Showing the Way: Oncolytic Adenoviruses as Chaperones of Immunostimulatory Adjuncts
Abstract
:1. Introduction
2. OAds (Oncolytic Adenoviruses) with Immunologic Effectors
2.1. Granulocyte Macrophage Colony-Stimulating Factor (GM-CSF)
2.2. CD40L
2.3. Interferon
2.4. Interleukin-12
2.5. CTLA-4 Antibody
2.6. Combination Therapy
3. OAds and the Immune System
4. Concluding Comments
Conflicts of Interest
References
- Pitt, J.M.; Marabelle, A.; Eggermont, A.; Soria, J.C.; Kroemer, G.; Zitvogel, L. Targeting the tumor microenvironment: Removing obstruction to anticancer immune responses and immunotherapy. Ann. Oncol. 2016, 27, 1482–1492. [Google Scholar] [CrossRef] [PubMed]
- Hanahan, D.; Coussens, L.M. Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell 2012, 21, 309–322. [Google Scholar] [CrossRef] [PubMed]
- Mantovani, A.; Romero, P.; Palucka, A.K.; Marincola, F.M. Tumour immunity: Effector response to tumour and role of the microenvironment. Lancet 2008, 371, 771–783. [Google Scholar] [CrossRef]
- Loskog, A. Immunostimulatory gene therapy using oncolytic viruses as vehicles. Viruses 2015, 7, 5780–5791. [Google Scholar] [CrossRef] [PubMed]
- Albini, A.; Tosetti, F.; Benelli, R.; Noonan, D.M. Tumor inflammatory angiogenesis and its chemoprevention. Cancer Res. 2005, 65, 10637–10641. [Google Scholar] [CrossRef] [PubMed]
- Senzer, N.N.; Kaufman, H.L.; Amatruda, T.; Nemunaitis, M.; Reid, T.; Daniels, G.; Gonzalez, R.; Glaspy, J.; Whitman, E.; Harrington, K.; et al. Phase II clinical trial of a granulocyte-macrophage colony-stimulating factor-encoding, second-generation oncolytic herpesvirus in patients with unresectable metastatic melanoma. J. Clin. Oncol. 2009, 27, 5763–5771. [Google Scholar] [CrossRef] [PubMed]
- Andtbacka, R.H.; Kaufman, H.L.; Collichio, F.; Amatruda, T.; Senzer, N.; Chesney, J.; Delman, K.A.; Spitler, L.E.; Puzanov, I.; Agarwala, S.S.; et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J. Clin. Oncol. 2015, 33, 2780–2788. [Google Scholar] [CrossRef] [PubMed]
- Larson, C.; Oronsky, B.; Scicinski, J.; Fanger, G.R.; Stirn, M.; Oronsky, A.; Reid, T.R. Going viral: A review of replication-selective oncolytic adenoviruses. Oncotarget 2015, 6, 19976–19989. [Google Scholar] [CrossRef] [PubMed]
- Miura, Y.; Yamasaki, S.; Davydova, J.; Brown, E.; Aoki, K.; Vickers, S.; Yamamoto, M. Infectivity-selective oncolytic adenovirus developed by high-throughput screening of adenovirus-formatted library. Mol. Ther. 2013, 21, 139–148. [Google Scholar] [CrossRef] [PubMed]
- Yamamoto, M.; Curiel, D.T. Current issues and future directions of oncolytic adenoviruses. Mol. Ther. 2010, 18, 243–250. [Google Scholar] [CrossRef] [PubMed]
- Demir, G.; Klein, H.O.; Tuzuner, N. Low dose daily rhGM-CSF application activates monocytes and dendritic cells in vivo. Leukemia Res. 2003, 27, 1105–1108. [Google Scholar] [CrossRef]
- Kaufman, H.L.; Kim, D.W.; DeRaffele, G.; Mitcham, J.; Coffin, R.S.; Kim-Schulze, S. Local and distant immunity induced by intralesional vaccination with an oncolytic herpes virus encoding GM-CSF in patients with stage IIIc and IV melanoma. Ann. Surg. Oncol. 2010, 17, 718–730. [Google Scholar] [CrossRef] [PubMed]
- Koski, A.; Kangasniemi, L.; Escutenaire, S.; Pesonen, S.; Cerullo, V.; Diaconu, I.; Nokisalmi, P.; Raki, M.; Rajecki, M.; Guse, K.; et al. Treatment of cancer patients with a serotype 5/3 chimeric oncolytic adenovirus expressing GMCSF. Mol. Ther. 2010, 18, 1874–1884. [Google Scholar] [CrossRef] [PubMed]
- Krasnykh, V.N.; Mikheeva, G.V.; Douglas, J.T.; Curiel, D.T. Generation of recombinant adenovirus vectors with modified fibers for altering viral tropism. J. Virol. 1996, 70, 6839–6846. [Google Scholar] [PubMed]
- Fueyo, J.; Gomez-Manzano, C.; Alemany, R.; Lee, P.S.; McDonnell, T.J.; Mitlianga, P.; Shi, Y.X.; Levin, V.A.; Yung, W.K.; Kyritsis, A.P. A mutant oncolytic adenovirus targeting the rb pathway produces anti-glioma effect in vivo. Oncogene 2000, 19, 2–12. [Google Scholar] [CrossRef] [PubMed]
- Cerullo, V.; Koski, A.; Vaha-Koskela, M.; Hemminki, A. Chapter eight—Oncolytic adenoviruses for cancer immunotherapy: Data from mice, hamsters, and humans. Adv. Cancer Res. 2012, 115, 265–318. [Google Scholar] [PubMed]
- Cerullo, V.; Pesonen, S.; Diaconu, I.; Escutenaire, S.; Arstila, P.T.; Ugolini, M.; Nokisalmi, P.; Raki, M.; Laasonen, L.; Sarkioja, M.; et al. Oncolytic adenovirus coding for granulocyte macrophage colony-stimulating factor induces antitumoral immunity in cancer patients. Cancer Res. 2010, 70, 4297–4309. [Google Scholar] [CrossRef] [PubMed]
- Kanerva, A.; Nokisalmi, P.; Diaconu, I.; Koski, A.; Cerullo, V.; Liikanen, I.; Tahtinen, S.; Oksanen, M.; Heiskanen, R.; Pesonen, S.; et al. Antiviral and antitumor T-cell immunity in patients treated with GM-CSF-coding oncolytic adenovirus. Clin. Cancer Res. 2013, 19, 2734–2744. [Google Scholar] [CrossRef] [PubMed]
- Hemminki, O.; Parviainen, S.; Juhila, J.; Turkki, R.; Linder, N.; Lundin, J.; Kankainen, M.; Ristimaki, A.; Koski, A.; Liikanen, I.; et al. Immunological data from cancer patients treated with Ad5/3-E2F-Delta24-GMCSF suggests utility for tumor immunotherapy. Oncotarget 2015, 6, 4467–4481. [Google Scholar] [CrossRef] [PubMed]
- Ranki, T.; Pesonen, S.; Hemminki, A.; Partanen, K.; Kairemo, K.; Alanko, T.; Lundin, J.; Linder, N.; Turkki, R.; Ristimaki, A.; et al. Phase I study with ONCOS-102 for the treatment of solid tumors—An evaluation of clinical response and exploratory analyses of immune markers. J. Immunother. Cancer 2016, 4. [Google Scholar] [CrossRef] [PubMed]
- Ramesh, N.; Ge, Y.; Ennist, D.L.; Zhu, M.; Mina, M.; Ganesh, S.; Reddy, P.S.; Yu, D.C. CG0070, a conditionally replicating granulocyte macrophage colony-stimulating factor—Armed oncolytic adenovirus for the treatment of bladder cancer. Clin. Cancer Res. 2006, 12, 305–313. [Google Scholar] [CrossRef] [PubMed]
- Flemington, E.K.; Speck, S.H.; Kaelin, W.G. E2F-1-mediated transactivation is inhibited by complex-formation with the retinoblastoma susceptibility gene-product. Proc. Natl. Acad. Sci. USA 1993, 90, 6914–6918. [Google Scholar] [CrossRef] [PubMed]
- Neuman, E.; Flemington, E.K.; Sellers, W.R.; Kaelin, W.G. Transcription of the E2F-1 gene is rendered cell-cycle dependent by E2F DNA-binding sites within its promoter. Mol. Cell. Biol. 1994, 14, 6607–6615. [Google Scholar] [CrossRef] [PubMed]
- Burke, J.M.; Lamm, D.L.; Meng, M.V.; Nemunaitis, J.J.; Stephenson, J.J.; Arseneau, J.C.; Aimi, J.; Lerner, S.; Yeung, A.W.; Kazarian, T.; et al. A first in human phase 1 study of CG0070, a GM-CSF expressing oncolytic adenovirus, for the treatment of nonmuscle invasive bladder cancer. J. Urol. 2012, 188, 2391–2397. [Google Scholar] [CrossRef] [PubMed]
- Tong, A.W.; Stone, M.J. Prospects for CD40-directed experimental therapy of human cancer. Cancer Gene Ther. 2003, 10, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Tong, A.W.; Papayoti, M.H.; Netto, G.; Armstrong, D.T.; Ordonez, G.; Lawson, J.M.; Stone, M.J. Growth-inhibitory effects of CD40 ligand (CD154) and its endogenous expression in human breast cancer. Clin. Cancer Res. 2001, 7, 691–703. [Google Scholar] [PubMed]
- Ghamande, S.; Hylander, B.L.; Oflazoglu, E.; Lele, S.; Fanslow, W.; Repasky, E.A. Recombinant CD40 ligand therapy has significant antitumor effects on CD40-positive ovarian tumor xenografts grown in SCID mice and demonstrates an augmented effect with cisplatin. Cancer Res. 2001, 61, 7556–7562. [Google Scholar] [PubMed]
- Gomes, E.M.; Rodrigues, M.S.; Phadke, A.P.; Butcher, L.D.; Starling, C.; Chen, S.; Chang, D.; Hernandez-Alcoceba, R.; Newman, J.T.; Stone, M.J.; et al. Antitumor activity of an oncolytic adenoviral-CD40 ligand (CD154) transgene construct in human breast cancer cells. Clin. Cancer Res. 2009, 15, 1317–1325. [Google Scholar] [CrossRef] [PubMed]
- Fernandes, M.S.; Gomes, E.M.; Butcher, L.D.; Hernandez-Alcoceba, R.; Chang, D.; Kansopon, J.; Newman, J.; Stone, M.J.; Tong, A.W. Growth inhibition of human multiple myeloma cells by an oncolytic adenovirus carrying the CD40 ligand transgene. Clin. Cancer Res. 2009, 15, 4847–4856. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.F.; Xue, S.Y.; Lu, Z.Z.; Xiao, F.J.; Yin, Y.; Zhang, Q.W.; Wu, C.T.; Wang, H.; Wang, L.S. Antitumor effects of oncolytic adenovirus armed with PSA-IZ-CD40l fusion gene against prostate cancer. Gene Ther. 2014, 21, 723–731. [Google Scholar] [CrossRef] [PubMed]
- Diaconu, I.; Cerullo, V.; Hirvinen, M.L.M.; Escutenaire, S.; Ugolini, M.; Pesonen, S.K.; Bramante, S.; Parviainen, S.; Kanerva, A.; Loskog, A.S.I.; et al. Immune response is an important aspect of the antitumor effect produced by a CD40l-encoding oncolytic adenovirus. Cancer Res. 2012, 72, 2327–2338. [Google Scholar] [CrossRef] [PubMed]
- Kawashima, T.; Kagawa, S.; Kobayashi, N.; Shirakiya, Y.; Umeoka, T.; Teraishi, F.; Taki, M.; Kyo, S.; Tanaka, N.; Fujiwara, T. Telomerase-specific replication-selective virotherapy for human cancer. Clin. Cancer Res. 2004, 10, 285–292. [Google Scholar] [CrossRef] [PubMed]
- Wirth, T.; Zender, L.; Schulte, B.; Mundt, B.; Plentz, R.; Rudolph, K.L.; Manns, M.; Kubicka, S.; Kuhnel, F. A telomerase-dependent conditionally replicating adenovirus for selective treatment of cancer. Cancer Res. 2003, 63, 3181–3188. [Google Scholar] [PubMed]
- Lanson, N.A., Jr.; Friedlander, P.L.; Schwarzenberger, P.; Kolls, J.K.; Wang, G. Replication of an adenoviral vector controlled by the human telomerase reverse transcriptase promoter causes tumor-selective tumor lysis. Cancer Res. 2003, 63, 7936–7941. [Google Scholar] [PubMed]
- Pesonen, S.; Diaconu, I.; Kangasniemi, L.; Ranki, T.; Kanerva, A.; Pesonen, S.K.; Gerdemann, U.; Leen, A.M.; Kairemo, K.; Oksanen, M.; et al. Oncolytic immunotherapy of advanced solid tumors with a CD40l-expressing replicating adenovirus: Assessment of safety and immunologic responses in patients. Cancer Res. 2012, 72, 1621–1631. [Google Scholar] [CrossRef] [PubMed]
- Pestka, S. Purification and cloning of interferon alpha. Curr. Top. Microbiol. Immunol. 2007, 316, 23–37. [Google Scholar] [PubMed]
- Decatris, M.; Santhanam, S.; O’Byrne, K. Potential of interferon-alpha in solid tumours: Part 1. BioDrugs 2002, 16, 261–281. [Google Scholar] [CrossRef] [PubMed]
- Ferrantini, M.; Capone, I.; Belardelli, F. Interferon-alpha and cancer: Mechanisms of action and new perspectives of clinical use. Biochimie 2007, 89, 884–893. [Google Scholar] [CrossRef] [PubMed]
- Koeller, J.M. Biologic response modifiers: The interferon alfa experience. Am. J. Hosp. Pharm. 1989, 46, S11–S15. [Google Scholar] [PubMed]
- Nukui, Y.; Picozzi, V.J.; Traverso, L.W. Interferon-based adjuvant chemoradiation therapy improves survival after pancreaticoduodenectomy for pancreatic adenocarcinoma. Am. J. Surg. 2000, 179, 367–371. [Google Scholar] [CrossRef]
- Rocha, F.G.; Hashimoto, Y.; Traverso, L.W.; Dorer, R.; Kozarek, R.; Helton, W.S.; Picozzi, V.J. Interferon-based adjuvant chemoradiation for resected pancreatic head cancer: Long-term follow-up of the virginia mason protocol. Ann. Surg. 2016, 263, 376–384. [Google Scholar] [CrossRef] [PubMed]
- Jensen, E.H.; Armstrong, L.; Lee, C.; Tuttle, T.M.; Vickers, S.M.; Sielaff, T.; Greeno, E.W. Neoadjuvant interferon-based chemoradiation for borderline resectable and locally advanced pancreas cancer: A phase II pilot study. HPB 2014, 16, 131–139. [Google Scholar] [CrossRef] [PubMed]
- Hatanaka, K.; Suzuki, K.; Miura, Y.; Yoshida, K.; Ohnami, S.; Kitade, Y.; Yoshida, T.; Aoki, K. Interferon-α and antisense K-ras RNA combination gene therapy against pancreatic cancer. J. Gene Med. 2004, 6, 1139–1148. [Google Scholar] [CrossRef] [PubMed]
- Ohashi, M.; Yoshida, K.; Kushida, M.; Miura, Y.; Ohnami, S.; Ikarashi, Y.; Kitade, Y.; Yoshida, T.; Aoki, K. Adenovirus-mediated interferon alpha gene transfer induces regional direct cytotoxicity and possible systemic immunity against pancreatic cancer. Br. J. Cancer 2005, 93, 441–449. [Google Scholar] [CrossRef] [PubMed]
- Hara, H.; Kobayashi, A.; Yoshida, K.; Ohashi, M.; Ohnami, S.; Uchida, E.; Higashihara, E.; Yoshida, T.; Aoki, K. Local interferon-alpha gene therapy elicits systemic immunity in a syngeneic pancreatic cancer model in hamster. Cancer Sci. 2007, 98, 455–463. [Google Scholar] [CrossRef] [PubMed]
- Armstrong, L.; Arrington, A.; Han, J.; Gavrikova, T.; Brown, E.; Yamamoto, M.; Vickers, S.M.; Davydova, J. Generation of a novel, cyclooxygenase-2-targeted, interferon-expressing, conditionally replicative adenovirus for pancreatic cancer therapy. Am. J. Surg. 2012, 204, 741–750. [Google Scholar] [CrossRef] [PubMed]
- Armstrong, L.; Davydova, J.; Brown, E.; Han, J.; Yamamoto, M.; Vickers, S.M. Delivery of interferon alpha using a novel cox2-controlled adenovirus for pancreatic cancer therapy. Surgery 2012, 152, 114–122. [Google Scholar] [CrossRef] [PubMed]
- LaRocca, C.J.; Han, J.; Gavrikova, T.; Armstrong, L.; Oliveira, A.R.; Shanley, R.; Vickers, S.M.; Yamamoto, M.; Davydova, J. Oncolytic adenovirus expressing interferon alpha in a syngeneic syrian hamster model for the treatment of pancreatic cancer. Surgery 2015, 157, 888–898. [Google Scholar] [CrossRef] [PubMed]
- Benedict, W.F.; Tao, Z.; Kim, C.S.; Zhang, X.; Zhou, J.H.; Adam, L.; McConkey, D.J.; Papageorgiou, A.; Munsell, M.; Philopena, J.; et al. Intravesical Ad-IFNα causes marked regression of human bladder cancer growing orthotopically in nude mice and overcomes resistance to IFN-α protein. Mol. Ther. 2004, 10, 525–532. [Google Scholar] [CrossRef] [PubMed]
- Dinney, C.P.; Fisher, M.B.; Navai, N.; O’Donnell, M.A.; Cutler, D.; Abraham, A.; Young, S.; Hutchins, B.; Caceres, M.; Kishnani, N.; et al. Phase I trial of intravesical recombinant adenovirus mediated interferon-α2b formulated in Syn3 for Bacillus Calmette-Guerin failures in nonmuscle invasive bladder cancer. J. Urol. 2013, 190, 850–856. [Google Scholar] [CrossRef] [PubMed]
- Tsugawa, T.; Kuwashima, N.; Sato, H.; Fellows-Mayle, W.K.; Dusak, J.E.; Okada, K.; Papworth, G.D.; Watkins, S.C.; Gambotto, A.; Yoshida, J.; et al. Sequential delivery of interferon-α gene and DCs to intracranial gliomas promotes an effective antitumor response. Gene Ther. 2004, 11, 1551–1558. [Google Scholar] [CrossRef] [PubMed]
- Shashkova, E.V.; Spencer, J.F.; Wold, W.S.; Doronin, K. Targeting interferon-α increases antitumor efficacy and reduces hepatotoxicity of E1A-mutated spread-enhanced oncolytic adenovirus. Mol. Ther. 2007, 15, 598–607. [Google Scholar] [CrossRef] [PubMed]
- Brin, E.; Atencio, I.; Helmich, B.K.; Maneval, D.; Laface, D. Adenovirus delivery provides extended interferon-α exposure and augments treatment of metastatic carcinoma. Cancer Gene Ther. 2006, 13, 664–675. [Google Scholar] [CrossRef] [PubMed]
- Ahmed, C.M.I.; Johnson, D.E.; Demers, G.W.; Engler, H.; Howe, J.A.; Wills, K.N.; Wen, S.F.; Shinoda, J.; Beltran, J.; Nodelman, M.; et al. Interferon α2b gene delivery using adenoviral vector causes inhibition of tumor growth in xenograft models from a variety of cancers. Cancer Gene Ther. 2001, 8, 788–795. [Google Scholar] [CrossRef] [PubMed]
- Atkins, M.B.; Robertson, M.J.; Gordon, M.; Lotze, M.T.; DeCoste, M.; DuBois, J.S.; Ritz, J.; Sandler, A.B.; Edington, H.D.; Garzone, P.D.; et al. Phase I evaluation of intravenous recombinant human interleukin 12 in patients with advanced malignancies. Clin. Cancer Res. 1997, 3, 409–417. [Google Scholar] [PubMed]
- Zaidi, M.R.; Merlino, G. The two faces of interferon-γ in cancer. Clin. Cancer Res. 2011, 17, 6118–6124. [Google Scholar] [CrossRef] [PubMed]
- Del Vecchio, M.; Bajetta, E.; Canova, S.; Lotze, M.T.; Wesa, A.; Parmiani, G.; Anichini, A. Interleukin-12: Biological properties and clinical application. Clin. Cancer Res. 2007, 13, 4677–4685. [Google Scholar] [CrossRef] [PubMed]
- Freytag, S.O.; Barton, K.N.; Zhang, Y. Efficacy of oncolytic adenovirus expressing suicide genes and interleukin-12 in preclinical model of prostate cancer. Gene Ther. 2013, 20, 1131–1139. [Google Scholar] [CrossRef] [PubMed]
- Freytag, S.O.; Rogulski, K.R.; Paielli, D.L.; Gilbert, J.D.; Kim, J.H. A novel three-pronged approach to kill cancer cells selectively: Concomitant viral, double suicide gene, and radiotherapy. Hum. Gene Ther. 1998, 9, 1323–1333. [Google Scholar] [CrossRef] [PubMed]
- Choi, I.K.; Lee, J.S.; Zhang, S.N.; Park, J.; Sonn, C.H.; Lee, K.M.; Yun, C.O. Oncolytic adenovirus co-expressing IL-12 and IL-18 improves tumor-specific immunity via differentiation of T cells expressing IL-12Rβ2 or IL-18Rα. Gene Ther. 2011, 18, 898–909. [Google Scholar] [CrossRef] [PubMed]
- Choi, K.J.; Zhang, S.N.; Choi, I.K.; Kim, J.S.; Yun, C.O. Strengthening of antitumor immune memory and prevention of thymic atrophy mediated by adenovirus expressing IL-12 and GM-CSF. Gene Ther. 2012, 19, 711–723. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.S.; Kim, J.H.; Choi, K.J.; Choi, I.K.; Kim, H.; Cho, S.; Cho, B.C.; Yun, C.O. Enhanced antitumor effect of oncolytic adenovirus expressing interleukin-12 and B7-1 in an immunocompetent murine model. Clin. Cancer Res. 2006, 12, 5859–5868. [Google Scholar] [CrossRef] [PubMed]
- Bortolanza, S.; Bunuales, M.; Otano, I.; Gonzalez-Aseguinolaza, G.; Ortiz-de-Solorzano, C.; Perez, D.; Prieto, J.; Hernandez-Alcoceba, R. Treatment of pancreatic cancer with an oncolytic adenovirus expressing interleukin-12 in syrian hamsters. Mol. Ther. 2009, 17, 614–622. [Google Scholar] [CrossRef] [PubMed]
- NCCN. NCCN Clinical Practice Guidelines in Oncology: Melanoma; NCCN: Fort Washington, PA, USA, 2016. [Google Scholar]
- Contardi, E.; Palmisano, G.L.; Tazzari, P.L.; Martelli, A.M.; Fala, F.; Fabbi, M.; Kato, T.; Lucarelli, E.; Donati, D.; Polito, L.; et al. CTLA-4 is constitutively expressed on tumor cells and can trigger apoptosis upon ligand interaction. Int. J. Cancer 2005, 117, 538–550. [Google Scholar] [CrossRef] [PubMed]
- Dias, J.D.; Hemminki, O.; Diaconu, I.; Hirvinen, M.; Bonetti, A.; Guse, K.; Escutenaire, S.; Kanerva, A.; Pesonen, S.; Loskog, A.; et al. Targeted cancer immunotherapy with oncolytic adenovirus coding for a fully human monoclonal antibody specific for CTLA-4. Gene Ther. 2012, 19, 988–998. [Google Scholar] [CrossRef] [PubMed]
- Zou, W.; Wolchok, J.D.; Chen, L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci. Transl. Med. 2016, 8. [Google Scholar] [CrossRef] [PubMed]
- Rajani, K.; Parrish, C.; Kottke, T.; Thompson, J.; Zaidi, S.; Ilett, L.; Shim, K.G.; Diaz, R.M.; Pandha, H.; Harrington, K.; et al. Combination therapy with reovirus and anti-PD-1 blockade controls tumor growth through innate and adaptive immune responses. Mol. Ther. 2016, 24, 166–174. [Google Scholar] [CrossRef] [PubMed]
- Engeland, C.E.; Grossardt, C.; Veinalde, R.; Bossow, S.; Lutz, D.; Kaufmann, J.K.; Shevchenko, I.; Umansky, V.; Nettelbeck, D.M.; Weichert, W.; et al. CTLA-4 and PD-l1 checkpoint blockade enhances oncolytic measles virus therapy. Mol. Ther. 2014, 22, 1949–1959. [Google Scholar] [CrossRef] [PubMed]
- Woller, N.; Gurlevik, E.; Fleischmann-Mundt, B.; Schumacher, A.; Knocke, S.; Kloos, A.M.; Saborowski, M.; Geffers, R.; Manns, M.P.; Wirth, T.C.; et al. Viral infection of tumors overcomes resistance to PD-1-immunotherapy by broadening neoantigenome-directed T-cell responses. Mol. Ther. 2015, 23, 1630–1640. [Google Scholar] [CrossRef] [PubMed]
- Nishio, N.; Diaconu, I.; Liu, H.; Cerullo, V.; Caruana, I.; Hoyos, V.; Bouchier-Hayes, L.; Savoldo, B.; Dotti, G. Armed oncolytic virus enhances immune functions of chimeric antigen receptor-modified T cells in solid tumors. Cancer Res. 2014, 74, 5195–5205. [Google Scholar] [CrossRef] [PubMed]
- Zhu, J.G.; Huang, X.P.; Yang, Y.P. Innate immune response to adenoviral vectors is mediated by both toll-like receptor-dependent and -independent pathways. J. Virol. 2007, 81, 3170–3180. [Google Scholar] [CrossRef] [PubMed]
- Thaci, B.; Ulasov, I.V.; Wainwright, D.A.; Lesniak, M.S. The challenge for gene therapy: Innate immune response to adenoviruses. Oncotarget 2011, 2, 113–121. [Google Scholar] [CrossRef] [PubMed]
- Chen, M.; Barnfield, C.; Naslund, T.I.; Fleeton, M.N.; Liljestrom, P. MyD88 expression is required for efficient cross-presentation of viral antigens from infected cells. J. Virol. 2005, 79, 2964–2972. [Google Scholar] [CrossRef] [PubMed]
- Uusi-Kerttula, H.; Hulin-Curtis, S.; Davies, J.; Parker, A.L. Oncolytic adenovirus: Strategies and insights for vector design and immuno-oncolytic applications. Viruses 2015, 7, 6009–6042. [Google Scholar] [CrossRef] [PubMed]
- Seregin, S.S.; Amalfitano, A. Improving adenovirus based gene transfer: Strategies to accomplish immune evasion. Viruses 2010, 2, 2013–2036. [Google Scholar] [CrossRef] [PubMed]
- Thomas, M.A.; Spencer, J.F.; Toth, K.; Sagartz, J.E.; Phillips, N.J.; Wold, W.S. Immunosuppression enhances oncolytic adenovirus replication and antitumor efficacy in the syrian hamster model. Mol. Ther. 2008, 16, 1665–1673. [Google Scholar] [CrossRef] [PubMed]
- Dhar, D.; Spencer, J.F.; Toth, K.; Wold, W.S. Effect of preexisting immunity on oncolytic adenovirus vector INGN 007 antitumor efficacy in immunocompetent and immunosuppressed Syrian hamsters. J. Virol. 2009, 83, 2130–2139. [Google Scholar] [CrossRef] [PubMed]
- Young, B.A.; Spencer, J.F.; Ying, B.; Tollefson, A.E.; Toth, K.; Wold, W.S. The role of cyclophosphamide in enhancing antitumor efficacy of an adenovirus oncolytic vector in subcutaneous Syrian hamster tumors. Cancer Gene Ther. 2013, 20, 521–530. [Google Scholar] [CrossRef] [PubMed]
- Rhee, E.G.; Blattman, J.N.; Kasturi, S.P.; Kelley, R.P.; Kaufman, D.R.; Lynch, D.M.; La Porte, A.; Simmons, N.L.; Clark, S.L.; Pulendran, B.; et al. Multiple innate immune pathways contribute to the immunogenicity of recombinant adenovirus vaccine vectors. J. Virol. 2011, 85, 315–323. [Google Scholar] [CrossRef] [PubMed]
- Salio, M.; Cerundolo, V. Viral immunity: Cross-priming with the help of TLR3. Curr. Biol. 2005, 15, R336–R339. [Google Scholar] [CrossRef] [PubMed]
- Edukulla, R.; Woller, N.; Mundt, B.; Knocke, S.; Gurlevik, E.; Saborowski, M.; Malek, N.; Manns, M.P.; Wirth, T.; Kuhnel, F.; et al. Antitumoral immune response by recruitment and expansion of dendritic cells in tumors infected with telomerase-dependent oncolytic viruses. Cancer Res. 2009, 69, 1448–1458. [Google Scholar] [PubMed]
- Gurlevik, E.; Woller, N.; Struver, N.; Schache, P.; Kloos, A.; Manns, M.P.; Zender, L.; Kuhnel, F.; Kubicka, S. Selectivity of oncolytic viral replication prevents antiviral immune response and toxicity, but does not improve antitumoral immunity. Mol. Ther. 2010, 18, 1972–1982. [Google Scholar] [CrossRef] [PubMed]
© 2016 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 ( http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Huang, J.L.; LaRocca, C.J.; Yamamoto, M. Showing the Way: Oncolytic Adenoviruses as Chaperones of Immunostimulatory Adjuncts. Biomedicines 2016, 4, 23. https://doi.org/10.3390/biomedicines4030023
Huang JL, LaRocca CJ, Yamamoto M. Showing the Way: Oncolytic Adenoviruses as Chaperones of Immunostimulatory Adjuncts. Biomedicines. 2016; 4(3):23. https://doi.org/10.3390/biomedicines4030023
Chicago/Turabian StyleHuang, Jing Li, Christopher J. LaRocca, and Masato Yamamoto. 2016. "Showing the Way: Oncolytic Adenoviruses as Chaperones of Immunostimulatory Adjuncts" Biomedicines 4, no. 3: 23. https://doi.org/10.3390/biomedicines4030023
APA StyleHuang, J. L., LaRocca, C. J., & Yamamoto, M. (2016). Showing the Way: Oncolytic Adenoviruses as Chaperones of Immunostimulatory Adjuncts. Biomedicines, 4(3), 23. https://doi.org/10.3390/biomedicines4030023