiebel, J. Replication-selective virotherapy for cancer: Biological principles, risk management and future directions. Nat. Med. 2001, 7, 781–787. [Google Scholar] [CrossRef]
- Wakimoto, H.; Fulci, G.; Tyminski, E.; Antonio Chiocca, E. Altered expression of antiviral cytokine mRNAs associated with cyclophosphamide’s enhancement of viral oncolysis. Gene Ther. 2004, 11, 214–223. [Google Scholar] [CrossRef] [Green Version]
- Nguyên, T.; Abdelbary, H.; Arguello, M.; Breitbach, C.; Leveille, S.; Diallo, J.-S.; Yasmeen, A.; Bismar, T.A.; Kirn, D.; Falls, T.; et al. Chemical targeting of the innate antiviral response by histone deacetylase inhibitors renders refractory cancers sensitive to viral oncolysis. Proc. Natl. Acad. Sci. USA 2008, 105, 14981–14986. [Google Scholar] [CrossRef] [Green Version]
- Phan, M.; Watson, M.F.; Alain, T.; Diallo, J.S. Oncolytic Viruses on Drugs: Achieving Higher Therapeutic Efficacy. ACS Infect. Dis. 2018, 4, 1448–1467. [Google Scholar] [CrossRef]
- Prestwich, R.J.; Errington, F.; Diaz, R.M.; Pandha, H.S.; Harrington, K.J.; Melcher, A.A.; Vile, R.G. The case of oncolytic viruses versus the immune system: Waiting on the judgment of Solomon. Hum. Gene Ther. 2009, 20, 1119–1132. [Google Scholar] [CrossRef]
- Workenhe, S.T.; Mossman, K.L. Oncolytic virotherapy and immunogenic cancer cell death: Sharpening the sword for improved cancer treatment strategies. Mol. Ther. 2014, 22, 251–256. [Google Scholar] [CrossRef] [Green Version]
- Workenhe, S.T.; Verschoor, M.L.; Mossman, K.L. The role of oncolytic virus immunotherapies to subvert cancer immune evasion. Future Oncol. 2015, 11, 675–689. [Google Scholar] [CrossRef]
- Harrington, K.; Freeman, D.J.; Kelly, B.; Harper, J.; Soria, J.-C. Optimizing oncolytic virotherapy in cancer treatment. Nat. Rev. Drug Discov. 2019, 18, 689–706. [Google Scholar] [CrossRef]
- Van Vloten, J.P.; Workenhe, S.T.; Wootton, S.K.; Mossman, K.L.; Bridle, B.W. Critical Interactions between Immunogenic Cancer Cell Death, Oncolytic Viruses, and the Immune System Define the Rational Design of Combination Immunotherapies. J. Immunol. 2018, 200, 450–458. [Google Scholar] [CrossRef]
- Choi, A.; O’Leary, M.; Fong, Y.; Chen, N. From Benchtop to Bedside: A Review of Oncolytic Virotherapy. Biomedicines 2016, 4, 18. [Google Scholar] [CrossRef]
- Gujar, S.; Pol, J.G.; Kroemer, G. Heating it up: Oncolytic viruses make tumors ‘hot’ and suitable for checkpoint blockade immunotherapies. Oncoimmunology 2018, 7, e1442169. [Google Scholar] [CrossRef]
- Krysko, D.V.; Garg, A.D.; Kaczmarek, A.; Krysko, O.; Agostinis, P.; Vandenabeele, P. Immunogenic cell death and DAMPs in cancer therapy. Nat. Rev. Cancer 2012, 12, 860–875. [Google Scholar] [CrossRef]
- Liikanen, I.; Ahtiainen, L.; Hirvinen, M.L.; Bramante, S.; Cerullo, V.; Nokisalmi, P.; Hemminki, O.; Diaconu, I.; Pesonen, S.; Koski, A.; et al. Oncolytic Adenovirus with Temozolomide Induces Autophagy and Antitumor Immune Responses in Cancer Patients. Mol. Ther. 2013, 21, 1212–1223. [Google Scholar] [CrossRef] [Green Version]
- Komorowski, M.P.; McGray, A.R.; Kolakowska, A.; Eng, K.; Gil, M.; Opyrchal, M.; Litwinska, B.; Nemeth, M.J.; Odunsi, K.O.; Kozbor, D. Reprogramming antitumor immunity against chemoresistant ovarian cancer by a CXCR4 antagonist-armed viral oncotherapy. Mol. Ther. Oncolytics 2016, 3, 16034. [Google Scholar] [CrossRef]
- Errington, F.; Steele, L.; Prestwich, R.; Harrington, K.J.; Pandha, H.S.; Vidal, L.; de Bono, J.; Selby, P.; Coffey, M.; Vile, R.; et al. Reovirus Activates Human Dendritic Cells to Promote Innate Antitumor Immunity. J. Immunol. 2014, 180, 6018–6026. [Google Scholar] [CrossRef] [Green Version]
- Gujar, S.; Dielschneider, R.; Clements, D.; Helson, E.; Shmulevitz, M.; Marcato, P.; Pan, D.; Pan, L.Z.; Ahn, D.G.; Alawadhi, A.; et al. Multifaceted therapeutic targeting of ovarian peritoneal carcinomatosis through virus-induced immunomodulation. Mol. Ther. 2013, 21, 338–347. [Google Scholar] [CrossRef] [Green Version]
- Gujar, S.A.; Lee, P.W.K. Oncolytic Virus-Mediated Reversal of Impaired Tumor Antigen Presentation. Front. Oncol. 2014, 4, 77. [Google Scholar] [CrossRef] [Green Version]
- Liu, Z.; Ravindranathan, R.; Kalinski, P.; Guo, Z.S.; Bartlett, D.L. Rational combination of oncolytic vaccinia virus and PD-L1 blockade works synergistically to enhance therapeutic efficacy. Nat. Commun. 2017, 8, 14754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huck, B.R.; Kötzner, L.; Urbahns, K. Small Molecules Drive Big Improvements in Immuno-Oncology Therapies. Angew. Chem. Int. Ed. 2018, 57, 4412–4428. [Google Scholar] [CrossRef] [Green Version]
- Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017, 168, 707–723. [Google Scholar] [CrossRef] [Green Version]
- Matveeva, O.V.; Chumakov, P.M. Defects in interferon pathways as potential biomarkers of sensitivity to oncolytic viruses. Rev. Med. Virol. 2018, 28, e2008. [Google Scholar] [CrossRef] [PubMed]
- Stojdl, D.F.; Lichty, B.D.; TenOever, B.R.; Paterson, J.M.; Power, A.T.; Knowles, S.; Marius, R.; Reynard, J.; Poliquin, L.; Atkins, H.; et al. VSV strains with defects in their ability to shutdown innate immunity are potent systemic anti-cancer agents. Cancer Cell 2003, 4, 263–275. [Google Scholar] [CrossRef] [Green Version]
- Kurokawa, C.; Galanis, E. Interferon signaling predicts response to oncolytic virotherapy. Oncotarget 2019, 10, 1544–1545. [Google Scholar] [CrossRef] [PubMed]
- Hastie, E.; Cataldi, M.; Moerdyk-Schauwecker, M.J.; Felt, S.A.; Steuerwald, N.; Grdzelishvili, V.Z.; Hastie, E.; Cataldi, M.; Moerdyk-Schauwecker, M.J.; Felt, S.A.; et al. Novel biomarkers of resistance of pancreatic cancer cells to oncolytic vesicular stomatitis virus. Oncotarget 2016, 7, 61601–61618. [Google Scholar] [CrossRef]
- Kurokawa, C.; Iankov, I.D.; Anderson, S.K.; Aderca, I.; Leontovich, A.A.; Maurer, M.J.; Oberg, A.L.; Schroeder, M.A.; Giannini, C.; Greiner, S.M.; et al. Constitutive Interferon Pathway Activation in Tumors as an Efficacy Determinant Following Oncolytic Virotherapy. JNCI J. Natl. Cancer Inst. 2018, 110, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Vähä-Koskela, M.; Hinkkanen, A. Tumor Restrictions to Oncolytic Virus. Biomedicines 2014, 2, 163–194. [Google Scholar] [CrossRef]
- Rojas, J.M.; Alejo, A.; Martín, V.; Sevilla, N. Viral pathogen-induced mechanisms to antagonize mammalian interferon (IFN) signaling pathway. Cell. Mol. Life Sci. 2021, 78, 1423–1444. [Google Scholar] [CrossRef]
- Lin, F.; Young, H.A. Interferons: Success in anti-viral immunotherapy. Cytokine Growth Factor Rev. 2014, 25, 369–376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leonard, W.J.; O’Shea, J.J. JAKS and STATS: Biological Implications. Annu. Rev. Immunol. 2002, 16, 293–322. [Google Scholar] [CrossRef] [Green Version]
- Dold, C.; Rodriguez Urbiola, C.; Wollmann, G.; Egerer, L.; Muik, A.; Bellmann, L.; Fiegl, H.; Marth, C.; Kimpel, J.; von Laer, D. Application of interferon modulators to overcome partial resistance of human ovarian cancers to VSV-GP oncolytic viral therapy. Mol. Ther. Oncolytics 2016, 3, 16021. [Google Scholar] [CrossRef]
- Moerdyk-Schauwecker, M.; Shah, N.R.; Murphy, A.M.; Hastie, E.; Mukherjee, P.; Grdzelishvili, V.Z. Resistance of pancreatic cancer cells to oncolytic vesicular stomatitis virus: Role of type I interferon signaling. Virology 2013, 436, 221–234. [Google Scholar] [CrossRef] [Green Version]
- Cataldi, M.; Shah, N.R.; Felt, S.A.; Grdzelishvili, V.Z. Breaking resistance of pancreatic cancer cells to an attenuated vesicular stomatitis virus through a novel activity of IKK inhibitor TPCA-1. Virology 2015, 485, 340–354. [Google Scholar] [CrossRef] [Green Version]
- Felt, S.A.; Droby, G.N.; Grdzelishvili, V.Z. Ruxolitinib and Polycation Combination Treatment Overcomes Multiple Mechanisms of Resistance of Pancreatic Cancer Cells to Oncolytic Vesicular Stomatitis Virus. J. Virol. 2017, 91, e00461-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Escobar-Zarate, D.; Liu, Y.-P.; Suksanpaisan, L.; Russell, S.J.; Peng, K.-W. Overcoming cancer cell resistance to VSV oncolysis with JAK1/2 inhibitors. Cancer Gene Ther. 2013, 20, 582–589. [Google Scholar] [CrossRef] [Green Version]
- Patel, M.R.; Dash, A.; Jacobson, B.A.; Ji, Y.; Baumann, D.; Ismail, K.; Kratzke, R.A. JAK/STAT inhibition with ruxolitinib enhances oncolytic virotherapy in non-small cell lung cancer models. Cancer Gene Ther. 2019, 26, 411–418. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, T.-T.; Ramsay, L.; Ahanfeshar-Adams, M.; Lajoie, M.; Schadendorf, D.; Alain, T.; Watson, I.R. Mutations in the IFNγ-JAK-STAT Pathway Causing Resistance to Immune Checkpoint Inhibitors in Melanoma Increase Sensitivity to Oncolytic Virus Treatment. Clin. Cancer Res. 2021. [Google Scholar] [CrossRef] [PubMed]
- Jackson, J.D.; Markert, J.M.; Li, L.; Carroll, S.L.; Cassady, K.A. STAT1 and NF- B Inhibitors Diminish Basal Interferon-Stimulated Gene Expression and Improve the Productive Infection of Oncolytic HSV in MPNST Cells. Mol. Cancer Res. 2016, 14, 482–492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Domingo, E.; Guerra, S.; Esteban, M.; Gil, J.; Rivas, C.; Garcia, M.A.; Ventoso, I. Impact of Protein Kinase PKR in Cell Biology: From Antiviral to Antiproliferative Action. Microbiol. Mol. Biol. Rev. 2006, 70, 1032–1060. [Google Scholar] [CrossRef] [Green Version]
- Tamanini, A.; Rolfini, R.; Nicolis, E.; Melotti, P.; Cabrini, G. MAP kinases and NF-κB collaborate to induce ICAM-1 gene expression in the early phase of adenovirus infection. Virology 2003, 307, 228–242. [Google Scholar] [CrossRef] [Green Version]
- Vlahopoulos, S.; Zoumpourlis, V.C. JNK: A key modulator of intracellular signaling. Biochemistry 2004, 69, 844–854. [Google Scholar] [CrossRef] [PubMed]
- Motiwala, A.; Pierce, S.; Satoh, Y.; Bhagwat, S.S.; Bennett, B.L.; Sakata, S.T.; O’Leary, E.C.; Manning, A.M.; Leisten, J.C.; Xu, W.; et al. SP600125, an anthrapyrazolone inhibitor of Jun N-terminal kinase. Proc. Natl. Acad. Sci. USA 2002, 98, 13681–13686. [Google Scholar] [CrossRef] [Green Version]
- Li, B.; Tournier, C.; Davis, R.J.; Flavell, R.A. Regulation of IL-4 expression by the transcription factor JunB during T helper cell differentiation. EMBO J. 1999, 18, 420–432. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Manaligod, J.M.; Bhat, R.K.; Brasier, A.R.; Hershenson, M.B.; Page, K.; Tan, A.; Iasvovskaia, S.; Kartha, S. Regulation of human airway epithelial cell IL-8 expression by MAP kinases. Am. J. Physiol. Cell. Mol. Physiol. 2015, 283, L690–L699. [Google Scholar] [CrossRef] [Green Version]
- Wang, Y.; Li, C.; Wang, X.; Zhang, J.; Chang, Z. Heat shock response inhibits IL-18 expression through the JNK pathway in murine peritoneal macrophages. Biochem. Biophys. Res. Commun. 2002, 296, 742–748. [Google Scholar] [CrossRef]
- Oltmanns, U.; Issa, R.; Sukkar, M.B.; John, M.; Chung, K.F. Role of c-jun N-terminal kinase in the induced release of GM-CSF, RANTES and IL-8 from human airway smooth muscle cells. Br. J. Pharmacol. 2003, 139, 1228–1234. [Google Scholar] [CrossRef] [Green Version]
- Holloway, G.; Coulson, B.S. Rotavirus Activates JNK and p38 Signaling Pathways in Intestinal Cells, Leading to AP-1-Driven Transcriptional Responses and Enhanced Virus Replication. J. Virol. 2006, 80, 10624–10633. [Google Scholar] [CrossRef] [Green Version]
- Hirasawa, K.; Kim, A.; Han, H.-S.; Han, J.; Jun, H.-S.; Yoon, J.-W. Effect of p38 mitogen-activated protein kinase on the replication of encephalomyocarditis virus. J. Virol. 2003, 77, 5649–5656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McLean, T.; Bachenmeier, S.L. Activation of cJUN N-Terminal Kinase by Herpes Simplex Virus Type 1 Enhances Viral Replication. J. Virol. 1999, 73, 8415–8426. [Google Scholar] [CrossRef] [Green Version]
- Hu, W.; Hofstetter, W.; Guo, W.; Li, H.; Pataer, A.; Peng, H.H.; Guo, Z.S.; Bartlett, D.L.; Lin, A.; Swisher, S.G.; et al. JNK-deficiency enhanced oncolytic vaccinia virus replication and blocked activation of double-stranded RNA-dependent protein kinase. Cancer Gene Ther. 2008, 15, 616–624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Perkins, N.D. Integrating cell-signalling pathways with NF-κB and IKK function. Nat. Rev. Mol. Cell Biol. 2007, 8, 49–62. [Google Scholar] [CrossRef]
- Struzik, J.; Szulc-Dąbrowska, L. NF-κB signaling in targeting tumor cells by oncolytic viruses—Therapeutic perspectives. Cancers 2018, 10, 426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Selman, M.; Ou, P.; Rousso, C.; Bergeron, A.; Krishnan, R.; Pikor, L.; Chen, A.; Keller, B.A.; Ilkow, C.; Bell, J.C.; et al. Dimethyl fumarate potentiates oncolytic virotherapy through NF-B inhibition. Sci. Transl. Med. 2018, 10, eaao1613. [Google Scholar] [CrossRef] [Green Version]
- Qiu, D.; Kao, P.N. Immunosuppressive and Anti-Inflammatory Mechanisms of Triptolide, the Principal Active Diterpenoid from the Chinese Medicinal Herb Tripterygium wilfordii Hook. f. Drugs R & D 2006, 4, 1–18. [Google Scholar] [CrossRef]
- Ben Yebdri, F.; Van Grevenynghe, J.; Tang, V.A.; Goulet, M.L.; Wu, J.H.; Stojdl, D.F.; Hiscott, J.; Lin, R. Triptolide-mediated inhibition of interferon signaling enhances vesicular stomatitis virus-based oncolysis. Mol. Ther. 2013, 21, 2043–2053. [Google Scholar] [CrossRef] [Green Version]
- Baumann, J.; Garner, J.M.; Davidoff, A.M.; Morton, C.L.; Du, Z.; Whitt, M.A.; Pfeffer, L.M. Inhibition of Type I Interferon-Mediated Antiviral Action in Human Glioma Cells by the IKK Inhibitors BMS-345541 and TPCA-1. J. Interferon Cytokine Res. 2012, 32, 368–377. [Google Scholar] [CrossRef] [Green Version]
- Wollmann, G.; Ozduman, K.; van den Pol, A.N. Oncolytic virus therapy for glioblastoma multiforme: Concepts and candidates. Cancer J. 2012, 18, 69–81. [Google Scholar] [CrossRef]
- Cao, W.; Manicassamy, S.; Tang, H.; Kasturi, S.P.; Pirani, A.; Murthy, N.; Pulendran, B. Toll-like receptor-mediated induction of type I interferon in plasmacytoid dendritic cells requires the rapamycin-sensitive PI(3)K-mTOR-p70S6K pathway. Nat. Immunol. 2008, 9, 1157–1164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weigelt, B.; Downward, J. Genomic Determinants of PI3K Pathway Inhibitor Response in Cancer. Front. Oncol. 2012, 2, 109. [Google Scholar] [CrossRef] [Green Version]
- Dienstmann, R.; Rodon, J.; Serra, V.; Tabernero, J. Picking the Point of Inhibition: A Comparative Review of PI3K/AKT/mTOR Pathway Inhibitors. Mol. Cancer Ther. 2014, 13, 1021–1031. [Google Scholar] [CrossRef] [Green Version]
- Alain, T.; Lun, X.; Martineau, Y.; Sean, P.; Pulendran, B.; Petroulakis, E.; Zemp, F.J.; Lemay, C.G.; Roy, D.; Bell, J.C.; et al. Vesicular stomatitis virus oncolysis is potentiated by impairing mTORC1-dependent type I IFN production. Proc. Natl. Acad. Sci. USA 2010, 107, 1576–1581. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Meng, S.; Xu, J.; Zhu, Q.; Liu, Q.; Jiang, K.; Zhang, G.; Deng, W.; Wang, Y. Pharmacological modulation of autophagy enhances Newcastle disease virus-mediated oncolysis in drug-resistant lung cancer cells. BMC Cancer 2014, 14, 551. [Google Scholar] [CrossRef] [Green Version]
- Comins, C.; Simpson, G.R.; Rogers, W.; Relph, K.; Harrington, K.; Melcher, A.; Roulstone, V.; Kyula, J.; Pandha, H. Synergistic antitumour effects of rapamycin and oncolytic reovirus. Cancer Gene Ther. 2018, 25, 148–160. [Google Scholar] [CrossRef] [PubMed]
- Zakaria, C.; Sean, P.; Hoang, H.D.; Leroux, L.P.; Watson, M.; Workenhe, S.T.; Hearnden, J.; Pearl, D.; Truong, V.T.; Robichaud, N.; et al. Active-site mTOR inhibitors augment HSV1-dICP0 infection in cancer cells via dysregulated eIF4E/4E-BP axis. PLoS Pathog. 2018, 14. [Google Scholar] [CrossRef]
- Lun, X.; Chan, J.; Zhou, H.; Sun, B.; Kelly, J.J.; Stechishin, O.O.; Bell, J.C.; Parato, K.; Hu, K.; Vaillant, D.; et al. Efficacy and safety/toxicity study of recombinant vaccinia virus JX-594 in two immunocompetent animal models of glioma. Mol. Ther. 2010, 18, 1927–1936. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Reilly, K.E.; Rojo, F.; She, Q.-B.; Solit, D.; Mills, G.B.; Smith, D.; Lane, H.; Hofmann, F.; Hicklin, D.J.; Ludwig, D.L.; et al. mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase Signaling and Activates Akt. Cancer Res. 2006, 66, 1500–1508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xue, Q.L.; Zhou, H.; Alain, T.; Sun, B.; Wang, L.; Barrett, J.W.; Stanford, M.M.; McFadden, G.; Bell, J.; Senger, D.L.; et al. Targeting human medulloblastoma: Oncolytic virotherapy with myxoma virus is enhanced by rapamycin. Cancer Res. 2007, 67, 8818–8827. [Google Scholar] [CrossRef] [Green Version]
- Stanford, M.M.; Shaban, M.; Barrett, J.W.; Werden, S.J.; Gilbert, P.A.; Bondy-Denomy, J.; MacKenzie, L.; Graham, K.C.; Chambers, A.F.; McFadden, G. Myxoma virus oncolysis of primary and metastatic B16F10 mouse tumors in vivo. Mol. Ther. 2008, 16, 52–59. [Google Scholar] [CrossRef]
- Zemp, F.J.; Lun, X.; McKenzie, B.A.; Zhou, H.; Maxwell, L.; Sun, B.; Kelly, J.J.P.; Stechishin, O.; Luchman, A.; Weiss, S.; et al. Treating brain tumor-initiating cells using a combination of myxoma virus and rapamycin. Neuro Oncol. 2013, 15, 904–920. [Google Scholar] [CrossRef] [Green Version]
- Costa-Mattioli, M.; Sonenberg, N. RAPping production of type I interferon in pDCs through mTOR. Nat. Immunol. 2008, 9, 1097–1099. [Google Scholar] [CrossRef]
- Livingstone, M.; Sikström, K.; Robert, P.A.; Uzé, G.; Larsson, O.; Pellegrini, S.P. Assessment of mTOR-Dependent translational regulation of interferon stimulated genes. PLoS ONE 2015, 10, e0133482. [Google Scholar] [CrossRef] [Green Version]
- Lun, X.; Alain, T.; Zemp, F.J.; Zhou, H.; Rahman, M.M.; Hamilton, M.G.; McFadden, G.; Bell, J.; Senger, D.L.; Forsyth, P.A. Myxoma virus virotherapy for glioma in immunocompetent animal models: Optimizing administration routes and synergy with rapamycin. Cancer Res. 2010, 70, 598–608. [Google Scholar] [CrossRef] [Green Version]
- Alonso, M.M.; Jiang, H.; Yokoyama, T.; Xu, J.; Bekele, N.B.; Lang, F.F.; Kondo, S.; Gomez-Manzano, C.; Fueyo, J. Delta-24-RGD in Combination with RAD001 Induces Enhanced Anti-glioma Effect via Autophagic Cell Death. Mol. Ther. 2008, 16, 487–493. [Google Scholar] [CrossRef] [PubMed]
- Alonso, M.M.; Gomez-Manzano, C.; Jiang, H.; Bekele, N.B.; Piao, Y.; Yung, W.K.A.; Alemany, R.; Fueyo, J. Combination of the oncolytic adenovirus ICOVIR-5 with chemotherapy provides enhanced anti-glioma effect in vivo. Cancer Gene Ther. 2007, 14, 756–761. [Google Scholar] [CrossRef]
- Cloughesy, T.F.; Yoshimoto, K.; Nghiemphu, P.; Brown, K.; Dang, J.; Zhu, S.; Hsueh, T.; Chen, Y.; Wang, W.; Youngkin, D.; et al. Antitumor Activity of Rapamycin in a Phase I Trial for Patients with Recurrent PTEN-Deficient Glioblastoma. PLoS Med. 2008, 5, e8. [Google Scholar] [CrossRef]
- Bhagwat, S.V.; Gokhale, P.C.; Crew, A.P.; Cooke, A.; Yao, Y.; Mantis, C.; Kahler, J.; Workman, J.; Bittner, M.; Dudkin, L.; et al. Preclinical Characterization of OSI-027, a Potent and Selective Inhibitor of mTORC1 and mTORC2: Distinct from Rapamycin. Mol. Cancer Ther. 2011, 10, 1394–1406. [Google Scholar] [CrossRef] [Green Version]
- Zaytseva, Y.Y.; Valentino, J.D.; Gulhati, P.; Mark Evers, B. mTOR inhibitors in cancer therapy. Cancer Lett. 2012, 319, 1–7. [Google Scholar] [CrossRef]
- Rodrik-Outmezguine, V.S.; Okaniwa, M.; Yao, Z.; Novotny, C.J.; McWhirter, C.; Banaji, A.; Won, H.; Wong, W.; Berger, M.; de Stanchina, E.; et al. Overcoming mTOR resistance mutations with a new-generation mTOR inhibitor. Nature 2016, 534, 272–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wen, P.Y.; Omuro, A.; Ahluwalia, M.S.; Fathallah-Shaykh, H.M.; Mohile, N.; Lager, J.J.; Laird, A.D.; Tang, J.; Jiang, J.; Egile, C.; et al. Phase I dose-escalation study of the PI3K/mTOR inhibitor voxtalisib (SAR245409, XL765) plus temozolomide with or without radiotherapy in patients with high-grade glioma. Neuro Oncol. 2015, 17, 1275–1283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, L.; Ning, J.; Wakimoto, H.; Wu, S.; Wu, C.L.; Humphrey, M.R.; Rabkin, S.D.; Martuza, R.L. Oncolytic Herpes Simplex Virus and PI3K Inhibitor BKM120 Synergize to Promote Killing of Prostate Cancer Stem-like Cells. Mol. Ther. Oncolytics 2019, 13, 58–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kanai, R.; Wakimoto, H.; Martuza, R.L.; Rabkin, S.D. A novel oncolytic herpes simplex virus that synergizes with phosphoinositide 3-kinase/Akt pathway inhibitors to target glioblastoma stem cells. Clin. Cancer Res. 2011, 17, 3686–3696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, T.C.; Wakimoto, H.; Martuza, R.L.; Rabkin, S.D. Herpes simplex virus Us3(-) mutant as oncolytic strategy and synergizes with phosphatidylinositol 3-kinase-Akt-targeting molecular therapeutics. Clin. Cancer Res. 2007, 13, 5897–5902. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ferguson, M.S.; Chard Dunmall, L.S.; Gangeswaran, R.; Marelli, G.; Tysome, J.R.; Burns, E.; Whitehead, M.A.; Aksoy, E.; Alusi, G.; Hiley, C.; et al. Transient Inhibition of PI3Kδ Enhances the Therapeutic Effect of Intravenous Delivery of Oncolytic Vaccinia Virus. Mol. Ther. 2020, 28, 1263–1275. [Google Scholar] [CrossRef] [Green Version]
- Yoo, J.Y.; Hurwitz, B.S.; Bolyard, C.; Yu, J.G.; Zhang, J.; Selvendiran, K.; Rath, K.S.; He, S.; Bailey, Z.; Eaves, D.; et al. Bortezomib-induced unfolded protein response increases oncolytic HSV-1 replication resulting in synergistic antitumor effects. Clin. Cancer Res. 2014, 20, 3787–3798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yoo, J.Y.; Jaime-Ramirez, A.C.; Bolyard, C.; Dai, H.; Nallanagulagari, T.; Wojton, J.; Hurwitz, B.S.; Relation, T.; Lee, T.J.; Lotze, M.T.; et al. Bortezomib treatment sensitizes oncolytic HSV-1-treated tumors to NK cell immunotherapy. Clin. Cancer Res. 2016, 22, 5265–5276. [Google Scholar] [CrossRef] [Green Version]
- Kim, Y.; Yoo, J.Y.; Lee, T.J.; Liu, J.; Yu, J.; Caligiuri, M.A.; Kaur, B.; Friedman, A. Complex role of NK cells in regulation of oncolytic virus–bortezomib therapy. Proc. Natl. Acad. Sci. USA 2018, 115, 4927–4932. [Google Scholar] [CrossRef] [Green Version]
- Yarde, D.N.; Nace, R.A.; Russell, S.J. Oncolytic vesicular stomatitis virus and bortezomib are antagonistic against myeloma cells invitro but have additive anti-myeloma activity in vivo. Exp. Hematol. 2013, 41, 1038–1049. [Google Scholar] [CrossRef] [Green Version]
- Dudek, S.E.; Luig, C.; Pauli, E.-K.; Schubert, U.; Ludwig, S. The Clinically Approved Proteasome Inhibitor PS-341 Efficiently Blocks Influenza A Virus and Vesicular Stomatitis Virus Propagation by Establishing an Antiviral State. J. Virol. 2010, 84, 9439–9451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tenbaum, S.P.; Ordóñez-Morán, P.; Puig, I.; Chicote, I.; Arqués, O.; Landolfi, S.; Fernández, Y.; Herance, J.R.; Gispert, J.D.; Mendizabal, L.; et al. β-catenin confers resistance to PI3K and AKT inhibitors and subverts FOXO3a to promote metastasis in colon cancer. Nat. Med. 2012, 18, 892–901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arqués, O.; Chicote, I.; Puig, I.; Tenbaum, S.P.; Argilés, G.; Dienstmann, R.; Fernández, N.; Caratù, G.; Matito, J.; Silberschmidt, D.; et al. Tankyrase Inhibition Blocks Wnt/b-Catenin Pathway and Reverts Resistance to PI3K and AKT Inhibitors in the Treatment of Colorectal Cancer. Clin. Cancer Res. 2016, 22, 644–656. [Google Scholar] [CrossRef] [Green Version]
- Deng, Z.; Atanasiu, C.; Zhao, K.; Marmorstein, R.; Sbodio, J.I.; Chi, N.-W.; Lieberman, P.M. Inhibition of Epstein-Barr virus OriP function by tankyrase, a telomere-associated poly-ADP ribose polymerase that binds and modifies EBNA1. J. Virol. 2005, 79, 4640–4650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Roy, S.; Liu, F.; Arav-Boger, R. Human cytomegalovirus inhibits the PARsylation activity of tankyrase—A potential strategy for suppression of the Wnt pathway. Viruses 2015, 8, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jha, B.K.; Polyakova, I.; Kessler, P.; Dong, B.; Dickerman, B.; Sen, G.C.; Silverman, R.H. Inhibition of RNase L and RNA-dependent protein kinase (PKR) by sunitinib impairs antiviral innate immunity. J. Biol. Chem. 2011, 286, 26319–26326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jha, B.K.; Dong, B.; Nguyen, C.T.; Polyakova, I.; Silverman, R.H. Suppression of antiviral innate immunity by sunitinib enhances oncolytic virotherapy. Mol. Ther. 2013, 21, 1749–1757. [Google Scholar] [CrossRef] [Green Version]
- Lawson, K.A.; Mostafa, A.A.; Shi, Z.Q.; Spurrell, J.; Chen, W.; Kawakami, J.; Gratton, K.; Thakur, S.; Morris, D.G. Repurposing sunitinib with oncolytic reovirus as a novel immunotherapeutic strategy for renal cell carcinoma. Clin. Cancer Res. 2016, 22, 5839–5850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, M.; Nitschke, M.; Sennino, B.; Murer, P.; Schriver, B.J.; Bell, A.; Subramanian, A.; McDonald, C.E.; Wang, J.; Cha, H.; et al. Amplification of oncolytic vaccinia virus widespread tumor cell killing by sunitinib through multiple mechanisms. Cancer Res. 2018, 78, 922–937. [Google Scholar] [CrossRef] [Green Version]
- Saha, D.; Wakimoto, H.; Peters, C.W.; Antoszczyk, S.J.; Rabkin, S.D.; Martuza, R.L. Combinatorial effects of vegfr kinase inhibitor axitinib and oncolytic virotherapy in mouse and human glioblastoma stem-like cell models. Clin. Cancer Res. 2018, 24, 3409–3422. [Google Scholar] [CrossRef] [Green Version]
- Heo, J.; Breitbach, C.J.; Moon, A.; Kim, C.W.; Patt, R.; Kim, M.K.; Lee, Y.K.; Oh, S.Y.; Woo, H.Y.; Parato, K.; et al. Sequential therapy with JX-594, a targeted oncolytic poxvirus, followed by sorafenib in hepatocellular carcinoma: Preclinical and clinical demonstration of combination efficacy. Mol. Ther. 2011, 19, 1170–1179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reeves, P.M.; Bommarius, B.; Lebeis, S.; McNulty, S.; Christensen, J.; Swimm, A.; Chahroudi, A.; Chavan, R.; Feinberg, M.B.; Veach, D.; et al. Disabling poxvirus pathogenesis by inhibition of Abl-family tyrosine kinases. Nat. Med. 2005, 11, 731–739. [Google Scholar] [CrossRef] [PubMed]
- Farsaci, B.; Higgins, J.P.; Hodge, J.W. Consequence of dose scheduling of sunitinib on host immune response elements and vaccine combination therapy. Int. J. Cancer 2012, 130, 1948–1959. [Google Scholar] [CrossRef] [Green Version]
- Ozao-Choy, J.; Ge, M.; Kao, J.; Wang, G.X.; Meseck, M.; Sung, M.; Schwartz, M.; Divino, C.M.; Pan, P.Y.; Chen, S.H. The novel role of tyrosine kinase inhibitor in the reversal of immune suppression and modulation of tumor microenvironment for immune-based cancer therapies. Cancer Res. 2009, 69, 2514–2522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kao, J.; Ko, E.C.; Eisenstein, S.; Sikora, A.G.; Fu, S.; Chen, S. Targeting immune suppressing myeloid-derived suppressor cells in oncology. Crit. Rev. Oncol. Hematol. 2011, 77, 12–19. [Google Scholar] [CrossRef] [Green Version]
- Ardiani, A.; Donahue, R.N.; Aftab, D.T.; Hodge, J.W.; Kwilas, A.R. Dual effects of a targeted small-molecule inhibitor (cabozantinib) on immune-mediated killing of tumor cells and immune tumor microenvironment permissiveness when combined with a cancer vaccine. J. Transl. Med. 2014, 12, 294. [Google Scholar] [CrossRef] [Green Version]
- Yamamura, K.; Kasuya, H.; Sahin, T.T.; Tan, G.; Hotta, Y.; Tsurumaru, N.; Fukuda, S.; Kanda, M.; Kobayashi, D.; Tanaka, C.; et al. Combination treatment of human pancreatic cancer xenograft models with the epidermal growth factor receptor tyrosine kinase inhibitor erlotinib and oncolytic herpes simplex virus hf10. Ann. Surg. Oncol. 2014, 21, 691–698. [Google Scholar] [CrossRef] [PubMed]
- Mahller, Y.Y.; Vaikunth, S.S.; Currier, M.A.; Miller, S.J.; Ripberger, M.C.; Hsu, Y.H.; Mehrian-Shai, R.; Collins, M.H.; Crombleholme, T.M.; Ratner, N.; et al. Oncolytic HSV and erlotinib inhibit tumor growth and angiogenesis in a novel malignant peripheral nerve sheath tumor xenograft model. Mol. Ther. 2007, 15, 279–286. [Google Scholar] [CrossRef]
- Stark, G.R.; Kerr, I.M.; Leung, S.; Muzaffar, R.; Vlieststra, R.J.; Levy, D.E.; Trapman, J.; Bluyssen, H.A.; van der Made, A.C. Combinatorial association and abundance of components of interferon-stimulated gene factor 3 dictate the selectivity of interferon responses. Proc. Natl. Acad. Sci. USA 2006, 92, 5645–5649. [Google Scholar] [CrossRef] [Green Version]
- Nusinzon, I.; Horvath, C.M. Positive and Negative Regulation of the Innate Antiviral Response and Beta Interferon Gene Expression by Deacetylation. Mol. Cell. Biol. 2006, 26, 3106–3113. [Google Scholar] [CrossRef] [Green Version]
- Genin, P.; Morin, P.; Civas, A. Impairment of Interferon-Induced IRF-7 Gene Expression due to Inhibition of ISGF3 Formation by Trichostatin A. J. Virol. 2003, 77, 7113–7119. [Google Scholar] [CrossRef] [Green Version]
- Liu, T.C.; Castelo-Branco, P.; Rabkin, S.D.; Martuza, R.L. Trichostatin A and oncolytic HSV combination therapy shows enhanced antitumoral and antiangiogenic effects. Mol. Ther. 2008, 16, 1041–1047. [Google Scholar] [CrossRef]
- Dornan, M.H.; Krishnan, R.; MacKlin, A.M.; Selman, M.; El Sayes, N.; Son, H.H.; Davis, C.; Chen, A.; Keillor, K.; Le, P.J.; et al. First-in-class small molecule potentiators of cancer virotherapy. Sci. Rep. 2016, 6, 26786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gilbert, J.; Gore, S.D.; Herman, J.G.; Carducci, M.A. The clinical application of targeting cancer through histone acetylation and hypomethylation. Clin. Cancer Res. 2004, 10, 4589–4596. [Google Scholar] [CrossRef] [Green Version]
- Archer, S.Y.; Hodin, R.A. Histone acetylation and cancer. Curr. Opin. Genet. Dev. 1999, 9, 171–174. [Google Scholar] [CrossRef]
- Jacobson, S.; Pillus, L. Modifying chromatin and concepts of cancer. Curr. Opin. Genet. Dev. 1999, 9, 175–184. [Google Scholar] [CrossRef]
- Place, R.F.; Noonan, E.J.; Giardina, C. HDAC inhibition prevents NF-κB activation by suppressing proteasome activity: Down-regulation of proteasome subunit expression stabilizes IκBα. Biochem. Pharmacol. 2005, 70, 394–406. [Google Scholar] [CrossRef] [PubMed]
- Bridle, B.W.; Stephenson, K.B.; Boudreau, J.E.; Koshy, S.; Kazdhan, N.; Pullenayegum, E.; Brunellière, J.; Bramson, J.L.; Lichty, B.D.; Wan, Y. Potentiating cancer immunotherapy using an oncolytic virus. Mol. Ther. 2010, 18, 1430–1439. [Google Scholar] [CrossRef]
- Bridle, B.W.; Chen, L.; Lemay, C.G.; Diallo, J.-S.; Pol, J.; Nguyen, A.; Capretta, A.; He, R.; Bramson, J.L.; Bell, J.C.; et al. HDAC inhibition suppresses primary immune responses, enhances secondary immune responses, and abrogates autoimmunity during tumor immunotherapy. Mol. Ther. 2013, 21, 887–894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shen, L.; Shrikant, P.; Ciesielski, M.; Ellis, L.; Fenstermaker, R.; Miles, K.M.; Ramakrishnan, S.; Pili, R.; Sotomayor, P. Class I Histone Deacetylase Inhibitor Entinostat Suppresses Regulatory T Cells and Enhances Immunotherapies in Renal and Prostate Cancer Models. PLoS ONE 2012, 7, e30815. [Google Scholar] [CrossRef] [PubMed]
- Otsuki, A.; Patel, A.; Kasai, K.; Suzuki, M.; Kurozumi, K.; Chiocca, E.A.; Saeki, Y. Histone deacetylase inhibitors augment antitumor efficacy of herpes-based oncolytic viruses. Mol. Ther. 2008, 16, 1546–1555. [Google Scholar] [CrossRef]
- Alvarez-Breckenridge, C.A.; Yu, J.; Price, R.; Wei, M.; Wang, Y.; Nowicki, M.O.; Ha, Y.P.; Bergin, S.; Hwang, C.; Fernandez, S.A.; et al. The histone deacetylase inhibitor valproic acid lessens NK cell action against oncolytic virus-infected glioblastoma cells by inhibition of STAT5/T-BET signaling and generation of gamma interferon. J. Virol. 2012, 86, 4566–4577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jennings, V.A.; Scott, G.B.; Rose, A.M.S.; Scott, K.J.; Migneco, G.; Keller, B.; Reilly, K.; Donnelly, O.; Peach, H.; Dewar, D.; et al. Potentiating Oncolytic Virus-Induced Immune-Mediated Tumor Cell Killing Using Histone Deacetylase Inhibition. Mol. Ther. 2019, 27, 1139–1152. [Google Scholar] [CrossRef] [Green Version]
- Vanhaecke, T.; Papeleu, P.; Elaut, G.; Rogiers, V. Trichostatin A-like Hydroxamate Histone Deacetylase Inhibitors as Therapeutic Agents: Toxicological Point of View. Curr. Med. Chem. 2004, 11, 1629–1643. [Google Scholar] [CrossRef]
- Bracker, T.U.; Sommer, A.; Fichtner, I.; Faus, H.; Haendler, B.; Hess-Stumpp, H. Efficacy of MS-275, a selective inhibitor of class I histone deacetylases, in human colon cancer models. Int. J. Oncol. 2009, 35, 909–920. [Google Scholar] [CrossRef] [PubMed]
- McKinsey, T.A. Isoform-selective HDAC inhibitors: Closing in on translational medicine for the heart. J. Mol. Cell. Cardiol. 2011, 51, 491–496. [Google Scholar] [CrossRef] [PubMed]
- Richon, V.M. Cancer biology: Mechanism of antitumour action of vorinostat (suberoylanilide hydroxamic acid), a novel histone deacetylase inhibitor. Br. J. Cancer 2006, 95, S2–S6. [Google Scholar] [CrossRef]
- Islam, S.; Espitia, C.M.; Persky, D.O.; Carew, J.S.; Nawrocki, S.T. Resistance to histone deacetylase inhibitors confers hypersensitivity to oncolytic reovirus therapy. Blood Adv. 2020, 4, 5297–5310. [Google Scholar] [CrossRef]
- Stiff, A.; Caserta, E.; Sborov, D.W.; Nuovo, G.J.; Mo, X.; Schlotter, S.Y.; Canella, A.; Smith, E.; Badway, J.; Old, M.; et al. Histone Deacetylase Inhibitors Enhance the Therapeutic Potential of Reovirus in Multiple Myeloma. Mol. Cancer Ther. 2016, 15, 830–841. [Google Scholar] [CrossRef] [Green Version]
- Bartlett, D.L.; Liu, Z.; Sathaiah, M.; Ravindranathan, R.; Guo, Z.; He, Y.; Guo, Z.S. Oncolytic viruses as therapeutic cancer vaccines. Mol. Cancer 2013, 12, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Katsura, T.; Iwai, S.; Ota, Y.; Shimizu, H.; Ikuta, K.; Yura, Y. The effects of trichostatin A on the oncolytic ability of herpes simplex virus for oral squamous cell carcinoma cells. Cancer Gene Ther. 2009, 16, 237–245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- MacTavish, H.; Diallo, J.; Huang, B.; Stanford, M.; Le Boeuf, F.; De Silva, N.; Cox, J.; Simmons, J.G.; Guimond, T.; Falls, T.; et al. Enhancement of vaccinia virus based oncolysis with histone deacetylase inhibitors. PLoS ONE 2010, 5, e14462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, J.; Bonifati, S.; Hristov, G.; Marttila, T.; Valmary-Degano, S.; Stanzel, S.; Schnölzer, M.; Mougin, C.; Aprahamian, M.; Grekova, S.P.; et al. Synergistic combination of valproic acid and oncolytic parvovirus H-1PV as a potential therapy against cervical and pancreatic carcinomas. EMBO Mol. Med. 2013, 5, 1537–1555. [Google Scholar] [CrossRef]
- Leenstra, S.; Kloezeman, J.J.; de Vrij, J.; Dirven, C.M.F.; Kleijn, A.; van den Bossche, W.; Berghauser Pont, L.M.E.; Lamfers, M.L.M.; Kaufmann, J.K. The HDAC Inhibitors Scriptaid and LBH589 Combined with the Oncolytic Virus Delta24-RGD Exert Enhanced Anti-Tumor Efficacy in Patient-Derived Glioblastoma Cells. PLoS ONE 2015, 10, e0127058. [Google Scholar] [CrossRef] [Green Version]
- Manning, J.; Indrova, M.; Lubyova, B.; Pribylova, H.; Bieblova, J.; Hejnar, J.; Simova, J.; Jandlova, T.; Bubenik, J.; Reinis, M. Induction of MHC class I molecule cell surface expression and epigenetic activation of antigen-processing machinery components in a murine model for human papilloma virus 16-associated tumours. Immunology 2008, 123, 218–227. [Google Scholar] [CrossRef]
- Leone, P.; Shin, E.C.; Perosa, F.; Vacca, A.; Dammacco, F.; Racanelli, V. MHC class I antigen processing and presenting machinery: Organization, function, and defects in tumor cells. J. Natl. Cancer Inst. 2013, 105, 1172–1187. [Google Scholar] [CrossRef] [Green Version]
- Marincola, F.M.; Nicolay, H.J.M.; Altomonte, M.; Guidoboni, M.; Colizzi, F.; Fonsatti, E.; Calabro, L.; Pezzani, L.; Maio, M.; Sigalotti, L. Functional Up-regulation of Human Leukocyte Antigen Class I Antigens Expression by 5-aza-2′-deoxycytidine in Cutaneous Melanoma: Immunotherapeutic Implications. Clin. Cancer Res. 2007, 13, 3333–3338. [Google Scholar] [CrossRef] [Green Version]
- Fernald, K.; Kurokawa, M. Evading apoptosis in cancer. Trends Cell Biol. 2013, 23, 620–633. [Google Scholar] [CrossRef] [Green Version]
- Gong, Y.; Fan, Z.; Luo, G.; Yang, C.; Huang, Q.; Fan, K.; Cheng, H.; Jin, K.; Ni, Q.; Yu, X.; et al. The role of necroptosis in cancer biology and therapy. Mol. Cancer 2019, 18, 100. [Google Scholar] [CrossRef] [Green Version]
- You, Y.; Cheng, A.-C.; Wang, M.-S.; Jia, R.-Y.; Sun, K.-F.; Yang, Q.; Wu, Y.; Zhu, D.; Chen, S.; Liu, M.-F.; et al. The suppression of apoptosis by α-herpesvirus. Cell Death Dis. 2017, 8, e2749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Veyer, D.L.; Carrara, G.; Maluquer de Motes, C.; Smith, G.L. Vaccinia virus evasion of regulated cell death. Immunol. Lett. 2017, 186, 68–80. [Google Scholar] [CrossRef] [Green Version]
- Sprague, L.; Braidwood, L.; Conner, J.; Cassady, K.A.; Benencia, F.; Cripe, T.P. Please stand by: How oncolytic viruses impact bystander cells. Future Virol. 2018, 13, 671–680. [Google Scholar] [CrossRef]
- Beug, S.T.; Tang, V.A.; LaCasse, E.C.; Cheung, H.H.; Beauregard, C.E.; Brun, J.; Nuyens, J.P.; Earl, N.; St-Jean, M.; Holbrook, J.; et al. Smac mimetics and innate immune stimuli synergize to promote tumor death. Nat. Biotechnol. 2014, 32, 182–190. [Google Scholar] [CrossRef] [Green Version]
- Yoshida, H. ER stress and diseases. FEBS J. 2007, 274, 630–658. [Google Scholar] [CrossRef]
- Lee, A.S.; Hendershot, L.M. ER stress and cancer. Cancer Biol. Ther. 2006, 5, 721–722. [Google Scholar] [CrossRef]
- Gugliotta, G.; Sudo, M.; Cao, Q.; Lin, D.C.; Sun, H.; Takao, S.; Le Moigne, R.; Rolfe, M.; Gery, S.; Müschen, M.; et al. Valosin-Containing Protein/p97 as a Novel Therapeutic Target in Acute Lymphoblastic Leukemia. Neoplasia 2017, 19, 750–761. [Google Scholar] [CrossRef]
- Prasad, V.; Suomalainen, M.; Pennauer, M.; Yakimovich, A.; Andriasyan, V.; Hemmi, S.; Greber, U.F. Chemical Induction of Unfolded Protein Response Enhances Cancer Cell Killing through Lytic Virus Infection. J. Virol. 2014, 88, 13086–13098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, H.; Li, K.; Lin, Y.; Xing, F.; Xiao, X.; Cai, J.; Zhu, W.; Liang, J.; Tan, Y.; Fu, L.; et al. Targeting VCP enhances anticancer activity of oncolytic virus M1 in hepatocellular carcinoma. Sci. Transl. Med. 2017, 9, eaam7996. [Google Scholar] [CrossRef] [Green Version]
- Prasad, V.; Greber, U.F. The endoplasmic reticulum unfolded protein response—Homeostasis, cell death and evolution in virus infections. FEMS Microbiol. Rev. 2021, 16, 1–19. [Google Scholar] [CrossRef]
- Taverner, W.K.; Jacobus, E.J.; Christianson, J.; Champion, B.; Paton, A.W.; Paton, J.C.; Su, W.; Cawood, R.; Seymour, L.W.; Lei-Rossmann, J. Calcium Influx Caused by ER Stress Inducers Enhances Oncolytic Adenovirus Enadenotucirev Replication and Killing through PKCα Activation. Mol. Ther. Oncolytics 2019, 15, 117–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Galmarini, C.M.; Mackey, J.R.; Dumontet, C. Nucleoside analogues and nucleobases in cancer treatment. Lancet Oncol. 2002, 3, 415–424. [Google Scholar] [CrossRef]
- Nakano, K.; Todo, T.; Zhao, G.; Yamaguchi, K.; Kuroki, S.; Cohen, J.B.; Glorioso, J.C.; Tanaka, M. Enhanced efficacy of conditionally replicating herpes simplex virus (G207) combined with 5-fluorouracil and surgical resection in peritoneal cancer dissemination models. J. Gene Med. 2005, 7, 638–648. [Google Scholar] [CrossRef]
- Leitner, S.; Sweeney, K.; Öberg, D.; Davies, D.; Miranda, E.; Lemoine, N.R.; Halldén, G. Oncolytic adenoviral mutants with E1B19KGene deletions enhance gemcitabine-induced apoptosis in pancreatic carcinoma cells and anti-tumor efficacy in vivo. Clin. Cancer Res. 2009, 15, 1730–1740. [Google Scholar] [CrossRef] [Green Version]
- Gutermann, A.; Mayer, E.; von Dehn-Rothfelser, K.; Breidenstein, C.; Weber, M.; Muench, M.; Gungor, D.; Suehnel, J.; Moebius, U.; Lechmann, M. Efficacy of oncolytic herpesvirus NV1020 can be enhanced by combination with chemotherapeutics in colon carcinoma cells. Hum. Gene Ther. 2006, 17, 1241–1253. [Google Scholar] [CrossRef]
- Angelova, A.L.; Aprahamian, M.; Grekova, S.P.; Hajri, A.; Leuchs, B.; Giese, N.A.; Dinsart, C.; Herrmann, A.; Balboni, G.; Rommelaere, J.; et al. Improvement of gemcitabine-based therapy of pancreatic carcinoma by means of oncolytic parvovirus H-1PV. Clin. Cancer Res. 2009, 15, 511–519. [Google Scholar] [CrossRef] [Green Version]
- Weiland, T.; Lampe, J.; Essmann, F.; Venturelli, S.; Berger, A.; Bossow, S.; Berchtold, S.; Schulze-Osthoff, K.; Lauer, U.M.; Bitzer, M. Enhanced killing of therapy-induced senescent tumor cells by oncolytic measles vaccine viruses. Int. J. Cancer 2014, 134, 235–243. [Google Scholar] [CrossRef]
- Chen, W.; Fan, W.; Ru, G.; Huang, F.; Lu, X.; Zhang, X.; Mou, X.; Wang, S. Gemcitabine combined with an engineered oncolytic vaccinia virus exhibits a synergistic suppressive effect on the tumor growth of pancreatic cancer. Oncol. Rep. 2019, 41, 67–76. [Google Scholar] [CrossRef]
- Liu, X.; Yang, Z.; Li, Y.; Zhu, Y.; Li, W.; Li, S.; Wang, J.; Cui, Y.; Shang, C.; Liu, Z.; et al. Chemovirotherapy of Lung Squamous Cell Carcinoma by Combining Oncolytic Adenovirus with Gemcitabine. Front. Oncol. 2020, 10, 229. [Google Scholar] [CrossRef]
- May, V.; Berchtold, S.; Berger, A.; Venturelli, S.; Burkard, M.; Leischner, C.; Malek, N.P.; Lauer, U.M. Chemovirotherapy for pancreatic cancer: Gemcitabine plus oncolytic measles vaccine virus. Oncol. Lett. 2019, 18, 5534–5542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mannhold, R.; Fulda, S.; Carosati, E. IAP antagonists: Promising candidates for cancer therapy. Drug Discov. Today 2010, 15, 210–219. [Google Scholar] [CrossRef]
- Michie, J.; Kearney, C.J.; Hawkins, E.D.; Silke, J.; Oliaro, J. The Immuno-Modulatory Effects of Inhibitor of Apoptosis Protein Antagonists in Cancer Immunotherapy. Cells 2020, 9, 207. [Google Scholar] [CrossRef] [Green Version]
- Tumilasci, V.F.; Oliere, S.; Nguyen, T.L.-A.; Shamy, A.; Bell, J.; Hiscott, J. Targeting the Apoptotic Pathway with BCL-2 Inhibitors Sensitizes Primary Chronic Lymphocytic Leukemia Cells to Vesicular Stomatitis Virus-Induced Oncolysis. J. Virol. 2008, 82, 8487–8499. [Google Scholar] [CrossRef] [Green Version]
- Sarkar, S.; Quinn, B.A.; Shen, X.-N.; Dash, R.; Das, S.K.; Emdad, L.; Klibanov, A.L.; Wang, X.-Y.; Pellecchia, M.; Sarkar, D.; et al. Therapy of prostate cancer using a novel cancer terminator virus and a small molecule BH-3 mimetic. Oncotarget 2015, 6, 10712–10727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karin, M. Nuclear factor-κB in cancer development and progression. Nature 2006, 441, 431–436. [Google Scholar] [CrossRef]
- Petersen, S.L.; Wang, L.; Yalcin-Chin, A.; Li, L.; Peyton, M.; Minna, J.; Harran, P.; Wang, X. Autocrine TNFα Signaling Renders Human Cancer Cells Susceptible to Smac-Mimetic-Induced Apoptosis. Cancer Cell 2007, 12, 445–456. [Google Scholar] [CrossRef] [Green Version]
- Du, C.; Fang, M.; Li, Y.; Li, L.; Wang, X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 2000, 102, 33–42. [Google Scholar] [CrossRef] [Green Version]
- Li, L.; Thomas, R.M.; Suzuki, H.; De Brabander, J.K.; Wang, X.; Harran, P.G. A small molecule Smac mimic potentiates TRAIL- and TNFα-mediated cell death. Science 2004, 305, 1471–1474. [Google Scholar] [CrossRef] [Green Version]
- Petersen, S.L.; Peyton, M.; Minna, J.D.; Wang, X. Overcoming cancer cell resistance to Smac mimetic induced apoptosis by modulating cIAP-2 expression. Proc. Natl. Acad. Sci. USA 2010, 107, 11936–11941. [Google Scholar] [CrossRef] [Green Version]
- Bai, L.; McEachern, D.; Yang, C.Y.; Lu, J.; Sun, H.; Wang, S. LRIG1 modulates cancer cell sensitivity to Smac mimetics by regulating TNFα expression and receptor tyrosine kinase signaling. Cancer Res. 2012, 72, 1229–1238. [Google Scholar] [CrossRef] [Green Version]
- Fulda, S. Smac mimetics as IAP antagonists. Semin. Cell Dev. Biol. 2015, 39, 132–138. [Google Scholar] [CrossRef]
- Fulda, S.; Wick, W.; Weller, M.; Debatin, K.M. Smac agonists sensitize for Apo2L/TRAIL-or anticancer drug-induced apoptosis and induce regression of malignant glioma in vivo. Nat. Med. 2002, 8, 808–815. [Google Scholar] [CrossRef]
- Cai, J.; Lin, Y.; Zhang, H.; Liang, J.; Tan, Y.; Cavenee, W.K.; Yan, G. Selective replication of oncolytic virus M1 results in a bystander killing effect that is potentiated by Smac mimetics. Proc. Natl. Acad. Sci. USA 2017, 114, 201701002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheung, H.H.; Beug, S.T.; St Jean, M.; Brewster, A.; Kelly, N.L.; Wang, S.; Korneluk, R.G. Smac mimetic compounds potentiate interleukin-1β-mediated cell death. J. Biol. Chem. 2010, 285, 40612–40623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lalaoui, N.; Hänggi, K.; Brumatti, G.; Chau, D.; Nguyen, N.Y.N.; Vasilikos, L.; Spilgies, L.M.; Heckmann, D.A.; Ma, C.; Ghisi, M.; et al. Targeting p38 or MK2 Enhances the Anti-Leukemic Activity of Smac-Mimetics. Cancer Cell 2016, 29, 145–158. [Google Scholar] [CrossRef] [Green Version]
- Beug, S.T.; Pichette, S.J.; St-Jean, M.; Holbrook, J.; Walker, D.E.; LaCasse, E.C.; Korneluk, R.G. Combination of IAP Antagonists and TNF-α-Armed Oncolytic Viruses Induce Tumor Vascular Shutdown and Tumor Regression. Mol. Ther. Oncolytics 2018, 10, 28–39. [Google Scholar] [CrossRef]
- Beug, S.T.; Conrad, D.P.; Alain, T.; Korneluk, R.G.; Lacasse, E.C. Combinatorial cancer immunotherapy strategies with proapoptotic small-molecule IAP antagonists. Int. J. Dev. Biol. 2015, 59, 141–147. [Google Scholar] [CrossRef]
- Beug, S.T.; Beauregard, C.E.; Healy, C.; Sanda, T.; St-Jean, M.; Chabot, J.; Walker, D.E.; Mohan, A.; Earl, N.; Lun, X.; et al. Smac mimetics synergize with immune checkpoint inhibitors to promote tumour immunity against glioblastoma. Nat. Commun. 2017, 8, 14278. [Google Scholar] [CrossRef] [Green Version]
- Dobson, C.C.; Naing, T.; Beug, S.T.; Faye, M.D.; Chabot, J.; St-Jean, M.; Walker, D.E.; LaCasse, E.C.; Stojdl, D.F.; Korneluk, R.G.; et al. Oncolytic virus synergizes with Smac mimetic compounds to induce rhabdomyosarcoma cell death in a syngeneic murine model. Oncotarget 2016. [Google Scholar] [CrossRef]
- Kim, D.-S.; Dastidar, H.; Zhang, C.; Zemp, F.J.; Lau, K.; Ernst, M.; Rakic, A.; Sikdar, S.; Rajwani, J.; Naumenko, V.; et al. Smac mimetics and oncolytic viruses synergize in driving anticancer T-cell responses through complementary mechanisms. Nat. Commun. 2017, 8, 344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cai, J.; Yan, G. The identification and development of a novel oncolytic virus: Alphavirus M1. Hum. Gene Ther. 2021, 32, 138–149. [Google Scholar] [CrossRef]
- Delbridge, A.R.D.; Strasser, A. The BCL-2 protein family, BH3-mimetics and cancer therapy. Cell Death Differ. 2015, 22, 1071–1080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Takai, S.; Hodge, J.W.; Farsaci, B.; Schlom, J.; Sabzevari, H.; Higgins, J.P.; Di Bari, M.G. Effect of a small molecule BCL-2 inhibitor on immune function and use with a recombinant vaccine. Int. J. Cancer 2010, 127, 1603–1613. [Google Scholar] [CrossRef] [Green Version]
- Samuel, S.; Tumilasci, V.F.; Oliere, S.; Nguyên, T.L.; Shamy, A.; Bell, J.; Hiscott, J. VSV oncolysis in combination with the BCL-2 inhibitor obatoclax overcomes apoptosis resistance in chronic lymphocytic leukemia. Mol. Ther. 2010, 18, 2094–2103. [Google Scholar] [CrossRef]
- Zhou, J.; Giannakakou, P. Targeting microtubules for cancer chemotherapy. Curr. Med. Chem. Anticancer Agents 2005, 5, 65–71. [Google Scholar] [CrossRef] [Green Version]
- Zeng, W.G.; Li, J.J.; Hu, P.; Lei, L.; Wang, J.N.; Liu, R. Bin An oncolytic herpes simplex virus vector, G47Δ, synergizes with paclitaxel in the treatment of breast cancer. Oncol. Rep. 2013, 29, 2355–2361. [Google Scholar] [CrossRef] [Green Version]
- Lin, S.F.; Gao, S.P.; Price, D.L.; Li, S.; Chou, T.C.; Singh, P.; Huang, Y.Y.; Fong, Y.; Wong, R.J. Synergy of a herpes oncolytic virus and paclitaxel for anaplastic thyroid cancer. Clin. Cancer Res. 2008, 14, 1519–1528. [Google Scholar] [CrossRef] [Green Version]
- Passer, B.J.; Castelo-Branco, P.; Buhrman, J.S.; Varghese, S.; Rabkin, S.D.; Martuza, R.L. Oncolytic herpes simplex virus vectors and taxanes synergize to promote killing of prostate cancer cells. Cancer Gene Ther. 2009, 16, 551–560. [Google Scholar] [CrossRef]
- Heinemann, L.; Simpson, G.R.; Boxall, A.; Kottke, T.; Relph, K.L.; Vile, R.; Melcher, A.; Prestwich, R.; Harrington, K.J.; Morgan, R.; et al. Synergistic effects of oncolytic reovirus and docetaxel chemotherapy in prostate cancer. BMC Cancer 2011, 11, 221. [Google Scholar] [CrossRef] [Green Version]
- Karapanagiotou, E.M.; Roulstone, V.; Twigger, K.; Ball, M.; Tanay, M.A.; Nutting, C.; Newbold, K.; Gore, M.E.; Larkin, J.; Syrigos, K.N.; et al. Phase I/II trial of carboplatin and paclitaxel chemotherapy in combination with intravenous oncolytic reovirus in patients with advanced malignancies. Clin. Cancer Res. 2012, 18, 2080–2089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fujiwara, T.; Kagawa, S.; Kishimoto, H.; Endo, Y.; Hioki, M.; Ikeda, Y.; Sakai, R.; Urata, Y.; Tanaka, N.; Fujiwara, T. Enhanced antitumor efficacy of telomerase-selective oncolytic adenoviral agent OBP-401 with docetaxel: Preclinical evaluation of chemovirotherapy. Int. J. Cancer 2006, 119, 432–440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bourgeois-Daigneault, M.-C.; St-Germain, L.E.; Roy, D.G.; Pelin, A.; Aitken, A.S.; Arulanandam, R.; Falls, T.; Garcia, V.; Diallo, J.-S.; Bell, J.C. Combination of Paclitaxel and MG1 oncolytic virus as a successful strategy for breast cancer treatment. Breast Cancer Res. 2016, 18, 83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sei, S.; Mussio, J.K.; Yang, Q.E.; Nagashima, K.; Parchment, R.E.; Coffey, M.C.; Shoemaker, R.H.; Tomaszewski, J.E. Synergistic antitumor activity of oncolytic reovirus and chemotherapeutic agents in non-small cell lung cancer cells. Mol. Cancer 2009, 8, 47. [Google Scholar] [CrossRef] [Green Version]
- Lal, G.; Rajala, M.S. Combination of oncolytic measles virus armed with BNiP3, a pro-apoptotic gene and paclitaxel induces breast cancer cell death. Front. Oncol. 2019, 9, 676. [Google Scholar] [CrossRef] [Green Version]
- Garofalo, M.; Saari, H.; Somersalo, P.; Crescenti, D.; Kuryk, L.; Aksela, L.; Capasso, C.; Madetoja, M.; Koskinen, K.; Oksanen, T.; et al. Antitumor effect of oncolytic virus and paclitaxel encapsulated in extracellular vesicles for lung cancer treatment. J. Control. Release 2018, 283, 223–234. [Google Scholar] [CrossRef]
- Arulanandam, R.; Batenchuk, C.; Varette, O.; Zakaria, C.; Garcia, V.; Forbes, N.E.; Davis, C.; Krishnan, R.; Karmacharya, R.; Cox, J.; et al. Microtubule disruption synergizes with oncolytic virotherapy by inhibiting interferon translation and potentiating bystander killing. Nat. Commun. 2015, 6, 6410. [Google Scholar] [CrossRef] [Green Version]
- Ziauddin, M.F.; Guo, Z.S.; O’Malley, M.E.; Austin, F.; Popovic, P.J.; Kavanagh, M.A.; Li, J.; Sathaiah, M.; Thirunavukarasu, P.; Fang, B.; et al. TRAIL gene-armed oncolytic poxvirus and oxaliplatin can work synergistically against colorectal cancer. Gene Ther. 2010, 17, 550–559. [Google Scholar] [CrossRef] [Green Version]
- Pan, Q.; Huang, Y.; Chen, L.; Gu, J.F.; Zhou, X. SMAC-armed vaccinia virus induces both apoptosis and necroptosis and synergizes the efficiency of vinblastine in HCC. Hum. Cell 2014, 27, 162–171. [Google Scholar] [CrossRef] [PubMed]
- Cinatl, J.; Cinatl, J.; Michaelis, M.; Kabickova, H.; Kotchetkov, R.; Vogel, J.U.; Doerr, H.W.; Klingebiel, T.; Hernáiz Driever, P. Potent oncolytic activity of multimutated herpes simplex virus G207 in combination with vincristine against human rhabdomyosarcoma. Cancer Res. 2003, 63, 1508–1514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moehler, M.; Sieben, M.; Roth, S.; Springsguth, F.; Leuchs, B.; Zeidler, M.; Dinsart, C.; Rommelaere, J.; Galle, P.R. Activation of the human immune system by chemotherapeutic or targeted agents combined with the oncolytic parvovirus H-1. BMC Cancer 2011, 11, 464. [Google Scholar] [CrossRef] [Green Version]
- Choi, C.-H.; Cha, Y.-J.; An, C.-S.; Kim, K.-J.; Kim, K.-C.; Moon, S.-P.; Lee, Z.; Min, Y.-D. Molecular mechanisms of heptaplatin effective against cisplatin-resistant cancer cell lines: Less involvement of metallothionein. Cancer Cell Int. 2004, 4, 6. [Google Scholar] [CrossRef] [Green Version]
- Khuri, F.R.; Nemunaitis, J.; Ganly, I.; Arseneau, J.; Tannock, I.F.; Romel, L.; Gore, M.; Ironside, J.; MacDougall, R.H.; Heise, C.; et al. A controlled trial of intratumoral ONYX-015, a selectively-replicating adenovirus, in combination with cisplatin and 5-fluorouracil in patients with recurrent head and neck cancer. Nat. Med. 2000, 6, 879–885. [Google Scholar] [CrossRef]
- Mell, L.K.; Brumund, K.T.; Daniels, G.A.; Advani, S.J.; Zakeri, K.; Wright, M.E.; Onyeama, S.J.; Weisman, R.A.; Sanghvi, P.R.; Martin, P.J.; et al. Phase I trial of intravenous oncolytic vaccinia virus (GL-ONC1) with cisplatin and radiotherapy in patients with locoregionally advanced head and neck carcinoma. Clin. Cancer Res. 2017, 23, 5696–5702. [Google Scholar] [CrossRef] [Green Version]
- Arulanandam, R.; Taha, Z.; Garcia, V.; Selman, M.; Chen, A.; Varette, O.; Jirovec, A.; Sutherland, K.; Macdonald, E.; Tzelepis, F.; et al. The strategic combination of trastuzumab emtansine with oncolytic rhabdoviruses leads to therapeutic synergy. Commun. Biol. 2020. [Google Scholar] [CrossRef]
- Hong, B.; Chapa, V.; Saini, U.; Modgil, P.; Cohn, D.E.; He, G.; Siddik, Z.H.; Sood, A.K.; Yan, Y.; Selvendiran, K.; et al. Oncolytic HSV therapy modulates vesicular trafficking inducing cisplatin sensitivity and antitumor immunity. Clin. Cancer Res. 2021, 27, 542–553. [Google Scholar] [CrossRef]
- You, L.; Wang, Y.; Jin, Y.; Qian, W. Downregulation of Mcl-1 synergizes the apoptotic response to combined treatment with cisplatin and a novel fiber chimeric oncolytic adenovirus. Oncol. Rep. 2012, 27, 971–978. [Google Scholar] [CrossRef]
- Champoux, J.J. DNA Topoisomerases: Structure, Function, and Mechanism. Annu. Rev. Biochem. 2002, 70, 369–413. [Google Scholar] [CrossRef] [Green Version]
- Hande, K.R. Etoposide: Four decades of development of a topoisomerase II inhibitor. Eur. J. Cancer 1998, 34, 1514–1521. [Google Scholar] [CrossRef]
- Ewesuedo, R.B.; Ratain, M.J. Topoisomerase I Inhibitors. Oncologist 1997, 2, 359–364. [Google Scholar] [CrossRef] [Green Version]
- Ezoe, S. Secondary leukemia associated with the anti-cancer agent, etoposide, a topoisomerase II inhibitor. Int. J. Environ. Res. Public Health 2012, 9, 2444–2453. [Google Scholar] [CrossRef] [Green Version]
- Felix, C.A. Leukemias related to treatment with DNA topoisomerase II inhibitors. Med. Pediatr. Oncol. Off. J. SIOP Int. Soc. Pediatr. Oncol. 2001, 36, 525–535. [Google Scholar] [CrossRef] [PubMed]
- Skelding, K.A.; Barry, R.D.; Shafren, D.R. Enhanced oncolysis mediated by Coxsackievirus A21 in combination with doxorubicin hydrochloride. Investig. New Drugs 2012, 30, 568–581. [Google Scholar] [CrossRef] [PubMed]
- Schache, P.; Gürlevik, E.; Strüver, N.; Woller, N.; Malek, N.; Zender, L.; Manns, M.; Wirth, T.; Kühnel, F.; Kubicka, S. VSV virotherapy improves chemotherapy by triggering apoptosis due to proteasomal degradation of Mcl-1. Gene Ther. 2009, 16, 849–861. [Google Scholar] [CrossRef] [Green Version]
- Siurala, M.; Bramante, S.; Vassilev, L.; Hirvinen, M.; Parviainen, S.; Tähtinen, S.; Guse, K.; Cerullo, V.; Kanerva, A.; Kipar, A.; et al. Oncolytic adenovirus and doxorubicin-based chemotherapy results in synergistic antitumor activity against soft-tissue sarcoma. Int. J. Cancer 2015, 136, 945–954. [Google Scholar] [CrossRef] [Green Version]
- Rodríguez Stewart, R.M.; Berry, J.T.L.; Berger, A.K.; Yoon, S.B.; Hirsch, A.L.; Guberman, J.A.; Patel, N.B.; Tharp, G.K.; Bosinger, S.E.; Mainou, B.A. Enhanced Killing of Triple-Negative Breast Cancer Cells by Reassortant Reovirus and Topoisomerase Inhibitors. J. Virol. 2019, 93. [Google Scholar] [CrossRef] [PubMed]
- Berry, J.T.L.; Muñoz, L.E.; Rodríguez Stewart, R.M.; Selvaraj, P.; Mainou, B.A. Doxorubicin Conjugation to Reovirus Improves Oncolytic Efficacy in Triple-Negative Breast Cancer. Mol. Ther. Oncolytics 2020, 18, 556–572. [Google Scholar] [CrossRef]
- Denard, B.; Seemann, J.; Chen, Q.; Gay, A.; Huang, H.; Chen, Y.; Ye, J. The membrane-bound transcription factor CREB3L1 is activated in response to virus infection to inhibit proliferation of virus-infected cells. Cell Host Microbe 2011, 10, 65–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mistarz, A.; Graczyk, M.; Winkler, M.; Singh, P.K.; Cortes, E.; Miliotto, A.; Liu, S.; Long, M.; Yan, L.; Stablewski, A.; et al. Induction of Cell Death in Ovarian Cancer Cells by Doxorubicin and Oncolytic Vaccinia Virus is Associated with CREB3L1 Activation. Mol. Ther. Oncolytics 2021. [Google Scholar] [CrossRef]
- Aitken, A.S.; Roy, D.G.; Bourgeois-Daigneault, M.-C. Taking a Stab at Cancer; Oncolytic Virus-Mediated Anti-Cancer Vaccination Strategies. Biomedicines 2017, 5, 3. [Google Scholar] [CrossRef] [Green Version]
- Watanabe, N.; McKenna, M.K.; Rosewell Shaw, A.; Suzuki, M. Clinical CAR-T Cell and Oncolytic Virotherapy for Cancer Treatment. Mol. Ther. 2021, 29, 505–520. [Google Scholar] [CrossRef] [PubMed]
- Mondal, M.; Guo, J.; He, P.; Zhou, D. Recent advances of oncolytic virus in cancer therapy. Hum. Vaccines Immunother. 2020, 16, 2389–2402. [Google Scholar] [CrossRef] [PubMed]
- Sivanandam, V.; LaRocca, C.J.; Chen, N.G.; Fong, Y.; Warner, S.G. Oncolytic Viruses and Immune Checkpoint Inhibition: The Best of Both Worlds. Mol. Ther. Oncolytics 2019, 13, 93–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zou, P.; Tang, R.; Luo, M. Oncolytic virotherapy, alone or in combination with immune checkpoint inhibitors, for advanced melanoma: A systematic review and meta-analysis. Int. Immunopharmacol. 2020, 78, 106050. [Google Scholar] [CrossRef] [PubMed]
- Roma-Rodrigues, C.; Mendes, R.; Baptista, P.; Fernandes, A. Targeting Tumor Microenvironment for Cancer Therapy. Int. J. Mol. Sci. 2019, 20, 840. [Google Scholar] [CrossRef] [Green Version]
- Zhang, B.; Wang, X.; Cheng, P. Remodeling of Tumor Immune Microenvironment by Oncolytic Viruses. Front. Oncol. 2021, 10, 3478. [Google Scholar] [CrossRef] [PubMed]
- Chaudhary, B.; Elkord, E. Regulatory T Cells in the Tumor Microenvironment and Cancer Progression: Role and Therapeutic Targeting. Vaccines 2016, 4, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Viehl, C.T.; Moore, T.T.; Liyanage, U.K.; Frey, D.M.; Ehlers, J.P.; Eberlein, T.J.; Goedegebuure, P.S.; Linehan, D.C. Depletion of CD4+CD25+ regulatory T cells promotes a tumor-specific immune response in pancreas cancer-bearing mice. Ann. Surg. Oncol. 2006, 13, 1252–1258. [Google Scholar] [CrossRef]
- Lun, X.Q.; Jang, J.-H.; Tang, N.; Deng, H.; Head, R.; Bell, J.C.; Stojdl, D.F.; Nutt, C.L.; Senger, D.L.; Forsyth, P.A.; et al. Efficacy of systemically administered oncolytic vaccinia virotherapy for malignant gliomas is enhanced by combination therapy with rapamycin or cyclophosphamide. Clin. Cancer Res. 2009, 15, 2777–2788. [Google Scholar] [CrossRef] [Green Version]
- Fulci, G.; Breymann, L.; Gianni, D.; Kurozomi, K.; Rhee, S.S.; Yu, J.; Kaur, B.; Louis, D.N.; Weissleder, R.; Caligiuri, M.A.; et al. Cyclophosphamide enhances glioma virotherapy by inhibiting innate immune responses. Proc. Natl. Acad. Sci. USA 2006, 103, 12873–12878. [Google Scholar] [CrossRef] [Green Version]
- Ikeda, K.; Ichikawa, T.; Wakimoto, H.; Silver, J.S.; Deisboeck, T.S.; Finkelstein, D.; Harsh, G.R.; Louis, D.N.; Bartus, R.T.; Hochberg, F.H.; et al. Oncolytic virus therapy of multiple tumors in the brain requires suppression of innate and elicited antiviral responses. Nat. Med. 1999, 5, 881–887. [Google Scholar] [CrossRef]
- Peng, K.-W.; Myers, R.; Greenslade, A.; Mader, E.; Greiner, S.; Federspiel, M.J.; Dispenzieri, A.; Russell, S.J. Using clinically approved cyclophosphamide regimens to control the humoral immune response to oncolytic viruses. Gene Ther. 2013, 20, 255–261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cerullo, V.; Diaconu, I.; Kangasniemi, L.; Rajecki, M.; Escutenaire, S.; Koski, A.; Romano, V.; Rouvinen, N.; Tuuminen, T.; Laasonen, L.; et al. Immunological Effects of Low-dose Cyclophosphamide in Cancer Patients Treated with Oncolytic Adenovirus. Mol. Ther. 2011, 19, 1737–1746. [Google Scholar] [CrossRef] [Green Version]
- Ghiringhelli, F.; Larmonier, N.; Schmitt, E.; Parcellier, A.; Cathelin, D.; Garrido, C.; Chauffert, B.; Solary, E.; Bonnotte, B.; Martin, F. CD4+CD25+ regulatory T cells suppress tumor immunity but are sensitive to cyclophosphamide which allows immunotherapy of established tumors to be curative. Eur. J. Immunol. 2004, 34, 336–344. [Google Scholar] [CrossRef] [PubMed]
- Madondo, M.T.; Quinn, M.; Plebanski, M. Low dose cyclophosphamide: Mechanisms of T cell modulation. Cancer Treat. Rev. 2016, 42, 3–9. [Google Scholar] [CrossRef] [PubMed]
- Pol, J.G.; Atherton, M.J.; Stephenson, K.B.; Bridle, B.W.; Workenhe, S.T.; Kazdhan, N.; McGray, A.J.R.; Wan, Y.; Kroemer, G.; Lichty, B.D. Enhanced immunotherapeutic profile of oncolytic virus-based cancer vaccination using cyclophosphamide preconditioning. J. Immunother. Cancer 2020, 8. [Google Scholar] [CrossRef]
- Roland, C.L.; Lynn, K.D.; Toombs, J.E.; Dineen, S.P.; Udugamasooriya, D.G.; Brekken, R.A. Cytokine levels correlate with immune cell infiltration after anti-VEGF therapy in preclinical mouse models of breast cancer. PLoS ONE 2009, 4, e7669. [Google Scholar] [CrossRef]
- Zhao, Q.; Guo, J.; Wang, G.; Chu, Y.; Hu, X. Suppression of immune regulatory cells with combined therapy of celecoxib and sunitinib in renal cell carcinoma. Oncotarget 2017, 8, 1668–1677. [Google Scholar] [CrossRef] [Green Version]
- Zhao, Y.; Ma, J.; Fan, Y.; Wang, Z.; Tian, R.; Ji, W.; Zhang, F.; Niu, R. TGF-β transactivates EGFR and facilitates breast cancer migration and invasion through canonical Smad3 and ERK/Sp1 signaling pathways. Mol. Oncol. 2018, 12, 305–321. [Google Scholar] [CrossRef]
- Akhurst, R.J.; Hata, A. Targeting the TGFβ signalling pathway in disease. Nat. Rev. Drug Discov. 2012, 11, 790–811. [Google Scholar] [CrossRef] [Green Version]
- Pickup, M.; Novitskiy, S.; Moses, H.L. The roles of TGFβ in the tumour microenvironment. Nat. Rev. Cancer 2013, 13, 788–799. [Google Scholar] [CrossRef] [Green Version]
- Padua, D.; Massagué, J. Roles of TGFβ in metastasis. Cell Res. 2009, 19, 89–102. [Google Scholar] [CrossRef] [PubMed]
- Yang, L.; Pang, Y.; Moses, H.L. TGF-β and immune cells: An important regulatory axis in the tumor microenvironment and progression. Trends Immunol. 2010, 31, 220–227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Groeneveldt, C.; van Hall, T.; van der Burg, S.H.; ten Dijke, P.; van Montfoort, N. Immunotherapeutic Potential of TGF-β Inhibition and Oncolytic Viruses. Trends Immunol. 2020, 41, 406–420. [Google Scholar] [CrossRef] [PubMed]
- Hamashima, Y.; Saitoh, M.; Node, M.; Hanyu, A.; Tojo, M.; Imamura, T.; Miyazono, K.; Kajimoto, T. The ALK-5 inhibitor A-83-01 inhibits Smad signaling and epithelial-to-mesenchymal transition by transforming growth factor-beta. Cancer Sci. 2005, 96, 791–800. [Google Scholar] [CrossRef]
- Hutzen, B.; Chen, C.Y.; Wang, P.Y.; Sprague, L.; Swain, H.M.; Love, J.; Conner, J.; Boon, L.; Cripe, T.P. TGF-β Inhibition Improves Oncolytic Herpes Viroimmunotherapy in Murine Models of Rhabdomyosarcoma. Mol. Ther. Oncolytics 2017, 7, 17–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yingling, J.M.; McMillen, W.T.; Yan, L.; Huang, H.; Sawyer, J.S.; Graff, J.; Clawson, D.K.; Britt, K.S.; Anderson, B.D.; Beight, D.W.; et al. Preclinical assessment of galunisertib (LY2157299 monohydrate), a first-in-class transforming growth factor-β receptor type I inhibitor. Oncotarget 2018, 9, 6659–6677. [Google Scholar] [CrossRef] [Green Version]
- Esaki, S.; Nigim, F.; Moon, E.; Luk, S.; Kiyokawa, J.; Curry, W.; Cahill, D.P.; Chi, A.S.; Iafrate, A.J.; Martuza, R.L.; et al. Blockade of transforming growth factor-β signaling enhances oncolytic herpes simplex virus efficacy in patient-derived recurrent glioblastoma models. Int. J. Cancer 2017, 141, 2348–2358. [Google Scholar] [CrossRef] [Green Version]
- Ishigami, I.; Shuwari, N.; Kaminade, T.; Mizuguchi, H.; Sakurai, F. A TGFβ Signaling Inhibitor, SB431542, inhibits reovirus-mediated lysis of human hepatocellular carcinoma cells in a TGFβ-independent Manner. Anticancer Res. 2021, 41, 2431–2440. [Google Scholar] [CrossRef]
- Huang, K.C.-Y.; Chiang, S.-F.; Yang, P.-C.; Ke, T.-W.; Chen, T.-W.; Hu, C.-H.; Huang, Y.-W.; Chang, H.-Y.; Chen, W.T.-L.; Chao, K.S.C. Immunogenic Cell Death by the Novel Topoisomerase I Inhibitor TLC388 Enhances the Therapeutic Efficacy of Radiotherapy. Cancers 2021, 13, 1218. [Google Scholar] [CrossRef]
- Haggerty, T.J.; Dunn, I.S.; Rose, L.B.; Newton, E.E.; Martin, S.; Riley, J.L.; Kurnick, J.T. Topoisomerase inhibitors modulate expression of melanocytic antigens and enhance T cell recognition of tumor cells. Cancer Immunol. Immunother. 2011, 60, 133–144. [Google Scholar] [CrossRef] [Green Version]
- Workenhe, S.T.; Pol, J.G.; Lichty, B.D.; Cummings, D.T.; Mossman, K.L. Combining Oncolytic HSV-1 with Immunogenic Cell Death-Inducing Drug Mitoxantrone Breaks Cancer Immune Tolerance and Improves Therapeutic Efficacy. Cancer Immunol. Res. 2013, 1, 309–319. [Google Scholar] [CrossRef] [Green Version]
- Fan, J.; Jiang, H.; Cheng, L.; Ma, B.; Liu, R. Oncolytic herpes simplex virus and temozolomide synergistically inhibit breast cancer cell tumorigenesis invitro and in vivo. Oncol. Lett. 2021, 21, 1-1. [Google Scholar] [CrossRef]
- Saha, D.; Rabkin, S.D.; Martuza, R.L. Temozolomide antagonizes oncolytic immunovirotherapy in glioblastoma. J. Immunother. Cancer 2020, 8, 345. [Google Scholar] [CrossRef]
- Kumar, S.; Wu, J.; Wang, F.; Wang, L.; Chen, L.; Sokirniy, I.; Wang, H.; Sterner, D.; Grove, C.; Cunnion, B.; et al. Abstract B202: Covalent irreversible usp7 inhibitors for cancer immunotherapy. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics. Mol. Cancer Ther. 2018, 17, B202. [Google Scholar]
- Fu, C.; Zhu, X.; Xu, P.; Li, Y. Pharmacological inhibition of USP7 promotes antitumor immunity and contributes to colon cancer therapy. OncoTargets Ther. 2019, 12, 609–617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hanna, B.S.; Roessner, P.M.; Scheffold, A.; Jebaraj, B.M.C.; Demerdash, Y.; Öztürk, S.; Lichter, P.; Stilgenbauer, S.; Seiffert, M. PI3Kδ inhibition modulates regulatory and effector T-cell differentiation and function in chronic lymphocytic leukemia. Leukemia 2018, 33, 1427–1438. [Google Scholar] [CrossRef]
- De Almeida Nagata, D.E.; Chiang, E.Y.; Jhunjhunwala, S.; Caplazi, P.; Arumugam, V.; Modrusan, Z.; Chan, E.; Merchant, M.; Jin, L.; Arnott, D.; et al. Regulation of Tumor-Associated Myeloid Cell Activity by CBP/EP300 Bromodomain Modulation of H3K27 Acetylation. Cell Rep. 2019, 27, 269–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shi, T.; Song, X.; Wang, Y.; Liu, F.; Wei, J. Combining Oncolytic Viruses with Cancer Immunotherapy: Establishing a New Generation of Cancer Treatment. Front. Immunol. 2020, 11, 683. [Google Scholar] [CrossRef]
- Marchini, A.; Scott, E.M.; Rommelaere, J. Overcoming barriers in oncolytic virotherapy with HDAC inhibitors and immune checkpoint blockade. Viruses 2016, 8, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rojas, J.J.; Sampath, P.; Hou, W.; Thorne, S.H. Defining effective combinations of immune checkpoint blockade and oncolytic virotherapy. Clin. Cancer Res. 2015, 21, 5543–5551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sasikumar, P.G.; Ramachandra, M. Small-molecule antagonists of the immune checkpoint pathways: Concept to clinic. Future Med. Chem. 2017, 9, 1305–1308. [Google Scholar] [CrossRef] [PubMed]
- Sasikumar, P.G.; Ramachandra, M. Small-Molecule Immune Checkpoint Inhibitors Targeting PD-1/PD-L1 and Other Emerging Checkpoint Pathways. BioDrugs 2018, 32, 481–497. [Google Scholar] [CrossRef] [PubMed]
- Chen, S.; Song, Z.; Zhang, A. Small-Molecule Immuno-Oncology Therapy: Advances, Challenges and New Directions. Curr. Top. Med. Chem. 2019, 19, 180–185. [Google Scholar] [CrossRef]
- Park, J.-J.; Thi, E.P.; Carpio, V.H.; Bi, Y.; Cole, A.G.; Dorsey, B.D.; Fan, K.; Harasym, T.; Iott, C.L.; Kadhim, S.; et al. Checkpoint inhibition through small molecule-induced internalization of programmed death-ligand 1. Nat. Commun. 2021, 12, 1222. [Google Scholar] [CrossRef] [PubMed]
- Ishikawa, H.; Ma, Z.; Barber, G.N. STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature 2009, 461, 788–792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ishikawa, H.; Barber, G.N. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature 2008, 455, 674–678. [Google Scholar] [CrossRef] [PubMed]
- Lau, A.; Gray, E.E.; Brunette, R.L.; Stetson, D.B. DNA tumor virus oncogenes antagonize the cGAS-STING DNA-sensing pathway. Science 2015, 350, 568–571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lam, E.; Stein, S.; Falck-Pedersen, E. Adenovirus Detection by the cGAS/STING/TBK1 DNA Sensing Cascade. J. Virol. 2014, 88, 974–981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Woo, S.R.; Fuertes, M.B.; Corrales, L.; Spranger, S.; Furdyna, M.J.; Leung, M.Y.K.; Duggan, R.; Wang, Y.; Barber, G.N.; Fitzgerald, K.A.; et al. STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity 2014, 41, 830–842. [Google Scholar] [CrossRef] [Green Version]
- Corrales, L.; McWhirter, S.M.; Dubensky, T.W.; Gajewski, T.F. The host STING pathway at the interface of cancer and immunity. J. Clin. Investig. 2016, 126, 2404–2411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deng, L.; Liang, H.; Xu, M.; Yang, X.; Burnette, B.; Arina, A.; Li, X.D.; Mauceri, H.; Beckett, M.; Darga, T.; et al. STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity 2014, 41, 843–852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Demaria, O.; De Gassart, A.; Coso, S.; Gestermann, N.; Di Domizio, J.; Flatz, L.; Gaide, O.; Michielin, O.; Hwu, P.; Petrova, T.V.; et al. STING activation of tumor endothelial cells initiates spontaneous and therapeutic antitumor immunity. Proc. Natl. Acad. Sci. USA 2015, 112, 15408–15413. [Google Scholar] [CrossRef] [Green Version]
- Jiang, M.; Jiang, M.; Chen, P.; Chen, P.; Wang, L.; Li, W.; Chen, B.; Liu, Y.; Liu, Y.; Wang, H.; et al. CGAS-STING, an important pathway in cancer immunotherapy. J. Hematol. Oncol. 2020, 13, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Pan, B.S.; Perera, S.A.; Piesvaux, J.A.; Presland, J.P.; Schroeder, G.K.; Cumming, J.N.; Wesley Trotter, B.; Altman, M.D.; Buevich, A.V.; Cash, B.; et al. An orally available non-nucleotide STING agonist with antitumor activity. Science 2020, 369. [Google Scholar] [CrossRef]
- Chin, E.N.; Yu, C.; Vartabedian, V.F.; Jia, Y.; Kumar, M.; Gamo, A.M.; Vernier, W.; Ali, S.H.; Kissai, M.; Lazar, D.C.; et al. Antitumor activity of a systemic STING-activating non-nucleotide cGAMP mimetic. Science 2020, 369, 993–999. [Google Scholar] [CrossRef] [PubMed]
- Xia, T.; Konno, H.; Ahn, J.; Barber, G.N. Deregulation of STING Signaling in Colorectal Carcinoma Constrains DNA Damage Responses and Correlates with Tumorigenesis. Cell Rep. 2016, 14, 282–297. [Google Scholar] [CrossRef] [Green Version]
- Xia, T.; Konno, H.; Barber, G.N. Recurrent loss of STING signaling in melanoma correlates with susceptibility to viral oncolysis. Cancer Res. 2016, 76, 6747–6759. [Google Scholar] [CrossRef] [Green Version]
- De Queiroz, N.M.G.P.; Xia, T.; Konno, H.; Barber, G.N. Ovarian cancer cells commonly exhibit defective STING signaling which affects sensitivity to viral oncolysis. Mol. Cancer Res. 2019, 17, 974–986. [Google Scholar] [CrossRef] [PubMed]
- Eaglesham, J.B.; Kranzusch, P.J. Conserved strategies for pathogen evasion of cGAS–STING immunity. Curr. Opin. Immunol. 2020, 66, 27–34. [Google Scholar] [CrossRef]
- Ahn, J.; Barber, G.N. STING signaling and host defense against microbial infection. Exp. Mol. Med. 2019, 51, 1–10. [Google Scholar] [CrossRef]
- Russell, L.; Peng, K.W.; Russell, S.J.; Diaz, R.M. Oncolytic Viruses: Priming Time for Cancer Immunotherapy. BioDrugs 2019, 33, 485–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zaikos, T.D.; Painter, M.M.; Sebastian Kettinger, N.T.; Terry, V.H.; Collins, K.L. Class 1-Selective Histone Deacetylase (HDAC) Inhibitors Enhance HIV Latency Reversal while Preserving the Activity of HDAC Isoforms Necessary for Maximal HIV Gene Expression. J. Virol. 2018, 92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nehme, Z.; Pasquereau, S.; Herbein, G. Control of viral infections by epigenetic-targeted therapy. Clin. Epigenet. 2019, 11, 1–17. [Google Scholar] [CrossRef] [Green Version]
- Wang, L.; Beier, U.H.; Akimova, T.; Dahiya, S.; Han, R.; Samanta, A.; Levine, M.H.; Hancock, W.W. Histone/protein deacetylase inhibitor therapy for enhancement of Foxp3+ T-regulatory cell function posttransplantation. Am. J. Transplant. 2018, 18, 1596–1603. [Google Scholar] [CrossRef] [PubMed]
- Chiu, M.; Armstrong, E.J.L.; Jennings, V.; Foo, S.; Crespo-Rodriguez, E.; Bozhanova, G.; Patin, E.C.; McLaughlin, M.; Mansfield, D.; Baker, G.; et al. Combination therapy with oncolytic viruses and immune checkpoint inhibitors. Expert Opin. Biol. Ther. 2020, 20, 635–652. [Google Scholar] [CrossRef] [PubMed]
- Hwang, J.K.; Hong, J.; Yun, C.O. Oncolytic viruses and immune checkpoint inhibitors: Preclinical developments to clinical trials. Int. J. Mol. Sci. 2020, 21, 8627. [Google Scholar] [CrossRef] [PubMed]
HDI | OV | Tumor | References |
---|---|---|---|
entinostat | VSV | B16-F10, CT26, L363(MM), HT29, M14, PC3, SW620, 4T1 | [17,129,130,140] |
vorinostat | VSV | B16-F10 | [130,141] |
trichostatin | HSV, vaccinia | SAS, Ca9-22, HSC, HCT116, B26-F10, U87, SW480, HeLa | [123,142,143] |
valproate | HSV, H1 | U87, AGS1, U251, Gli36, HeLa | [132,133,134,144] |
Scriptaid & LBH589 | Adenovirus | Glioblastoma | [145] |
SMC | OV | Tumor Model | References |
---|---|---|---|
LCL-161 | VSV, M1 | EMT-6, CT26, MOC-11, SNB75, SG539, BTIC, HCT-116, Kym-1, M-3 | [154,183,186,188,189,190,191] |
Birinapant | M1 | HCT-116, Huh-7 | [183,191] |
Virus Family | Oncolytic Virus Design | Small Molecule Compound | Indication | Phase/Status | CinicalTrials.gov Reference |
---|---|---|---|---|---|
HSV | rQNestin34.5v.2 HSV-1 with viral gene ICP34.5 under glioma specific nestin promoter control | Cyclophosphamide | Glioma | I recruiting | NCT03152318 |
TBI-1401(HF10) naturally attenuated HSV-1 | Gemcitabine + nab-pactitaxel | Pancreatic cancer | I not recruiting | NCT03252808 | |
AdV | ONCOS-102 Ad5/3-24 expressing a GM-CSF transgene | Cyclophosphamide | Melanoma | I not recruiting | NCT03003676 |
ONCOS-102 Ad5/3-24 expressing a GM-CSF transgene | Cyclophosphamide | Mesothelioma | II not recruiting | NCT02879669 | |
LOAd703 AdV5/35 expressing TMZ-CD40L and 4-1BBL transgenes | Gemcitabine + nab-pactitaxel | Pancreatic cancer | I/IIa recruiting | NCT02705196 | |
RV | Pelareorep Unmodified human reovirus typ 3 (Dearing strain) | Paclitaxel | Breast cancer | II recruiting | NCT04215146 |
Pelareorep Unmodified human reovirus typ 3 (Dearing strain) | Carfilzomib | Multiple myeloma | I recruiting | NCT03605719 | |
VV | JX-594 (Pexa-Vec) Wyeth strain VV expressing a GM-CSF transgene | Cyclophosphamide | Sarcoma, breast cancer | II recruiting | NCT02630368 |
VSV | VSV-hIFN-NIS VSV expressing an interferon and a sodium iodide symporter transgene | Ruxolitinib | Multiple myeloma, AML, T-cell lymphoma | I recruiting | NCT03017820 |
VSV-hIFN-NIS VSV expressing an interferon and a sodium iodide symporter transgene | Ruxolitinib | Endometrial cancer | I recruiting | NCT03120624 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Spiesschaert, B.; Angerer, K.; Park, J.; Wollmann, G. Combining Oncolytic Viruses and Small Molecule Therapeutics: Mutual Benefits. Cancers 2021, 13, 3386. https://doi.org/10.3390/cancers13143386
Spiesschaert B, Angerer K, Park J, Wollmann G. Combining Oncolytic Viruses and Small Molecule Therapeutics: Mutual Benefits. Cancers. 2021; 13(14):3386. https://doi.org/10.3390/cancers13143386
Chicago/Turabian StyleSpiesschaert, Bart, Katharina Angerer, John Park, and Guido Wollmann. 2021. "Combining Oncolytic Viruses and Small Molecule Therapeutics: Mutual Benefits" Cancers 13, no. 14: 3386. https://doi.org/10.3390/cancers13143386
APA StyleSpiesschaert, B., Angerer, K., Park, J., & Wollmann, G. (2021). Combining Oncolytic Viruses and Small Molecule Therapeutics: Mutual Benefits. Cancers, 13(14), 3386. https://doi.org/10.3390/cancers13143386