High Frequency of p53/MDM2/p14ARF Pathway Abnormalities in Relapsed Neuroblastoma

Jane Carr-Wilkinson, Kieran O'Toole, Katrina M Wood, Christine C Challen, Angela G Baker, Julian R Board, Laura Evans, Michael Cole, Nai-Kong V Cheung, Joachim Boos, Gabriele Köhler, Ivo Leuschner, Andrew D J Pearson, John Lunec, Deborah A Tweddle, Jane Carr-Wilkinson, Kieran O'Toole, Katrina M Wood, Christine C Challen, Angela G Baker, Julian R Board, Laura Evans, Michael Cole, Nai-Kong V Cheung, Joachim Boos, Gabriele Köhler, Ivo Leuschner, Andrew D J Pearson, John Lunec, Deborah A Tweddle

Abstract

Purpose: Most neuroblastomas initially respond to therapy but many relapse with chemoresistant disease. p53 mutations are rare in diagnostic neuroblastomas, but we have previously reported inactivation of the p53/MDM2/p14(ARF) pathway in 9 of 17 (53%) neuroblastoma cell lines established at relapse.

Hypothesis: Inactivation of the p53/MDM2/p14(ARF) pathway develops during treatment and contributes to neuroblastoma relapse.

Methods: Eighty-four neuroblastomas were studied from 41 patients with relapsed neuroblastoma including 38 paired neuroblastomas at different stages of therapy. p53 mutations were detected by automated sequencing, p14(ARF) methylation and deletion by methylation-specific PCR and duplex PCR, respectively, and MDM2 amplification by fluorescent in situ hybridization.

Results: Abnormalities in the p53 pathway were identified in 20 of 41 (49%) cases. Downstream defects due to inactivating missense p53 mutations were identified in 6 of 41 (15%) cases, 5 following chemotherapy and/or at relapse and 1 at diagnosis, postchemotherapy, and relapse. The presence of a p53 mutation was independently prognostic for overall survival (hazard ratio, 3.4; 95% confidence interval, 1.2-9.9; P = 0.02). Upstream defects were present in 35% of cases: MDM2 amplification in 3 cases, all at diagnosis and relapse and p14(ARF) inactivation in 12 of 41 (29%) cases: 3 had p14(ARF) methylation, 2 after chemotherapy, and 9 had homozygous deletions, 8 at diagnosis and relapse.

Conclusions: These results show that a high proportion of neuroblastomas which relapse have an abnormality in the p53 pathway. The majority have upstream defects suggesting that agents which reactivate wild-type p53 would be beneficial, in contrast to those with downstream defects in which p53-independent therapies are indicated.

Figures

Figure 1. p53 status and function in…
Figure 1. p53 status and function in neuroblastoma at different stages of treatment
(a) Case 14, (ai) codon 203 mutation (aii) DHPLC chromatogram. (b-d) Case 1 (bi) codon 270 mutation present post-chemotherapy and relapse but not at diagnosis. (bii) DHPLC chromatogram (c) p53 and p21 immunohistochemistry showing lack of p21 expression at relapse (scale bar = 50μm) (d) Codon 270 mutation-specific PCR.
Figure 1. p53 status and function in…
Figure 1. p53 status and function in neuroblastoma at different stages of treatment
(a) Case 14, (ai) codon 203 mutation (aii) DHPLC chromatogram. (b-d) Case 1 (bi) codon 270 mutation present post-chemotherapy and relapse but not at diagnosis. (bii) DHPLC chromatogram (c) p53 and p21 immunohistochemistry showing lack of p21 expression at relapse (scale bar = 50μm) (d) Codon 270 mutation-specific PCR.
Figure 2. MDM2 amplification in neuroblastoma
Figure 2. MDM2 amplification in neuroblastoma
(a & b) Case 4 (ai) MDM2 amplification by FISH on a relapsed tumour imprint (×100), (aii) Non-MDM2 amplified tumour. (b) 50K SNP arrays log2ratio from relapsed tumour DNA showing (bi) MDM2 amplification (12q15) (bii) no MYCN amplification (2p24). (c) Case 19, MDM2 amplification by FISH on paraffin sections from (ci) diagnostic tumour and (cii) relapse.
Figure 3. p14 ARF abnormalities in neuroblastoma
Figure 3. p14ARF abnormalities in neuroblastoma
(a & b) Case 9 (a) MSP showing unmethylated p14ARF (diagnosis) and partial methylation (progression) (b) p14ARF mRNA expression showing decreased expression at progression. (c) Duplex PCR showing (ci) homozygous deletion at diagnosis and relapse (case 20) and other relapsed tumours, (cii) the presence of p14ARF in constitutional DNA from cases homozygously deleted for p14ARF.

References

    1. Keshelava N, Zuo JJ, Chen P, et al. Loss of p53 function confers high-level multidrug resistance in neuroblastoma cell lines. Cancer Res. 2001;61:6185–93.
    1. Lane DP. Cancer. p53, guardian of the genome. Nature. 1992;358:15–6.
    1. Michalak E, Villunger A, Erlacher M, Strasser A. Death squads enlisted by the tumour suppressor p53. Biochem Biophys Res Commun. 2005;331:786–98.
    1. Barak Y, Juven T, Haffner R, Oren M. mdm2 expression is induced by wild type p53 activity. The EMBO journal. 1993;12:461–8.
    1. Stone S, Jiang P, Dayananth P, et al. Complex structure and regulation of the P16 (MTS1) locus. Cancer Res. 1995;55:2988–94.
    1. Honda R, Yasuda H. Association of p19(ARF) with Mdm2 inhibits ubiquitin ligase activity of Mdm2 for tumor suppressor p53. The EMBO journal. 1999;18:22–7.
    1. Gallagher SJ, Kefford RF, Rizos H. The ARF tumour suppressor. The international journal of biochemistry & cell biology. 2006;38:1637–41.
    1. Robertson KD, Jones PA. The human ARF cell cycle regulatory gene promoter is a CpG island which can be silenced by DNA methylation and down-regulated by wild-type p53. Molecular and cellular biology. 1998;18:6457–73.
    1. Tweddle DA, Pearson AD, Haber M, et al. The p53 pathway and its inactivation in neuroblastoma. Cancer letters. 2003;197:93–8.
    1. Tweddle DA, Malcolm AJ, Bown N, Pearson AD, Lunec J. Evidence for the development of p53 mutations after cytotoxic therapy in a neuroblastoma cell line. Cancer Res. 2001;61:8–13.
    1. Carr J, Bell E, Pearson AD, et al. Increased frequency of aberrations in the p53/MDM2/p14(ARF) pathway in neuroblastoma cell lines established at relapse. Cancer Res. 2006;66:2138–45.
    1. Brodeur GM, Pritchard J, Berthold F, et al. Revisions of the international criteria for neuroblastoma diagnosis, staging, and response to treatment. J Clin Oncol. 1993;11:1466–77.
    1. Ambros IM, Benard J, Boavida M, et al. Quality assessment of genetic markers used for therapy stratification. J Clin Oncol. 2003;21:2077–84.
    1. Lamont JM, McManamy CS, Pearson AD, Clifford SC, Ellison DW. Combined histopathological and molecular cytogenetic stratification of medulloblastoma patients. Clin Cancer Res. 2004;10:5482–93.
    1. Carr J, Bown NP, Case MC, Hall AG, Lunec J, Tweddle DA. High-resolution analysis of allelic imbalance in neuroblastoma cell lines by single nucleotide polymorphism arrays. Cancer genetics and cytogenetics. 2007;172:127–38.
    1. Chen L, Malcolm AJ, Wood KM, et al. Cell cycle. Vol. 6. Georgetown, Tex: 2007. p53 is nuclear and functional in both undifferentiated and differentiated neuroblastoma; pp. 2685–96.
    1. Hosoi G, Hara J, Okamura T, et al. Low frequency of the p53 gene mutations in neuroblastoma. Cancer. 1994;73:3087–93.
    1. Imamura J, Bartram CR, Berthold F, Harms D, Nakamura H, Koeffler HP. Mutation of the p53 gene in neuroblastoma and its relationship with N-myc amplification. Cancer Res. 1993;53:4053–8.
    1. Manhani R, Cristofani LM, Odone Filho V, Bendit I. Concomitant p53 mutation and MYCN amplification in neuroblastoma. Medical and pediatric oncology. 1997;29:206–7.
    1. Omura-Minamisawa M, Diccianni MB, Chang RC, et al. p16/p14(ARF) cell cycle regulatory pathways in primary neuroblastoma: p16 expression is associated with advanced stage disease. Clin Cancer Res. 2001;7:3481–90.
    1. Rainwater R, Parks D, Anderson ME, Tegtmeyer P, Mann K. Role of cysteine residues in regulation of p53 function. Molecular and cellular biology. 1995;15:3892–903.
    1. Butler JS, Loh SN. Structure, function, and aggregation of the zinc-free form of the p53 DNA binding domain. Biochemistry. 2003;42:2396–403.
    1. Sidransky D, Mikkelsen T, Schwechheimer K, Rosenblum ML, Cavanee W, Vogelstein B. Clonal expansion of p53 mutant cells is associated with brain tumour progression. Nature. 1992;355:846–7.
    1. Ishii N, Tada M, Hamou MF, et al. Cells with TP53 mutations in low grade astrocytic tumors evolve clonally to malignancy and are an unfavorable prognostic factor. Oncogene. 1999;18:5870–8.
    1. Pollock RE, Lang A, Luo J, El-Naggar AK, Yu D. Soft tissue sarcoma metastasis from clonal expansion of p53 mutated tumor cells. Oncogene. 1996;12:2035–9.
    1. Wada H, Asada M, Nakazawa S, et al. Clonal expansion of p53 mutant cells in leukemia progression in vitro. Leukemia. 1994;8:53–9.
    1. Righetti SC, Perego P, Corna E, Pierotti MA, Zunino F. Emergence of p53 mutant cisplatin-resistant ovarian carcinoma cells following drug exposure: spontaneously mutant selection. Cell Growth Differ. 1999;10:473–8.
    1. de Vries A, Flores ER, Miranda B, et al. Targeted point mutations of p53 lead to dominant-negative inhibition of wild-type p53 function. Proceedings of the National Academy of Sciences of the United States of America. 2002;99:2948–53.
    1. Wijnhoven SW, Speksnijder EN, Liu X, et al. Dominant-negative but not gain-of-function effects of a p53.R270H mutation in mouse epithelium tissue after DNA damage. Cancer Res. 2007;67:4648–56.
    1. Xue C, Haber M, Flemming C, et al. p53 determines multidrug sensitivity of childhood neuroblastoma. Cancer Res. 2007;67:10351–60.
    1. Storey A, Thomas M, Kalita A, et al. Role of a p53 polymorphism in the development of human papillomavirus-associated cancer. Nature. 1998;393:229–34.
    1. Dumont P, Leu JI, Della Pietra AC, 3rd, George DL, Murphy M. The codon 72 polymorphic variants of p53 have markedly different apoptotic potential. Nature genetics. 2003;33:357–65.
    1. Su WT, Alaminos M, Mora J, Cheung NK, La Quaglia MP, Gerald WL. Positional gene expression analysis identifies 12q overexpression and amplification in a subset of neuroblastomas. Cancer genetics and cytogenetics. 2004;154:131–7.
    1. Spitz R, Oberthuer A, Zapatka M, et al. Oligonucleotide array-based comparative genomic hybridization (aCGH) of 90 neuroblastomas reveals aberration patterns closely associated with relapse pattern and outcome. Genes Chromosomes Cancer. 2006;45:1130–42.
    1. Cattelani S, Defferrari R, Marsilio S, et al. Impact of a single nucleotide polymorphism in the MDM2 gene on neuroblastoma development and aggressiveness: results of a pilot study on 239 patients. Clin Cancer Res. 2008;14:3248–53.
    1. Perfumo C, Parodi S, Mazzocco K, et al. MDM2 SNP309 genotype influences survival of metastatic but not of localized neuroblastoma. Pediatr Blood Cancer. 2009;53:576–83.
    1. Gonzalez-Gomez P, Bello MJ, Lomas J, et al. Aberrant methylation of multiple genes in neuroblastic tumours. relationship with MYCN amplification and allelic status at 1p. Eur J Cancer. 2003;39:1478–85.
    1. Zhang Y, Xiong Y, Yarbrough WG. ARF promotes MDM2 degradation and stabilizes p53: ARF-INK4a locus deletion impairs both the Rb and p53 tumor suppression pathways. Cell. 1998;92:725–34.
    1. Bassi CL, Martelli L, Cipolotti R, Scrideli CA, Defavery R, Tone LG, et al. Lack of evidence for mutations or deletions in the CDKN2A/p16 and CDKN2B/p15 genes of Brazilian neuroblastoma patients. Brazilian journal of medical and biological research = Revista brasileira de pesquisas medicas e biologicas / Sociedade Brasileira de Biofisica. 2004;37:1683–7.
    1. Castresana JS, Gomez L, Garcia-Miguel P, Queizan A, Pestana A. Mutational analysis of the p16 gene in human neuroblastomas. Molecular carcinogenesis. 1997;18:129–33.
    1. Diccianni MB, Chau LS, Batova A, Vu TQ, Yu AL. The p16 and p18 tumor suppressor genes in neuroblastoma: implications for drug resistance. Cancer letters. 1996;104:183–92.
    1. Thompson PM, Maris JM, Hogarty MD, et al. Homozygous deletion of CDKN2A (p16INK4a/p14ARF) but not within 1p36 or at other tumor suppressor loci in neuroblastoma. Cancer Res. 2001;61:679–86.
    1. Chen Z, Lin Y, Barbieri E, et al. Mdm2 deficiency suppresses MYCN-Driven neuroblastoma tumorigenesis in vivo. Neoplasia. 2009;11:753–62.
    1. Van Maerken T, Speleman F, Vermeulen J, et al. Small-Molecule MDM2 Antagonists as a New Therapy Concept for Neuroblastoma. Cancer Res. 2006;66:9646–55.
    1. Barbieri E, Mehta P, Chen Z, et al. MDM2 inhibition sensitizes neuroblastoma to chemotherapy-induced apoptotic cell death. Mol Cancer Ther. 2006;5:2358–65.
    1. Van Maerken T, Ferdinande L, Taildeman J, et al. Antitumor Activity of the Selective MDM2 Antagonist Nutlin-3 Against Chemoresistant Neuroblastoma With Wild-Type p53. J Natl Cancer Inst. 2009;101:1562–74.
    1. Pearson AD, Pinkerton CR, Lewis IJ, Imeson J, Ellershaw C, Machin D. High-dose rapid and standard induction chemotherapy for patients aged over 1 year with stage 4 neuroblastoma: a randomised trial. The lancet oncology. 2008;9:247–56.
    1. Tweddle DA, Pinkerton CR, Lewis IJ, Ellershaw C, Cole M, Pearson AD. OPEC/OJEC for stage 4 neuroblastoma in children over 1 year of age. Medical and pediatric oncology. 2001;36:239–42.
    1. Cheung NK, Kushner BH, LaQuaglia M, et al. N7: a novel multi-modality therapy of high risk neuroblastoma (NB) in children diagnosed over 1 year of age. Medical and pediatric oncology. 2001;36:227–30.
    1. Berthold F, Hero B, Kremens B, et al. Long-term results and risk profiles of patients in five consecutive trials (1979-1997) with stage 4 neuroblastoma over 1 year of age. Cancer letters. 2003;197:11–7.

Source: PubMed

Подписаться