800
Views
4
CrossRef citations to date
0
Altmetric
Commentaries

MYCN-targeting vaccines and immunotherapeutics

&
Pages 2257-2258 | Received 04 Mar 2016, Accepted 21 Mar 2016, Published online: 10 Jun 2016

ABSTRACT

Amplification and concomitant overexpression of the MYCN oncogene is a frequent event in many malignancies including the childhood tumors, neuroblastoma and medulloblastoma. MYCN is only expressed in a defined time frame during early developmental processes,Citation1 which is beneficial for approaches combatting tumor-specific MYCN. However, MYCN is a transcription factors that was considered a poor drug target, until recent approaches suggested that down-regulation of MYCN could be possible by indirect targeting using Aurora kinase inhibitors or BET inhibitors. These concepts were proven using preclinical modelsCitation2–6 and are now entering clinical trials.

In neuroblastoma (NB), a common solid tumor of childhood, the MYCN oncogene is amplified in ˜20 % of cases.Citation7 Following induction chemotherapy and consolidation with high dose chemotherapy and stem cell rescue, maintenance treatment regimens focus on passive immunotherapy by targeting NB-specific expression of the disialoganglioside GD2 with GD2-specific monoclonal antibodies (MAbs). As GD2 is devoid of any intracellular signal transduction component, the mechanism of action of this approach is the induction of complement-dependent (CDC) and antibody-dependent cellular cytotoxicity (ADCC). In this context, treatment with mouse-human chimeric Mab ch14.18 showed superior survival rates when used as a single agent approachCitation8 or in combination with IL-2 and GM-CSF.Citation9 These two studies from independent cooperative groups demonstrated the potential of targeted immunotherapies in neuroblastoma. Ongoing clinical trials in the context of passive immunotherapy address the role of cytokines as well as the potential of novel delivery methods by long term continuous infusion. The specific components of ch14.18 were also used to design GD2-specific chimeric antigen receptor-engineered (CAR) T cells. These were generated and administered to NB patients in the context of a phase I study. Complete remission was achieved in 3/11 patients with active disease and persistence of CARs in vivo > 6 weeks was found to correlate with clinical outcome.Citation10 One disadvantage of passive immunotherapy is the absence of long lasting and persistent immunity against the malignancy. Based on the high relapse rate in NB combined with limited strategies for therapeutic intervention new approaches are urgently needed.Citation11

Globally, neuroblastomas escape from destruction by the immune system using a combinatorial strategy involving MYCN-dependent downregulation of MHC moleculesCitation12 and inhibition of NKT cells, which in turn causes up-regulation of tumor-associated macrophages (TAMs,Citation13,14 Earlier reports also indicated that MYCN-specific cytotoxic T cells (CTLs) are present in neuroblastoma patients harboring tumor-specific MYCN amplification.Citation15 Surprisingly, little is known on the usefulness of MYCN as a target for cancer immunotherapy.Citation16 Peptide vaccination using a HLA-A2 restricted peptide derived from MYCN has been shown to effectively induce a cytotoxic T cell response.Citation17 In principle, vaccination against MYCN as a tumor antigen could be an interesting strategy, especially for those patients with MYCN amplification and thus high MYCN expression.

In a recent paper, DNA vaccination has been investigated as a means to exploit high MYCN expression on tumor cells and thus to overcome MYCN immune-suppressive activities in neuroblastoma.Citation18 Here, the following improvements over previous attempts were incorporated in the study design: first, use of a “minigene” avoided transfer of a potentially harmful gene sequence to a mammalian host; second, an attenuated S. typhimurium strain (SL7207) was used as DNA delivery vehicle.Citation19 Immunocompetent mice were immunized with a MYCN minigene displaying high binding affinity to MHC class I H2-Kk by three subsequent oral applications of S. typhimurium (SL7207) carrying the MYCN constructs. Mice were then randomly assigned to one of two groups receiving either mouse NB cells with low and high MYCN expression, respectively, as syngeneic grafts. Tumor volume was significantly reduced by vaccination with a MYCN minigene displaying high affinity to MHC class I H2-Kk in comparison to vaccination with a minigene epitope displaying low MHC class I affinity. Additionally, vaccination with full-length MYCN-cDNA was also less effective in reducing tumor volume. MYCN-DNA vaccination induced a cytotoxic MYCN-specific anti-NB immune response involving IFN- and increased target cell lysis. Interestingly, absence of MYCN in tumor cells abrogated IFN- release. Importantly, no signs of autoimmunity were noted. Ex vivo, splenocytes from MYCN-vaccinated mice receiving MYCN-expressing tumor cells presented with significant higher specific cytotoxicity toward MYCN-high expressing tumor cells or SCK mammary carcinoma cells pulsed with MYCN-peptides.Citation18

Taken together, strategies to exploit MYCN as a tumor-associated antigen for immune therapy deserve further functional validation. In tumors with high MYCN expression, targeting MYCN could be useful to overcome MYCN-mediated immune suppression. While passive immune therapies in a MYCN-driven disease such as high-risk, MYCN amplified neuroblastoma, are already in clinical use, vaccination strategies have the potential to evoke long lasting effects by inducing a memory immune response. Here, strategies involving multi-peptide cancer vaccinesCitation20–22 might be an attractive route also for MYCN-based therapies. Alternatively, DNA vaccination should be further evaluated as a cost-effective and easy-to-handle option.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Funding

A.S. acknowledges the support of the Deutsche Forschungsgemeinschaft (DFG) within the Collaborative Research Center SFB 876 (http://sfb876.tu-dortmund.de), “Providing Information by Resource-Constrained Analysis”, subproject C1. The funder had no role in decision to publish or preparation of this manuscript.

References

  • Zimmerman KA, Yancopoulos GD, Collum RG, Smith RK, Kohl NE, Denis KA, Nau MM, Witte ON, Toran-Allerand D, Gee CE. Differential expression of myc family genes during murine development. In: Nature 1986; 319(6056):780-3; PMID:2419762; http://dx.doi.org/10.1038/319780a0
  • Brockmann M, Poon E, Berry T, Carstensen A, Deubzer HE, Rycak L, Jamin Y, Thway K, Robinson SP, Roels F, et al. Small molecule inhibitors of aurora-a induce proteasomal degradation of N-myc in childhood neuroblastoma. In: Cancer Cell 2013; 24(1):75-89; PMID:23792191
  • Gustafson WC, Meyerowitz JG, Nekritz EA, Chen J, Benes C, Charron E, Simonds EF, Seeger R, Matthay KK, Hertz NT, et al. Drugging MYCN through an allosteric transition in Aurora kinase A. In: Cancer Cell 2014; 26(3):414-27; PMID:25175806
  • Otto T, Horn S, Brockmann M, Eilers U, Schuttrumpf L, Popov N, Kenney AM, Schulte JH, Beijersbergen R, Christiansen H, et al. Stabilization of N-Myc is a critical function of Aurora A in human neuroblastoma. In: Cancer Cell 2009; 15(1):67-78; PMID:19111882
  • Henssen A, Thor T, Odersky A, Heukamp L, El-Hindy N, Beckers A, Speleman F, Althoff K, Schäfers S, Schramm A, et al. BET bromodomain protein inhibition is a therapeutic option for medulloblastoma. In: Oncotarget 2013; 4(11):2080-95; PMID:24231268
  • Henssen AG, Althoff K, Odersky A, Beckers A, Koche R, Speleman F, Schäfers S, Bell E, Nortmeyer M, Westermann F, et al. Targeting MYCN-driven transcription by BET-bromodomain inhibition. In: Clin Cancer Res 2015; PMID:26631615
  • Cheung Nai-Kong V, Dyer MA. Neuroblastoma: developmental biology, cancer genomics and immunotherapy. In: Nat Rev Cancer 2013; 13(6):397-411; PMID:23702928
  • Simon T, Hero B, Faldum A, Handgretinger R, Schrappe M, Klingebiel T, Berthold F. Long-term outcome of high-risk neuroblastoma patients after immunotherapy with antibody ch14.18 or oral metronomic chemotherapy. In: BMC Cancer 2011b; 11:21; http://dx.doi.org/10.1186/1471-2407-11-21
  • Yu AL, Gilman AL, Ozkaynak MF, London WB, Kreissman SG, Chen HX, Smith M, Anderson B, Villablanca JG, Matthay KK, et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. In: N Eng J Med 2010; 363(14):1324-34; http://dx.doi.org/10.1056/NEJMoa0911123
  • Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD, Rossig C, Russell HV, Diouf O, Liu E, et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. In: Blood 2011; 118(23):6050-6; PMID:21984804
  • Simon T, Berthold F, Borkhardt A, Kremens B, Carolis B de, Hero B. Treatment and outcomes of patients with relapsed, high-risk neuroblastoma: results of German trials. In: Pediatric Blood Cancer 2011a; 56(4):578-83
  • Versteeg R, van der Minne C, Plomp A, Sijts A, van Leeuwen A, Schrier P. N-myc expression switched off and class I human leukocyte antigen expression switched on after somatic cell fusion of neuroblastoma cells. In: Mol Cell Biol 1990; 10(10):5416-23; PMID:2204814; http://dx.doi.org/10.1128/MCB.10.10.5416
  • Song L, Ara T, Wu Hong-wei, Woo CW, Reynolds CP, Seeger RC, DeClerck YA, Thiele CJ, Sposto R, Metelitsa LS. Oncogene MYCN regulates localization of NKT cells to the site of disease in neuroblastoma. In: J Clin Invest 2007; 117(9):2702-12; PMID:17710228; http://dx.doi.org/10.1172/JCI30751
  • Song L, Asgharzadeh S, Salo J, Engell K, Wu Hong-wei, Sposto R, Ara T, Silverman AM, DeClerck YA, Seeger RC, et al. Valpha24-invariant NKT cells mediate antitumor activity via killing of tumor-associated macrophages. In: J Clin Invest 2009; 119(6):1524-36; PMID:19411762; http://dx.doi.org/10.1172/JCI37869
  • Sarkar AK, Nuchtern JG. Lysis of MYCN-amplified neuroblastoma cells by MYCN peptide-specific cytotoxic T lymphocytes. In: Cancer Res 2000; 60(7):1908-13.; PMID:10766179
  • Pistoia V, Morandi F, Pezzolo A, Raffaghello L; Prigione I. MYCN: from oncoprotein to tumor-associated antigen. In: Frontiers Oncol 2012; 2:174
  • Himoudi N, Yan M, Papanastasiou A, Anderson J. MYCN as a target for cancer immunotherapy. In: Cancer Immunol Immunotherapy 2008; 57(5):693-700
  • Stermann A, Huebener N, Seidel D, Fest S, Eschenburg G, Stauder M, Schramm A, Eggert A, Lode HN. Targeting of MYCN by means of DNA vaccination is effective against neuroblastoma in mice. In: Cancer Immunol Immunotherapy 2015; 64(10):1215-27; http://dx.doi.org/10.1007/s00262-015-1733-1
  • Berger E, Soldati R, Huebener N, Hohn O; Stermann A, Durmus T, Lobitz S, Zenclussen AC, Christiansen H, Lode HN, et al. Salmonella SL7207 application is the most effective DNA vaccine delivery method for successful tumor eradication in a murine model for neuroblastoma. In: Cancer Letters 2013; 331(2):167-73; PMID:23337288
  • Fenoglio D, Parodi A, Lavieri R, Kalli F, Ferrera F, Tagliamacco A, Guastalla A, Lamperti MG, Giacomini M, Filaci G. Immunogenicity of GX301 cancer vaccine: Four (telomerase peptides) are better than one. In: Hum Vaccines Immunotherapeutics 2015; 11(4):838-50
  • Feyerabend S, Stevanovic S, Gouttefangeas C, Wernet D, Hennenlotter J, Bedke J, Dietz K, Pascolo S, Kuczyk M, Rammensee HG, et al. Novel multi-peptide vaccination in Hla-A2+ hormone sensitive patients with biochemical relapse of prostate cancer. In: Prostate 2009; 69(9):917-27; PMID:19267352
  • Kirner A, Mayer-Mokler A, Reinhardt C. IMA901: a multi-peptide cancer vaccine for treatment of renal cell cancer. In: Hum Vaccines Immunotherapeutics 2014; 10(11):3179-89

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.