Skip to main content

EDITORIAL article

Front. Oncol., 13 May 2015
Sec. Cancer Molecular Targets and Therapeutics
This article is part of the Research Topic Targeting MYCN in paediatric cancers View all 9 articles

Editorial: Targeting MYCN in pediatric cancers

  • Brunel Institute of Cancer Genetics and Pharmacogenomics, College of Health and Life Sciences, Brunel University London, London, UK

MYCN is the product of a gene frequently deregulated in childhood tumors that belongs to a small but very famous family of transcription factors whose prototype member is c-MYC. The other member of the family is L-MYC, identified as a gene amplified in a subset of lung cancers. c-MYC is widely expressed in normal tissues, is the most deregulated protooncogene in human cancer, and not surprisingly, it is the subject of intensive investigations in many laboratories worldwide. Clearly, decoding its function in tumorigenesis and finding ways of inhibiting its oncogenic activity would have a very large impact in terms of human health. In contrast to the broad significance of c-MYC, the expression of MYCN is temporally and spatially restricted during embryonal development, being detected mostly in cells of the developing nervous system. This more limited function is also reflected in human pathology, with only a few types of tumors presenting alterations of MYCN. These cancers arise in the nervous system, both central and peripheral, manifesting as medulloblastomas, gliomas, and neuroblastomas. Despite the fact that MYCN-positive tumors are relatively rare, their very aggressive nature and the pediatric setting make therapeutic treatments a clinical challenge.

Increased expression of MYCN can be observed in cancers in the absence of overt aberrations of its gene structure, but about half of high-risk, metastatic neuroblastomas are characterized by amplification of the MYCN gene, leading to high mRNA and protein expression in tumor cells. Amplification of MYCN is also detected in neoplasias of the central nervous system, although at lower frequency than neuroblastoma. Importantly, it is now well established that MYCN is a direct driver of pediatric cancers: transgenic expression of MYCN in the neuroectoderm, the sympathetic tissue from which neuroblastoma originates in humans, or the cerebella that causes the development of neuroblastomas and medulloblastomas in mice (1, 2). Furthermore, other oncogenes have been shown to transform cells of the nervous system by enhancing the expression of MYCN or by stabilizing its protein product (35). The central role of MYCN in pediatric cancers renders it an ideal candidate for gene therapy, if specific inhibitors be developed. Unfortunately, many years of research from the private and academic sectors have made evident that it is extremely difficult to find small molecule inhibitors targeting transcription factors such as c-MYC and MYCN. An answer to this almost intractable problem could be the use of drugs that indirectly affected the transcriptional activity of MYCN. For example, MYC gene expression depends on the activity of the co-factor bromodomain and extra-terminal (BET) family member BRD4 that can be inhibited by a cell permeable compound called JQ1. Different research teams have demonstrated that tumors with deregulated MYCN are susceptible to JQ1 inhibition in vitro and in vivo (6, 7). The MYCN protein requires the activity of the PI3K kinase pathway for stability. A research paper in this topic shows that newly engineered PI3K inhibitors, PIK-75 and PW-12, are able to destroy the MYCN protein in mouse models of neuroblastoma and medulloblastoma, suggesting that they might be developed into useful drugs for these cancers (8). Another approach is to dissect the signaling pathways that lie downstream and upstream of MYCN, trying to illuminate the gene networks required for homeostasis of tumor cells with deregulated MYCN. Indeed, increased MYC expression is known to induce oncogenic stress that requires the balancing activity of a plethora of factors that can be exploited pharmacologically to induce “synthetic lethality.” In this regard, several papers in the research topic describe MYCN-activated genes [i.e., MDM2, SKP2, ornithine decarboxylase, ATP-binding cassette (ABC) transporters] that could be used as potential targets for therapy (912). Another interesting aspect of MYCN biology highlighted in the papers from the Perini’s and Thiele’s laboratories is that the transcription factor is not only an activator of gene expression but also a mediator of transcriptional repression (13, 14). The most recent view is that this activity might be as important as that of transcriptional activation in tumorigenesis. Several putative tumor suppressors have been identified as MYCN-repressed genes, including p75, TRKA, CASZ1, and clusterin (1518). Notably, transcriptional repression is also achieved by MYCN via epigenetic modifications, suggesting that epigenetic drugs could be used in the clinic to successfully treat MYCN-amplified tumors. In this regard, preliminary evidence in vitro and in vivo indicates that histone deacetylase inhibitors or small molecule drugs targeting the histone methyltransferase EZH2 might be useful in the context of MYCN-amplified tumors (16, 18).

Last, but not least, MYCN could serve as a tumor-associated antigen given that its expression is only marginal in postnatal tissues. Overexpression of the MYCN protein outside the embryonal context could induce a break in the immunological tolerance and expose tumor cells to the attack of the immune system. The paper by the Pistoia’s team describes the possible role of MYCN as a tumor-associated antigen and strategies to generate cytotoxic T cells directed against pediatric tumors expressing the oncogene (19). Immunological therapy with the antibody against tumor-associated disialoganglioside GD2 has shown some success in inducing remission in children with relapsed metastatic neuroblastomas (20). Other forms of immunotherapy in clinical development are the generation of chimeric antigen receptor-engineered T cells that can be directed against tumor cells expressing the GD2 antigen such as neuroblastoma, sarcoma, and melanoma. One could speculate that MYCN is an even better antigen for immunotherapy because it is almost exclusively expressed by tumor, but not normal, cells.

Despite pediatric cancers bearing activation of MYCN are extremely malignant, their dependence on this oncoprotein for proliferation and survival makes them potentially curable diseases when more efficient ways of gene targeting will be developed.

Conflict of Interest Statement

The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Swartling FJ, Grimmer MR, Hackett CS, Northcott PA, Fan QW, Goldenberg DD, et al. Pleiotropic role for MYCN in medulloblastoma. Genes Dev (2010) 24(10):1059–72. doi: 10.1101/gad.1907510

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Weiss WA, Aldape K, Mohapatra G, Feuerstein BG, Bishop JM. Targeted expression of MYCN causes neuroblastoma in transgenic mice. EMBO J (1997) 16(11):2985–95. doi:10.1093/emboj/16.11.2985

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Berry T, Luther W, Bhatnagar N, Jamin Y, Poon E, Sanda T, et al. The ALK(F1174L) mutation potentiates the oncogenic activity of MYCN in neuroblastoma. Cancer Cell (2012) 22(1):117–30. doi:10.1016/j.ccr.2012.06.001

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Molenaar JJ, Domingo-Fernandez R, Ebus ME, Lindner S, Koster J, Drabek K, et al. LIN28B induces neuroblastoma and enhances MYCN levels via let-7 suppression. Nat Genet (2012) 44(11):1199–206. doi:10.1038/ng.2436

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Northcott PA, Fernandez LA, Hagan JP, Ellison DW, Grajkowska W, Gillespie Y, et al. The miR-17/92 polycistron is up-regulated in sonic hedgehog-driven medulloblastomas and induced by N-myc in sonic hedgehog-treated cerebellar neural precursors. Cancer Res (2009) 69(8):3249–55. doi:10.1158/0008-5472.CAN-08-4710

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Bandopadhayay P, Bergthold G, Nguyen B, Schubert S, Gholamin S, Tang Y, et al. BET bromodomain inhibition of MYC-amplified medulloblastoma. Clin Cancer Res (2014) 20(4):912–25. doi:10.1158/1078-0432.CCR-13-2281

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Puissant A, Frumm SM, Alexe G, Bassil CF, Qi J, Chanthery YH, et al. Targeting MYCN in neuroblastoma by BET bromodomain inhibition. Cancer Discov (2013) 3(3):308–23. doi:10.1158/2159-8290.CD-12-0418

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Cage TA, Chanthery Y, Chesler L, Grimmer M, Knight Z, Shokat K, et al. Downregulation of MYCN through PI3K inhibition in mouse models of pediatric neural cancer. Front Oncol (2015) 5:111. doi:10.3389/fonc.2015.00111

CrossRef Full Text | Google Scholar

9. Gamble LD, Hogarty MD, Liu X, Ziegler DS, Marshall G, Norris MD, et al. Polyamine pathway inhibition as a novel therapeutic approach to treating neuroblastoma. Front Oncol (2012) 2:162. doi:10.3389/fonc.2012.00162

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Huynh T, Norris MD, Haber M, Henderson MJ. ABCC4/MRP4: a MYCN-regulated transporter and potential therapeutic target in neuroblastoma. Front Oncol (2012) 2:178. doi:10.3389/fonc.2012.00178

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Petroni M, Veschi V, Gulino A, Giannini G. Molecular mechanisms of MYCN-dependent apoptosis and the MDM2-p53 pathway: an Achille’s heel to be exploited for the therapy of MYCN-amplified neuroblastoma. Front Oncol (2012) 2:141. doi:10.3389/fonc.2012.00141

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Chen L, Tweddle DA. p53, SKP2, and DKK3 as MYCN target genes and their potential therapeutic significance. Front Oncol (2012) 2:173. doi:10.3389/fonc.2012.00173

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Gherardi S, Valli E, Erriquez D, Perini G. MYCN-mediated transcriptional repression in neuroblastoma: the other side of the coin. Front Oncol (2013) 3:42. doi:10.3389/fonc.2013.00042

PubMed Abstract | CrossRef Full Text | Google Scholar

14. He S, Liu Z, Oh DY, Thiele CJ. MYCN and the epigenome. Front Oncol (2013) 3:1. doi:10.3389/fonc.2013.00001

CrossRef Full Text | Google Scholar

15. Sala A, Bettuzzi S, Pucci S, Chayka O, Dews M, Thomas-Tikhonenko A. Regulation of CLU gene expression by oncogenes and epigenetic factors implications for tumorigenesis. Adv Cancer Res (2009) 105:115–32. doi:10.1016/S0065-230X(09)05007-6

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Wang C, Liu Z, Woo CW, Li Z, Wang L, Wei JS, et al. EZH2 Mediates epigenetic silencing of neuroblastoma suppressor genes CASZ1, CLU, RUNX3, and NGFR. Cancer Res (2012) 72(1):315–24. doi:10.1158/0008-5472.CAN-11-0961

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Iraci N, Diolaiti D, Papa A, Porro A, Valli E, Gherardi S, et al. A SP1/MIZ1/MYCN repression complex recruits HDAC1 at the TRKA and p75NTR promoters and affects neuroblastoma malignancy by inhibiting the cell response to NGF. Cancer Res (2011) 71(2):404–12. doi:10.1158/0008-5472.CAN-10-2627

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Corvetta D, Chayka O, Gherardi S, D’Acunto CW, Cantilena S, Valli E, et al. Physical interaction between MYCN oncogene and polycomb repressive complex 2 (PRC2) in neuroblastoma: functional and therapeutic implications. J Biol Chem (2013) 288(12):8332–41. doi:10.1074/jbc.M113.454280

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Pistoia V, Morandi F, Pezzolo A, Raffaghello L, Prigione I. MYCN: from oncoprotein to tumor-associated antigen. Front Oncol (2012) 2:174. doi:10.3389/fonc.2012.00174

CrossRef Full Text | Google Scholar

20. Yu AL, Gilman AL, Ozkaynak MF, London WB, Kreissman SG, Chen HX, et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N Engl J Med (2010) 363(14):1324–34. doi:10.1056/NEJMoa0911123

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: neuroblastoma, paediatric cancer, MYC, MYCN, tran

Citation: Sala A (2015) Editorial: Targeting MYCN in pediatric cancers. Front. Oncol. 4:330. doi: 10.3389/fonc.2014.00330

Received: 09 October 2014; Accepted: 31 October 2014;
Published: 13 May 2015

Edited and reviewed by: Yisong Wang, National Institute of Health, USA

Copyright: © 2015 Sala. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Arturo Sala, arturo.sala@brunel.ac.uk

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.