This article was downloaded by: [New York University] On: 06 May 2015, At: 00:57 Publisher: Taylor & Francis Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House, 37-41 Mortimer Street, London W1T 3JH, UK

OncoImmunology Publication details, including instructions for authors and subscription information: http://www.tandfonline.com/loi/koni20

The therapeutic efficacy of the oncolytic virus Delta24RGD in a murine glioma model depends primarily on antitumor immunity a

a

b

c

Anne Kleijn , Jenneke Kloezeman , Elike Treffers-Westerlaken , Giulia Fulci , Sieger a

a

b

Leenstra , Clemens Dirven , Reno Debets & Martine Lamfers

a

a

Department of Neurosurgery; Brain Tumor Center; ErasmusMC; Rotterdam, The Netherlands b

Laboratory of Experimental Tumor Immunology; Dept of Medical Oncology; ErasmusMC; Rotterdam, The Netherlands c

Click for updates

Brain Tumor Research Center; Molecular Neurosurgery Laboratory; Department of Neurosurgery; Massachusetts General Hospital; Boston, MA USA Accepted author version posted online: 29 Oct 2014.Published online: 08 Dec 2014.

To cite this article: Anne Kleijn, Jenneke Kloezeman, Elike Treffers-Westerlaken, Giulia Fulci, Sieger Leenstra, Clemens Dirven, Reno Debets & Martine Lamfers (2014) The therapeutic efficacy of the oncolytic virus Delta24-RGD in a murine glioma model depends primarily on antitumor immunity, OncoImmunology, 3:9, e955697, DOI: 10.4161/21624011.2014.955697 To link to this article: http://dx.doi.org/10.4161/21624011.2014.955697

PLEASE SCROLL DOWN FOR ARTICLE Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) contained in the publications on our platform. However, Taylor & Francis, our agents, and our licensors make no representations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the Content. Any opinions and views expressed in this publication are the opinions and views of the authors, and are not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon and should be independently verified with primary sources of information. Taylor and Francis shall not be liable for any losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoever or howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use of the Content. This article may be used for research, teaching, and private study purposes. Any substantial or systematic reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any form to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http:// www.tandfonline.com/page/terms-and-conditions

AUTHOR'S VIEW OncoImmunology 3:9, e955697; October 1, 2014; © 2014 Taylor & Francis Group, LLC

The therapeutic efficacy of the oncolytic virus Delta24-RGD in a murine glioma model depends primarily on antitumor immunity Anne Kleijn1, Jenneke Kloezeman1, Elike Treffers-Westerlaken2, Giulia Fulci3, Sieger Leenstra1, Clemens Dirven1, Reno Debets2, and Martine Lamfers1,* 1 Department of Neurosurgery; Brain Tumor Center; ErasmusMC; Rotterdam, The Netherlands; 2Laboratory of Experimental Tumor Immunology; Dept of Medical Oncology; ErasmusMC; Rotterdam, The Netherlands; 3Brain Tumor Research Center; Molecular Neurosurgery Laboratory; Department of Neurosurgery; Massachusetts General Hospital; Boston, MA USA

Downloaded by [New York University] at 00:57 06 May 2015

Keywords: adenovirus, virotherapy, immune response, glioma, antitumor immunity

Oncolytic viruses selectively lyse tumor cells, making these agents a promising treatment modality for glioma. Accumulating data suggest that the immune system plays an important role in the anti-glioma activity of oncolytic viruses. In an immune competent glioma model, the therapeutic effect of the oncolytic adenovirus Delta24-RGD was found to depend primarily on antitumor immune responses.

The oncolytic adenovirus Delta24RGD specifically replicates in tumor cells harboring a mutated retinoblastoma (Rb)-pathway, which is the case in more than 80% of human gliomas.1,2 The anti-glioma oncolytic efficacy of Delta24-RGD adenovirus has been independently demonstrated in various xenograft models.1,3,4 Clinical trials for patients with recurrent glioblastoma have been initiated.5 However, little is known about the role of the immune system in oncolytic adenovirus therapy, especially in the context of brain tumors. On one hand, the antiviral immune response could hamper oncolytic efficacy,6 whereas on the other hand, a beneficial antitumor immune response could be elicited, as has been reported for other oncolytic virus therapies.7 We hypothesized that intratumoral replication of Delta24-RGD would drive a pro-inflammatory immune response remodeling the glioma-induced immunosuppressive tumor microenvironment to one that fosters antitumor immunity.8 To study this, we employed an established immunocompetent orthotopic glioma mouse model using murine GL261

tumor cells transplanted in syngeneic C57BL/6 mice.9 In our study, we investigated the ability of Delta24-RGD oncolytic virus to infect, replicate and induce cytotoxicity in GL261 cells, allowing this model to be used to map the role of the immune response to Delta24-RGD therapy.10 Treatment resulted in approximately 50% long-term survivors, an effect that was completely abolished when mice were co-treated with the immunosuppressive agent dexamethasone. Upon intratumoral injection of Delta24-RGD a local rapid release of acute-phase cytokines, including the proinflammatory interleukins IL-1b and IL-6 was detected within 6–12 hours. Furthermore, the antiviral cytokine interferon g (IFNg) was also rapidly induced. This induction was completely inhibited by dexamethasone treatment. Apart from the cytokines, also significantly upregulated were the chemokines (C-X-C) motif 10 (CXCL10, better known as IP-10) and (C-C) motif 3 (CCL3, also known as MIP-1a), signals eliciting the recruitment of lymphocytes and monocytes, respectively. Following these signals, macrophages and CD8C T cells migrated into the tumor

microenvironment by day 1 and 14, respectively, as revealed by immunohistochemistry. Again, dexamethasone treatment had an inhibitory effect, diminishing the intratumoral influx of macrophages and CD8C T-cells. Analysis of the humoral arm of the immune system revealed the presence of adenovirus neutralizing antibodies in the blood starting 96 h after viral injection. The cellular response revealed specific GL261 cancer cell recognition by the splenocytic T cells of virus-treated mice at 14 d after treatment. This sequence of events, as summarized in Figure 1, led to the induction of a memory response, which prohibited tumor growth upon rechallenge in the long-term survivors of the Delta24-RGD treatment group. Collectively, our results indicate that the therapeutic efficacy of the oncolytic adenovirus Delta24-RGD in a murine glioma model is mainly dependent on a virusinduced antitumor immune response. Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

*Correspondence to: Martine Lamfers; Email: [email protected] Submitted: 08/07/2014; Accepted: 08/10/2014 http://dx.doi.org/10.4161/21624011.2014.955697

www.landesbioscience.com

OncoImmunology

e955697-1

Delta24-RGD T-cell

B-cell

IFNγ IL-6

memory

IP-10 T-cell

Tumor

MIP-1α

Downloaded by [New York University] at 00:57 06 May 2015

recognition

Mᴓ

6 hr

Tumor cell

96 hr

14 d

Figure 1. The antiglioma immune response during Delta24-RGD oncolytic virus therapy underlies therapeutic efficacy. The Delta24-RGD virus intratumorally injected, infects glioma cells (black) and induces cell lysis. As a result, inflammatory and immunostimulatory cytokines (IL-6, IFNg) and chemokines (IP-10, MIP-1a) are upregulated locally, leading to an intratumoral influx of macrophages (mø, purple) and CD8C T cells (blue) by 6 h and 14 d, respectively. Meanwhile, plasma cells (yellow) start producing adenoviral neutralizing antibodies. Splenocytic T cells recognize the cancer cells, leading to an antitumor immune response and long term memory .

References 1. Fueyo J, Alemany R, Gomez-Manzano C, Fuller GN, Khan A, Conrad CA, Liu TJ, Jiang H, Lemoine MG, Suzuki K, et al. Preclinical characterization of the antiglioma activity of a tropism-enhanced adenovirus targeted to the retinoblastoma pathway. J Natl Cancer Inst 2003; 95:652-60; PMID:12734316; http://dx.doi. org/10.1093/jnci/95.9.652 2. Ueki K, Ono Y, Henson JW, Efird JT, von Deimling A, Louis DN. CDKN2p16 or RB alterations occur in the majority of glioblastomas and are inversely correlated. Cancer Res 1996; 56:150-3; PMID:8548755 3. Jiang H, Gomez-Manzano C, Aoki H, Alonso MM, Kondo S, McCormick F, Xu J, Kondo Y, Bekele BN, Colman H, et al. Examination of the therapeutic potential of Delta-24-RGD in brain tumor stem cells: role of autophagic cell death. J Natl Cancer Inst 2007; 99:1410-4; PMID:17848677; http://dx.doi.org/ 10.1093/jnci/djm102

e955697-2

4. Lamfers ML, Idema S, Bosscher L, Heukelom S, Moeniralm S, van der Meulen-Muileman IH, Overmeer RM, van der Valk P, van Beusechem VW, Gerritsen WR, et al. Differential effects of combined Ad5- delta 24RGD and radiation therapy in in vitro versus in vivo models of malignant glioma. Clin Cancer Res: An Off J Am Assoc Cancer Res 2007; 13:7451-8; PMID:18094429; http:// dx.doi.org/10.1158/1078-0432.CCR-07-1265 5. https://clinicaltrials.gov/ct2/results?termDdelta24rgd& SearchDSearch 6. Alvarez-Breckenridge CA, Yu J, Price R, Wojton J, Pradarelli J, Mao H, Wei M, Wang Y, He S, Hardcastle J, et al. NK cells impede glioblastoma virotherapy through NKp30 and NKp46 natural cytotoxicity receptors. Nat Med 2012; 18:1827-34; PMID:23178246; http://dx.doi.org/10.1038/nm.3013 7. Russell SJ, Peng KW, Bell JC. Oncolytic virotherapy. Nat Biotechnol 2012; 30:658-70; PMID:22781695; http://dx.doi.org/10.1038/nbt.2287

OncoImmunology

8. Rolle CE, Sengupta S, Lesniak MS. Mechanisms of immune evasion by gliomas. Adv Exp Med Biol 2012; 746:53-76; PMID:22639159; http://dx.doi.org/ 10.1007/978-1-4614-3146-6_5 9. Szatmari T, Lumniczky K, Desaknai S, Trajcevski S, Hidvegi EJ, Hamada H, Safrany G. Detailed characterization of the mouse glioma 261 tumor model for experimental glioblastoma therapy. Cancer Sci 2006; 97:546-53; PMID:16734735; http://dx.doi.org/ 10.1111/j.1349-7006.2006.00208.x 10. Kleijn A, Kloezeman J, Treffers-Westerlaken E, Fulci G, Leenstra S, Dirven C, Debets R, Lamfers M. The in vivo therapeutic efficacy of the oncolytic adenovirus delta24-RGD is mediated by tumor-specific immunity. PloS One 2014; 9:e97495; PMID:24866126; http:// dx.doi.org/10.1371/journal.pone.0097495

Volume 3 Issue 9

The therapeutic efficacy of the oncolytic virus Delta24-RGD in a murine glioma model depends primarily on antitumor immunity.

Oncolytic viruses selectively lyse tumor cells, making these agents a promising treatment modality for glioma. Accumulating data suggest that the immu...
111KB Sizes 0 Downloads 5 Views