Page 15 - 2019_04-Haematologica-web
P. 15

Editorials
and 15q (Figure 1).11-13 Deletion of the 9p21.3 locus is the most recurrent event in BPDCN, found to be associated with a poor outcome when biallelic.11 More recently, the presence of a recurrent gene rearrangement involving the MYC locus, specifically t(6;8)(p21;q24) has been reported in several studies including one by our own group, and has been seen in association with older onset and shorter median survival.14-16 Whole-exome sequencing (WES) and targeted sequencing studies have identified recurrent mutations involving TET2, ASXL1, TP53, and NPM1,17,18 while separate studies have appointed E-box transcription factor TCF4 as a master regulator of the BPDCN onco- genic program as bromodomain and extra-terminal domain inhibitors (BETis) induced BPDCN apoptosis due to disruption of the TCF4 dependent regulatory network.19 Finally, aberrant activation of the NF-kB pathway has been identified through gene expression profiling.20 However, as can be seen, our understanding of the genetic aspects of BPDCN has been somewhat limited.
In the current issue of the Journal, Sapienza et al.2 signif- icantly advance our understanding of BPDCN by analyz- ing 14 patients, as well as the patient-derived CAL-1 cell line, by WES. This broad and detailed sequencing demon- strated that BPDCN patients were affected by mutations of genes involved in epigenetic regulation, with 25 mutat- ed epigenetic modifier genes including those implicated in DNA methylation (TET2 and IDH2), chromatin accessibil- ity (ARID1a, CHD8, SMARCA1), and histone modification including: methylation (ASXL1, SUZ12, MLL), demethyla- tion (KDM4D), acetylation (EP300, EP400), ubiquitination (PHC1, PHC2), dephosphorylation (EYA2) and exchange (SRCAP) (Figure 1). This finding highlights the dysregula- tion of the epigenetic program in BPDCN as a hallmark of the disease indicating possible therapeutic interventions. In addition, by analyzing the transcriptome of the samples studied by WES, they examined the specific impact of these epigenetic-associated gene mutations. Gene set enrichment analysis additionally revealed two significant deregulation signatures associated with methylation of DNA: one driven by KDM5B34 histone demethylase and another by the PRMT5 methyltransferase-associated gene. The authors also detected gene set enrichment of those genes associated with response to decitabine, a DNA demethylating agent. Finally, based on the apparent signif- icance of epigenetics in BPDCN, the authors did what few have done before, and tested in vivo the efficacy of four US Food and Drug Administration (FDA)-approved epigenetic drugs (5’-Azacytidine, decitabine, romidepsin and borte- zomib) in a mouse xenograft model using the CAL-1 cell line. These drugs were used as single agents or in combi- nation, and when used as a single agent, 5’-Azacytidine and decitabine significantly prolonged mice overall sur- vival, while among all the combinations tested, treatment with 5’-Azacytidine in combination with decitabine achieved the best result in terms of survival. The signifi- cance of this study cannot be understated as it links geno- type to advancements in epigenetic forms of treatment and fundamental biological processes.
Blastic plasmacytoid dendritic cell neoplasm is a rare dis- ease with an extremely aggressive behavior; the advances here in the genetics and molecular aspects of this entity, as well as the therapeutic approach, are quite revealing and
significantly open the door for future clinical trials. This work nicely provides further support for the essence of why development of new epigenetic treatment strategies are a rational approach for this aggressive disease.
References
1. Laribi K, Denizon N, Besancon A, et al. Blastic Plasmacytoid Dendritic Cell Neoplasm: From Origin of the Cell to Targeted Therapies. Biol Blood Marrow Transplant. 2016;22(8):1357-1367.
2. SapienzaMR,AbateF,MelleF,OrecchioniFF,EtebariM.Blasticplas- macytoid dendritic cell neoplasm: genomics mark epigenetic dysreg- ulation as a primary therapeutic target. Haematologica. 2019;104(4): 729-737.
3. Tauchi T, Ohyashiki K, Ohyashiki JH, et al. CD4+ and CD56+ acute monoblastic leukemia. Am J Hematol. 1990;34(3):228-229.
4. Gattei V, Carbone A, Zagonel V, Pinto A. Expression of natural killer antigens in a subset of 'non-T, non-B lymphoma/leukaemia with his- tiocytic features'. Br J Haematol. 1990;76(3):444-448.
5. Kimura S, Kakazu N, Kuroda J, et al. Agranular CD4+CD56+ blastic natural killer leukemia/lymphoma. Ann Hematol. 2001;80(4):228- 231.
6. Kameoka J, Ichinohasama R, Tanaka M, et al. A cutaneous agranular CD2- CD4+ CD56+ "lymphoma": report of two cases and review of the literature. Am J Clin Pathol. 1998;110(4):478-488.
7. Lucio P, Parreira A, Orfao A. CD123hi dendritic cell lymphoma: an unusual case of non-Hodgkin lymphoma. Ann Intern Med. 1999;131(7):549-550.
8. Grouard G, Rissoan MC, Filgueira L, Durand I, Banchereau J, Liu YJ. The enigmatic plasmacytoid T cells develop into dendritic cells with interleukin (IL)-3 and CD40-ligand. J Exp Med. 1997;185(6):1101- 1111.
9. Herling M, Teitell MA, Shen RR, Medeiros LJ, Jones D. TCL1 expres- sion in plasmacytoid dendritic cells (DC2s) and the related CD4+ CD56+ blastic tumors of skin. Blood. 2003;101(12):5007-5009.
10. PetrellaT,ComeauMR,MaynadieM,etal.'AgranularCD4+CD56+ hematodermic neoplasm' (blastic NK-cell lymphoma) originates from a population of CD56+ precursor cells related to plasmacytoid mono- cytes. Am J Surg Pathol. 2002;26(7):852-862.
11. Lucioni M, Novara F, Fiandrino G, et al. Twenty-one cases of blastic plasmacytoid dendritic cell neoplasm: focus on biallelic locus 9p21.3 deletion. Blood. 2011;118(17):4591-4594.
12. Dijkman R, van Doorn R, Szuhai K, Willemze R, Vermeer MH, Tensen CP. Gene-expression profiling and array-based CGH classify CD4+CD56+ hematodermic neoplasm and cutaneous myelomono- cytic leukemia as distinct disease entities. Blood. 2007;109(4):1720- 1727.
13. Leroux D, Mugneret F, Callanan M, et al. CD4(+), CD56(+) DC2 acute leukemia is characterized by recurrent clonal chromosomal changes affecting 6 major targets: a study of 21 cases by the Groupe Francais de Cytogenetique Hematologique. Blood. 2002;99(11):4154- 4159.
14. Sakamoto K, Katayama R, Asaka R, et al. Recurrent 8q24 rearrange- ment in blastic plasmacytoid dendritic cell neoplasm: association with immunoblastoid cytomorphology, MYC expression, and drug response. Leukemia. 2018;32(12):2590-2603.
15. Boddu PC, Wang SA, Pemmaraju N, et al. 8q24/MYC rearrangement is a recurrent cytogenetic abnormality in blastic plasmacytoid den- dritic cell neoplasms. Leuk Res. 2018;66:73-78.
16. Sumarriva Lezama L, Chisholm KM, Carneal E, et al. An analysis of blastic plasmacytoid dendritic cell neoplasm with translocations involving the MYC locus identifies t(6;8)(p21;q24) as a recurrent cyto- genetic abnormality. Histopathology. 2018;73(5):767-776.
17. MenezesJ,AcquadroF,WisemanM,etal.Exomesequencingreveals novel and recurrent mutations with clinical impact in blastic plasma- cytoid dendritic cell neoplasm. Leukemia. 2014;28(4):823-829.
18. Jardin F, Ruminy P, Parmentier F, et al. TET2 and TP53 mutations are frequently observed in blastic plasmacytoid dendritic cell neoplasm. Br J Haematol. 2011;153(3):413-416.
19. Ceribelli M, Hou ZE, Kelly PN, et al. A Druggable TCF4- and BRD4- Dependent Transcriptional Network Sustains Malignancy in Blastic Plasmacytoid Dendritic Cell Neoplasm. Cancer Cell. 2016;30(5):764- 778.
20. Sapienza MR, Fuligni F, Agostinelli C, et al. Molecular profiling of blastic plasmacytoid dendritic cell neoplasm reveals a unique pattern and suggests selective sensitivity to NF-kB pathway inhibition. Leukemia. 2014;28(8):1606-1616.
haematologica | 2019; 104(4)
643


































































































   13   14   15   16   17