Targeting EBV Episome for Anti-Cancer Therapy: Emerging Strategies and Challenges
Abstract
:1. Introduction
2. Targeting the EBV Episome for Anti-Cancer Therapy
2.1. Targeting EBV Episome Maintenance Protein EBNA1
2.2. Targeting Viral Genome by Genome Editing
2.3. Lytic Induction Therapy
3. Limitations and Future Perspective
Author Contributions
Funding
Conflicts of Interest
References
- Epstein, M.A.; Achong, B.G.; Barr, Y.M. Virus particles in cultured lymphoblasts from burkitt’s lymphoma. Lancet 1964, 1, 702–703. [Google Scholar] [CrossRef] [PubMed]
- Magrath, I. Epidemiology: Clues to the pathogenesis of Burkitt lymphoma. Br. J. Haematol. 2012, 156, 744–756. [Google Scholar] [CrossRef] [PubMed]
- Grande, B.M.; Gerhard, D.S.; Jiang, A.; Griner, N.B.; Abramson, J.S.; Alexander, T.B.; Allen, H.; Ayers, L.W.; Bethony, J.M.; Bhatia, K.; et al. Genome-wide discovery of somatic coding and noncoding mutations in pediatric endemic and sporadic Burkitt lymphoma. Blood 2019, 133, 1313–1324. [Google Scholar] [CrossRef] [PubMed]
- Balfour, H.H., Jr.; Sifakis, F.; Sliman, J.A.; Knight, J.A.; Schmeling, D.O.; Thomas, W. Age-specific prevalence of Epstein-Barr virus infection among individuals aged 6-19 years in the United States and factors affecting its acquisition. J. Infect. Dis. 2013, 208, 1286–1293. [Google Scholar] [CrossRef]
- Hjalgrim, H.; Askling, J.; Rostgaard, K.; Hamilton-Dutoit, S.; Frisch, M.; Zhang, J.S.; Madsen, M.; Rosdahl, N.; Konradsen, H.B.; Storm, H.H.; et al. Characteristics of Hodgkin’s lymphoma after infectious mononucleosis. N. Engl. J. Med. 2003, 349, 1324–1332. [Google Scholar] [CrossRef] [PubMed]
- Xu, M.; Feng, R.; Liu, Z.; Zhou, X.; Chen, Y.; Cao, Y.; Valeri, L.; Li, Z.; Liu, Z.; Cao, S.M.; et al. Host genetic variants, Epstein-Barr virus subtypes, and the risk of nasopharyngeal carcinoma: Assessment of interaction and mediation. Cell Genom. 2024, 4, 100474. [Google Scholar] [CrossRef]
- Zeng, Y.; Luo, C.L.; Lin, G.W.; Li, F.; Bai, X.; Ko, J.M.; Xiong, L.; Liu, Y.; He, S.; Jiang, J.X.; et al. Whole-exome sequencing association study reveals genetic effects on tumor microenvironment components in nasopharyngeal carcinoma. J. Clin. Investig. 2025, 135. [Google Scholar] [CrossRef] [PubMed]
- Young, L.S.; Dawson, C.W. Epstein-Barr virus and nasopharyngeal carcinoma. Chin. J. Cancer 2014, 33, 581–590. [Google Scholar] [CrossRef] [PubMed]
- Burke, A.P.; Yen, T.S.; Shekitka, K.M.; Sobin, L.H. Lymphoepithelial carcinoma of the stomach with Epstein-Barr virus demonstrated by polymerase chain reaction. Mod. Pathol. 1990, 3, 377–380. [Google Scholar]
- Stanland, L.J.; Luftig, M.A. The Role of EBV-Induced Hypermethylation in Gastric Cancer Tumorigenesis. Viruses 2020, 12, 1222. [Google Scholar] [CrossRef] [PubMed]
- Kojima, Y.; Hamada, M.; Naruse, A.; Goto, K.; Khine, H.T.; Arai, H.; Akutsu, Y.; Satou, A.; Nakaguro, M.; Kato, S.; et al. The landscape of 142 Epstein-Barr viral whole genomes in gastric cancer. J. Gastroenterol. 2024, 60, 55–65. [Google Scholar] [CrossRef]
- Young, L.S.; Yap, L.F.; Murray, P.G. Epstein-Barr virus: More than 50 years old and still providing surprises. Nat. Rev. Cancer 2016, 16, 789–802. [Google Scholar] [CrossRef] [PubMed]
- Damania, B.; Kenney, S.C.; Raab-Traub, N. Epstein-Barr virus: Biology and clinical disease. Cell 2022, 185, 3652–3670. [Google Scholar] [CrossRef] [PubMed]
- Bjornevik, K.; Münz, C.; Cohen, J.I.; Ascherio, A. Epstein-Barr virus as a leading cause of multiple sclerosis: Mechanisms and implications. Nat. Rev. Neurol. 2023, 19, 160–171. [Google Scholar] [CrossRef] [PubMed]
- Soldan, S.S.; Lieberman, P.M. Epstein-Barr virus and multiple sclerosis. Nat. Rev. Microbiol. 2023, 21, 51–64. [Google Scholar] [CrossRef] [PubMed]
- Lanz, T.V.; Brewer, R.C.; Ho, P.P.; Moon, J.S.; Jude, K.M.; Fernandez, D.; Fernandes, R.A.; Gomez, A.M.; Nadj, G.S.; Bartley, C.M.; et al. Clonally expanded B cells in multiple sclerosis bind EBV EBNA1 and GlialCAM. Nature 2022, 603, 321–327. [Google Scholar] [CrossRef] [PubMed]
- Bjornevik, K.; Cortese, M.; Healy, B.C.; Kuhle, J.; Mina, M.J.; Leng, Y.; Elledge, S.J.; Niebuhr, D.W.; Scher, A.I.; Munger, K.L.; et al. Longitudinal analysis reveals high prevalence of Epstein-Barr virus associated with multiple sclerosis. Science 2022, 375, 296–301. [Google Scholar] [CrossRef]
- Lieberman, P.M. Keeping it quiet: Chromatin control of gammaherpesvirus latency. Nat. Rev. Microbiol. 2013, 11, 863–875. [Google Scholar] [CrossRef] [PubMed]
- Shimizu, N.; Yoshiyama, H.; Takada, K. Clonal propagation of Epstein-Barr virus (EBV) recombinants in EBV-negative Akata cells. J. Virol. 1996, 70, 7260–7263. [Google Scholar] [CrossRef] [PubMed]
- Nanbo, A.; Katano, H.; Kataoka, M.; Hoshina, S.; Sekizuka, T.; Kuroda, M.; Ohba, Y. Infection of Epstein–Barr Virus in Type III Latency Modulates Biogenesis of Exosomes and the Expression Profile of Exosomal miRNAs in the Burkitt Lymphoma Mutu Cell Lines. Cancers 2018, 10, 237. [Google Scholar] [CrossRef] [PubMed]
- Pan, Y.R.; Fang, C.Y.; Chang, Y.S.; Chang, H.Y. Analysis of Epstein-Barr virus gene expression upon phorbol ester and hydroxyurea treatment by real-time quantitative PCR. Arch. Virol. 2005, 150, 755–770. [Google Scholar] [CrossRef] [PubMed]
- Zuo, L.; Yu, H.; Liu, L.; Tang, Y.; Wu, H.; Yang, J.; Zhu, M.; Du, S.; Zhao, L.; Cao, L.; et al. The copy number of Epstein-Barr virus latent genome correlates with the oncogenicity by the activation level of LMP1 and NF-κB. Oncotarget 2015, 6, 41033–41044. [Google Scholar] [CrossRef] [PubMed]
- Houldcroft, C.J.; Petrova, V.; Liu, J.Z.; Frampton, D.; Anderson, C.A.; Gall, A.; Kellam, P. Host genetic variants and gene expression patterns associated with Epstein-Barr virus copy number in lymphoblastoid cell lines. PLoS ONE 2014, 9, e108384. [Google Scholar] [CrossRef]
- Kim, D.N.; Seo, M.K.; Choi, H.; Kim, S.Y.; Shin, H.J.; Yoon, A.R.; Tao, Q.; Rha, S.Y.; Lee, S.K. Characterization of naturally Epstein-Barr virus-infected gastric carcinoma cell line YCCEL1. J. Gen. Virol. 2013, 94, 497–506. [Google Scholar] [CrossRef] [PubMed]
- Dheekollu, J.; Wiedmer, A.; Ayyanathan, K.; Deakyne, J.S.; Messick, T.E.; Lieberman, P.M. Cell-cycle-dependent EBNA1-DNA crosslinking promotes replication termination at oriP and viral episome maintenance. Cell 2021, 184, 643–654.e613. [Google Scholar] [CrossRef] [PubMed]
- Mei, Y.; Messick, T.E.; Dheekollu, J.; Kim, H.J.; Molugu, S.; Muñoz, L.J.C.; Moiskeenkova-Bell, V.; Murakami, K.; Lieberman, P.M. Cryo-EM Structure and Functional Studies of EBNA1 Binding to the Family of Repeats and Dyad Symmetry Elements of Epstein-Barr Virus oriP. J. Virol. 2022, 96, e0094922. [Google Scholar] [CrossRef] [PubMed]
- Jiang, L.; Xie, C.; Lung, H.L.; Lo, K.W.; Law, G.L.; Mak, N.K.; Wong, K.L. EBNA1-targeted inhibitors: Novel approaches for the treatment of Epstein-Barr virus-associated cancers. Theranostics 2018, 8, 5307–5319. [Google Scholar] [CrossRef]
- Li, J.S.Z.; Abbasi, A.; Kim, D.H.; Lippman, S.M.; Alexandrov, L.B.; Cleveland, D.W. Chromosomal fragile site breakage by EBV-encoded EBNA1 at clustered repeats. Nature 2023, 616, 504–509. [Google Scholar] [CrossRef]
- Lu, F.; Wikramasinghe, P.; Norseen, J.; Tsai, K.; Wang, P.; Showe, L.; Davuluri, R.V.; Lieberman, P.M. Genome-wide analysis of host-chromosome binding sites for Epstein-Barr Virus Nuclear Antigen 1 (EBNA1). Virol. J. 2010, 7, 262. [Google Scholar] [CrossRef] [PubMed]
- Ian, M.X.; Lan, S.Z.; Cheng, Z.F.; Dan, H.; Qiong, L.H. Suppression of EBNA1 expression inhibits growth of EBV-positive NK/T cell lymphoma cells. Cancer Biol. Ther. 2008, 7, 1602–1606. [Google Scholar] [CrossRef] [PubMed]
- Li, N.; Thompson, S.; Schultz, D.C.; Zhu, W.; Jiang, H.; Luo, C.; Lieberman, P.M. Discovery of selective inhibitors against EBNA1 via high throughput in silico virtual screening. PLoS ONE 2010, 5, e10126. [Google Scholar] [CrossRef] [PubMed]
- Thompson, S.; Messick, T.; Schultz, D.C.; Reichman, M.; Lieberman, P.M. Development of a high-throughput screen for inhibitors of Epstein-Barr virus EBNA1. J. Biomol. Screen. 2010, 15, 1107–1115. [Google Scholar] [CrossRef] [PubMed]
- Kang, M.S.; Lee, E.K.; Soni, V.; Lewis, T.A.; Koehler, A.N.; Srinivasan, V.; Kieff, E. Roscovitine inhibits EBNA1 serine 393 phosphorylation, nuclear localization, transcription, and episome maintenance. J. Virol. 2011, 85, 2859–2868. [Google Scholar] [CrossRef] [PubMed]
- Bochkarev, A.; Barwell, J.A.; Pfuetzner, R.A.; Furey, W., Jr.; Edwards, A.M.; Frappier, L. Crystal structure of the DNA-binding domain of the Epstein-Barr virus origin-binding protein EBNA 1. Cell 1995, 83, 39–46. [Google Scholar] [CrossRef] [PubMed]
- Cruickshank, J.; Shire, K.; Davidson, A.R.; Edwards, A.M.; Frappier, L. Two domains of the epstein-barr virus origin DNA-binding protein, EBNA1, orchestrate sequence-specific DNA binding. J. Biol. Chem. 2000, 275, 22273–22277. [Google Scholar] [CrossRef]
- Deakyne, J.S.; Malecka, K.A.; Messick, T.E.; Lieberman, P.M. Structural and Functional Basis for an EBNA1 Hexameric Ring in Epstein-Barr Virus Episome Maintenance. J. Virol. 2017, 91. [Google Scholar] [CrossRef]
- Kim, S.Y.; Song, K.A.; Kieff, E.; Kang, M.S. Small molecule and peptide-mediated inhibition of Epstein-Barr virus nuclear antigen 1 dimerization. Biochem. Biophys. Res. Commun. 2012, 424, 251–256. [Google Scholar] [CrossRef] [PubMed]
- Lee, E.K.; Kim, S.Y.; Noh, K.W.; Joo, E.H.; Zhao, B.; Kieff, E.; Kang, M.S. Small molecule inhibition of Epstein-Barr virus nuclear antigen-1 DNA binding activity interferes with replication and persistence of the viral genome. Antiviral Res. 2014, 104, 73–83. [Google Scholar] [CrossRef]
- Yasuda, A.; Noguchi, K.; Minoshima, M.; Kashiwazaki, G.; Kanda, T.; Katayama, K.; Mitsuhashi, J.; Bando, T.; Sugiyama, H.; Sugimoto, Y. DNA ligand designed to antagonize EBNA1 represses Epstein-Barr virus-induced immortalization. Cancer Sci. 2011, 102, 2221–2230. [Google Scholar] [CrossRef] [PubMed]
- Jiang, L.; Lui, Y.L.; Li, H.; Chan, C.F.; Lan, R.; Chan, W.L.; Lau, T.C.; Tsao, G.S.; Mak, N.K.; Wong, K.L. EBNA1-specific luminescent small molecules for the imaging and inhibition of latent EBV-infected tumor cells. Chem. Commun. 2014, 50, 6517–6519. [Google Scholar] [CrossRef]
- Jiang, L.; Lan, R.; Huang, T.; Chan, C.-F.; Li, H.; Lear, S.; Zong, J.; Wong, W.-Y.; Muk-Lan Lee, M.; Dow Chan, B.; et al. EBNA1-targeted probe for the imaging and growth inhibition of tumours associated with the Epstein–Barr virus. Nat. Biomed. Eng. 2017, 1, 42. [Google Scholar] [CrossRef]
- Messick, T.E.; Smith, G.R.; Soldan, S.S.; McDonnell, M.E.; Deakyne, J.S.; Malecka, K.A.; Tolvinski, L.; van den Heuvel, A.P.J.; Gu, B.W.; Cassel, J.A.; et al. Structure-based design of small-molecule inhibitors of EBNA1 DNA binding blocks Epstein-Barr virus latent infection and tumor growth. Sci. Transl. Med. 2019, 11, eaau5612. [Google Scholar] [CrossRef] [PubMed]
- Soldan, S.S.; Anderson, E.M.; Frase, D.M.; Zhang, Y.; Caruso, L.B.; Wang, Y.; Deakyne, J.S.; Gewurz, B.E.; Tempera, I.; Lieberman, P.M.; et al. EBNA1 inhibitors have potent and selective antitumor activity in xenograft models of Epstein-Barr virus-associated gastric cancer. Gastric Cancer 2021, 24, 1076–1088. [Google Scholar] [CrossRef] [PubMed]
- Davis, M.T.; Anders, N.M.; Colevas, A.D.; Messick, T.E.; Rudek, M.A. Validation of a robust and rapid liquid chromatography tandem mass spectrometric method for the quantitative analysis of VK-2019, a selective EBNA1 inhibitor. Biomed. Chromatogr. 2024, 38, e5775. [Google Scholar] [CrossRef] [PubMed]
- Han, Y.; Wu, F.; Zhang, Y.; Liu, J.; Wu, Y.; Wang, Y.; Jiang, X.; Chen, X.; Xu, W. Structure-based design of antibodies targeting the EBNA1 DNA-binding domain to block Epstein-Barr virus latent infection and tumor growth. MedComm 2024, 5, e739. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; Yang, Y.; Hong, W.; Huang, M.; Wu, M.; Zhao, X. Applications of genome editing technology in the targeted therapy of human diseases: Mechanisms, advances and prospects. Signal Transduct. Target. Ther. 2020, 5, 1. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Quake, S.R. RNA-guided endonuclease provides a therapeutic strategy to cure latent herpesviridae infection. Proc. Natl. Acad. Sci. USA 2014, 111, 13157–13162. [Google Scholar] [CrossRef] [PubMed]
- van Diemen, F.R.; Kruse, E.M.; Hooykaas, M.J.; Bruggeling, C.E.; Schürch, A.C.; van Ham, P.M.; Imhof, S.M.; Nijhuis, M.; Wiertz, E.J.; Lebbink, R.J. CRISPR/Cas9-Mediated Genome Editing of Herpesviruses Limits Productive and Latent Infections. PLoS Pathog. 2016, 12, e1005701. [Google Scholar] [CrossRef]
- Yuen, K.S.; Wang, Z.M.; Wong, N.M.; Zhang, Z.Q.; Cheng, T.F.; Lui, W.Y.; Chan, C.P.; Jin, D.Y. Suppression of Epstein-Barr virus DNA load in latently infected nasopharyngeal carcinoma cells by CRISPR/Cas9. Virus Res. 2018, 244, 296–303. [Google Scholar] [CrossRef]
- Huo, H.; Hu, G. CRISPR/Cas9-mediated LMP1 knockout inhibits Epstein-Barr virus infection and nasopharyngeal carcinoma cell growth. Infect. Agent. Cancer 2019, 14, 30. [Google Scholar] [CrossRef] [PubMed]
- Feng, W.H.; Israel, B.; Raab-Traub, N.; Busson, P.; Kenney, S.C. Chemotherapy induces lytic EBV replication and confers ganciclovir susceptibility to EBV-positive epithelial cell tumors. Cancer Res. 2002, 62, 1920–1926. [Google Scholar]
- Feng, W.H.; Westphal, E.; Mauser, A.; Raab-Traub, N.; Gulley, M.L.; Busson, P.; Kenney, S.C. Use of adenovirus vectors expressing Epstein-Barr virus (EBV) immediate-early protein BZLF1 or BRLF1 to treat EBV-positive tumors. J. Virol. 2002, 76, 10951–10959. [Google Scholar] [CrossRef] [PubMed]
- Fu, D.X.; Tanhehco, Y.; Chen, J.; Foss, C.A.; Fox, J.J.; Chong, J.M.; Hobbs, R.F.; Fukayama, M.; Sgouros, G.; Kowalski, J.; et al. Bortezomib-induced enzyme-targeted radiation therapy in herpesvirus-associated tumors. Nat. Med. 2008, 14, 1118–1122. [Google Scholar] [CrossRef]
- Westphal, E.M.; Blackstock, W.; Feng, W.; Israel, B.; Kenney, S.C. Activation of lytic Epstein-Barr virus (EBV) infection by radiation and sodium butyrate in vitro and in vivo: A potential method for treating EBV-positive malignancies. Cancer Res. 2000, 60, 5781–5788. [Google Scholar]
- Westphal, E.M.; Mauser, A.; Swenson, J.; Davis, M.G.; Talarico, C.L.; Kenney, S.C. Induction of lytic Epstein-Barr virus (EBV) infection in EBV-associated malignancies using adenovirus vectors in vitro and in vivo. Cancer Res. 1999, 59, 1485–1491. [Google Scholar]
- Yiu, S.P.T.; Dorothea, M.; Hui, K.F.; Chiang, A.K.S. Lytic Induction Therapy against Epstein-Barr Virus-Associated Malignancies: Past, Present, and Future. Cancers 2020, 12, 2142. [Google Scholar] [CrossRef] [PubMed]
- Li, R.; Liao, G.; Nirujogi, R.S.; Pinto, S.M.; Shaw, P.G.; Huang, T.C.; Wan, J.; Qian, J.; Gowda, H.; Wu, X.; et al. Phosphoproteomic Profiling Reveals Epstein-Barr Virus Protein Kinase Integration of DNA Damage Response and Mitotic Signaling. PLoS Pathog. 2015, 11, e1005346. [Google Scholar] [CrossRef]
- Zhang, K.; Lv, D.W.; Li, R. Conserved Herpesvirus Protein Kinases Target SAMHD1 to Facilitate Virus Replication. Cell Rep. 2019, 28, 449–459.e445. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.T.; Yang, P.W.; Lee, C.P.; Han, C.H.; Tsai, C.H.; Chen, M.R. Detection of Epstein-Barr virus BGLF4 protein kinase in virus replication compartments and virus particles. J. Gen. Virol. 2005, 86, 3215–3225. [Google Scholar] [CrossRef]
- Li, R.; Zhu, J.; Xie, Z.; Liao, G.; Liu, J.; Chen, M.R.; Hu, S.; Woodard, C.; Lin, J.; Taverna, S.D.; et al. Conserved herpesvirus kinases target the DNA damage response pathway and TIP60 histone acetyltransferase to promote virus replication. Cell Host Microbe 2011, 10, 390–400. [Google Scholar] [CrossRef] [PubMed]
- Zhu, J.; Liao, G.; Shan, L.; Zhang, J.; Chen, M.R.; Hayward, G.S.; Hayward, S.D.; Desai, P.; Zhu, H. Protein array identification of substrates of the Epstein-Barr virus protein kinase BGLF4. J. Virol. 2009, 83, 5219–5231. [Google Scholar] [CrossRef] [PubMed]
- Meng, Q.; Hagemeier, S.R.; Fingeroth, J.D.; Gershburg, E.; Pagano, J.S.; Kenney, S.C. The Epstein-Barr virus (EBV)-encoded protein kinase, EBV-PK, but not the thymidine kinase (EBV-TK), is required for ganciclovir and acyclovir inhibition of lytic viral production. J. Virol. 2010, 84, 4534–4542. [Google Scholar] [CrossRef]
- Li, R.; Hayward, S.D. Potential of protein kinase inhibitors for treating herpesvirus-associated disease. Trends Microbiol. 2013, 21, 286–295. [Google Scholar] [CrossRef] [PubMed]
- Colombo, B.M.; Benedetti, S.; Ottolenghi, S.; Mora, M.; Pollo, B.; Poli, G.; Finocchiaro, G. The “bystander effect”: Association of U-87 cell death with ganciclovir-mediated apoptosis of nearby cells and lack of effect in athymic mice. Hum. Gene Ther. 1995, 6, 763–772. [Google Scholar] [CrossRef]
- Haverkos, B.; Alpdogan, O.; Baiocchi, R.; Brammer, J.E.; Feldman, T.A.; Capra, M.; Brem, E.A.; Nair, S.; Scheinberg, P.; Pereira, J.; et al. Targeted therapy with nanatinostat and valganciclovir in recurrent EBV-positive lymphoid malignancies: A phase 1b/2 study. Blood Adv. 2023, 7, 6339–6350. [Google Scholar] [CrossRef]
- Cong, L.; Ran, F.A.; Cox, D.; Lin, S.; Barretto, R.; Habib, N.; Hsu, P.D.; Wu, X.; Jiang, W.; Marraffini, L.A.; et al. Multiplex genome engineering using CRISPR/Cas systems. Science 2013, 339, 819–823. [Google Scholar] [CrossRef]
- Gebre, M.; Nomburg, J.L.; Gewurz, B.E. CRISPR-Cas9 Genetic Analysis of Virus-Host Interactions. Viruses 2018, 10, 55. [Google Scholar] [CrossRef] [PubMed]
- Mali, P.; Yang, L.; Esvelt, K.M.; Aach, J.; Guell, M.; DiCarlo, J.E.; Norville, J.E.; Church, G.M. RNA-guided human genome engineering via Cas9. Science 2013, 339, 823–826. [Google Scholar] [CrossRef]
- Wang, L.W.; Trudeau, S.J.; Wang, C.; Gerdt, C.; Jiang, S.; Zhao, B.; Gewurz, B.E. Modulating Gene Expression in Epstein-Barr Virus (EBV)-Positive B Cell Lines with CRISPRa and CRISPRi. Curr. Protoc. Mol. Biol. 2018, 121, 31.13.1–31.13.18. [Google Scholar] [CrossRef] [PubMed]
- Cheng, A.W.; Wang, H.; Yang, H.; Shi, L.; Katz, Y.; Theunissen, T.W.; Rangarajan, S.; Shivalila, C.S.; Dadon, D.B.; Jaenisch, R. Multiplexed activation of endogenous genes by CRISPR-on, an RNA-guided transcriptional activator system. Cell Res. 2013, 23, 1163–1171. [Google Scholar] [CrossRef]
- Konermann, S.; Brigham, M.D.; Trevino, A.E.; Joung, J.; Abudayyeh, O.O.; Barcena, C.; Hsu, P.D.; Habib, N.; Gootenberg, J.S.; Nishimasu, H.; et al. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature 2015, 517, 583–588. [Google Scholar] [CrossRef]
- Maeder, M.L.; Linder, S.J.; Cascio, V.M.; Fu, Y.; Ho, Q.H.; Joung, J.K. CRISPR RNA-guided activation of endogenous human genes. Nat. Methods 2013, 10, 977–979. [Google Scholar] [CrossRef] [PubMed]
- Perez-Pinera, P.; Kocak, D.D.; Vockley, C.M.; Adler, A.F.; Kabadi, A.M.; Polstein, L.R.; Thakore, P.I.; Glass, K.A.; Ousterout, D.G.; Leong, K.W.; et al. RNA-guided gene activation by CRISPR-Cas9-based transcription factors. Nat. Methods 2013, 10, 973–976. [Google Scholar] [CrossRef] [PubMed]
- Kenney, S.C.; Mertz, J.E. Regulation of the latent-lytic switch in Epstein-Barr virus. Semin. Cancer Biol. 2014, 26, 60–68. [Google Scholar] [CrossRef]
- Sugiokto, F.G.; Li, R. Targeted eradication of EBV-positive cancer cells by CRISPR/dCas9-mediated EBV reactivation in combination with ganciclovir. mBio 2024, 15, e0079524. [Google Scholar] [CrossRef] [PubMed]
- Wu, M.; Hau, P.M.; Li, L.; Tsang, C.M.; Yang, Y.; Taghbalout, A.; Chung, G.T.; Hui, S.Y.; Tang, W.C.; Jillette, N.; et al. Synthetic BZLF1-targeted transcriptional activator for efficient lytic induction therapy against EBV-associated epithelial cancers. Nat. Commun. 2024, 15, 3729. [Google Scholar] [CrossRef]
- Bond, M.J.; Crews, C.M. Proteolysis targeting chimeras (PROTACs) come of age: Entering the third decade of targeted protein degradation. RSC Chem. Biol. 2021, 2, 725–742. [Google Scholar] [CrossRef]
- Békés, M.; Langley, D.R.; Crews, C.M. PROTAC targeted protein degraders: The past is prologue. Nat. Rev. Drug Discov. 2022, 21, 181–200. [Google Scholar] [CrossRef]
- Sakamoto, K.M.; Kim, K.B.; Kumagai, A.; Mercurio, F.; Crews, C.M.; Deshaies, R.J. Protacs: Chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradation. Proc. Natl. Acad. Sci. USA 2001, 98, 8554–8559. [Google Scholar] [CrossRef]
- Lu, J.; Qian, Y.; Altieri, M.; Dong, H.; Wang, J.; Raina, K.; Hines, J.; Winkler, J.D.; Crew, A.P.; Coleman, K.; et al. Hijacking the E3 Ubiquitin Ligase Cereblon to Efficiently Target BRD4. Chem. Biol. 2015, 22, 755–763. [Google Scholar] [CrossRef]
- Asmamaw Mengstie, M. Viral Vectors for the in Vivo Delivery of CRISPR Components: Advances and Challenges. Front. Bioeng. Biotechnol. 2022, 10, 895713. [Google Scholar] [CrossRef] [PubMed]
- Hamilton, J.R.; Chen, E.; Perez, B.S.; Sandoval Espinoza, C.R.; Kang, M.H.; Trinidad, M.; Ngo, W.; Doudna, J.A. In vivo human T cell engineering with enveloped delivery vehicles. Nat. Biotechnol. 2024, 42, 1684–1692. [Google Scholar] [CrossRef] [PubMed]
- Hamilton, J.R.; Tsuchida, C.A.; Nguyen, D.N.; Shy, B.R.; McGarrigle, E.R.; Sandoval Espinoza, C.R.; Carr, D.; Blaeschke, F.; Marson, A.; Doudna, J.A. Targeted delivery of CRISPR-Cas9 and transgenes enables complex immune cell engineering. Cell Rep. 2021, 35, 109207. [Google Scholar] [CrossRef] [PubMed]
- Fire, A.; Xu, S.; Montgomery, M.K.; Kostas, S.A.; Driver, S.E.; Mello, C.C. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998, 391, 806–811. [Google Scholar] [CrossRef] [PubMed]
- Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993, 75, 843–854. [Google Scholar] [CrossRef]
- Stephenson, M.L.; Zamecnik, P.C. Inhibition of Rous sarcoma viral RNA translation by a specific oligodeoxyribonucleotide. Proc. Natl. Acad. Sci. USA 1978, 75, 285–288. [Google Scholar] [CrossRef] [PubMed]
- Portnoy, V.; Lin, S.H.; Li, K.H.; Burlingame, A.; Hu, Z.H.; Li, H.; Li, L.C. saRNA-guided Ago2 targets the RITA complex to promoters to stimulate transcription. Cell Res. 2016, 26, 320–335. [Google Scholar] [CrossRef] [PubMed]
- Jiao, A.L.; Slack, F.J. RNA-mediated gene activation. Epigenetics 2014, 9, 27–36. [Google Scholar] [CrossRef]
- Li, L.C.; Okino, S.T.; Zhao, H.; Pookot, D.; Place, R.F.; Urakami, S.; Enokida, H.; Dahiya, R. Small dsRNAs induce transcriptional activation in human cells. Proc. Natl. Acad. Sci. USA 2006, 103, 17337–17342. [Google Scholar] [CrossRef] [PubMed]
- Aubert, M.; Haick, A.K.; Strongin, D.E.; Klouser, L.M.; Loprieno, M.A.; Stensland, L.; Santo, T.K.; Huang, M.L.; Hyrien, O.; Stone, D.; et al. Gene editing for latent herpes simplex virus infection reduces viral load and shedding in vivo. Nat. Commun. 2024, 15, 4018. [Google Scholar] [CrossRef]
- Aubert, M.; Strongin, D.E.; Roychoudhury, P.; Loprieno, M.A.; Haick, A.K.; Klouser, L.M.; Stensland, L.; Huang, M.L.; Makhsous, N.; Tait, A.; et al. Gene editing and elimination of latent herpes simplex virus in vivo. Nat. Commun. 2020, 11, 4148. [Google Scholar] [CrossRef]
- Takeuchi, R.; Choi, M.; Stoddard, B.L. Engineering of customized meganucleases via in vitro compartmentalization and in cellulo optimization. Methods Mol. Biol. 2015, 1239, 105–132. [Google Scholar] [CrossRef]
- Silva, G.; Poirot, L.; Galetto, R.; Smith, J.; Montoya, G.; Duchateau, P.; Pâques, F. Meganucleases and other tools for targeted genome engineering: Perspectives and challenges for gene therapy. Curr. Gene Ther. 2011, 11, 11–27. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 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
Sugiokto, F.G.; Li, R. Targeting EBV Episome for Anti-Cancer Therapy: Emerging Strategies and Challenges. Viruses 2025, 17, 110. https://doi.org/10.3390/v17010110
Sugiokto FG, Li R. Targeting EBV Episome for Anti-Cancer Therapy: Emerging Strategies and Challenges. Viruses. 2025; 17(1):110. https://doi.org/10.3390/v17010110
Chicago/Turabian StyleSugiokto, Febri Gunawan, and Renfeng Li. 2025. "Targeting EBV Episome for Anti-Cancer Therapy: Emerging Strategies and Challenges" Viruses 17, no. 1: 110. https://doi.org/10.3390/v17010110
APA StyleSugiokto, F. G., & Li, R. (2025). Targeting EBV Episome for Anti-Cancer Therapy: Emerging Strategies and Challenges. Viruses, 17(1), 110. https://doi.org/10.3390/v17010110