Page 29 - 2018_11-Haematologica-web
P. 29

Preclinical models of mastocytosis
LUVA, ROSA and MCPV-1. While none of these cell lines simultaneously expresses the KIT D816V mutant and a functional FcεRI, making them useless for testing MC-sta- bilizing drugs or drugs interfering with FcεRI-induced sig- naling in the context of KIT D816V+ SM, some of these cell lines may qualify as MCL-like since they harbor SM-relat- ed KIT variants and/or other oncogenic molecules relevant to SM. Among all MC lines, HMC-1 cells have been most frequently used, but other more recently established MC lines, such as ROSA and MCPV-1, are now available and are being used in various preclinical studies. For example, these cell lines have been used to analyze in vitro the growth-inhibitory effects of antineoplastic drugs, including various KIT-TKI, on neoplastic MC. However, because neoplastic MC in advanced SM are triggered by KIT-inde- pendent signaling pathways and additional genetic lesions that confer resistance against KIT-TKI, it might be interest- ing to establish in vitro models of multi-mutated neoplastic MC, starting from established human KIT mutant-positive MC lines in which additional lesions, such as the S/A/R
combination of molecular lesions might be introduced. Such multi-mutated neoplastic MC lines should be useful to test combination therapies in vitro, and later in clinical tri- als in patients with advanced SM. With these approaches, new therapeutic concepts should be established in order to improve therapy in advanced SM.
Acknowledgments
The authors would like to thank Dr Patrick T. Gunning (Department of Medical Biophysics, University of Toronto, Ontario, Canada) for kindly providing the STAT-specific inhibitors presented in the manuscript. We also express our deep thanks to Dr Fawzia Louache (Inserm UMRS-1170, CNRS GDR 3697 Micronit, Institut Gustave Roussy, Université Paris- Sud, Université Paris-Saclay, Villejuif, France) for her invalu- able help with the in vivo studies presented in the manuscript. K.H. is supported by a research grant from the German Research Council (DFG; HA 2393/6-1). DM is supported by the DIR, NIAID, and PV is supported by the Austrian Science Fund (FWF) grants F 4701-B20 and F 4704-B20.
References
1. DeBruinEJ,GoldM,LoBC,etal.Mastcells in human health and disease. Methods Mol Biol. 2015;1220:93-119.
2. GalliSJ,TsaiM.IgEandmastcellsinallergic disease. Nat Med. 2012;18(5):693-704.
3. Kirshenbaum AS, Metcalfe DD. Growth of human mast cells from bone marrow and peripheral blood-derived CD34+ pluripo- tent progenitor cells. Methods Mol Biol. 2006;315:105-112.
4. GalliSJ,TsaiM,WershilBK,TamSY,Costa JJ. Regulation of mouse and human mast cell development, survival and function by stem cell factor, the ligand for the c-kit receptor. Int Arch Allergy Immunol. 1995;107(1-3):51- 53.
5. Brown JM, Wilson TM, Metcalfe DD. The mast cell and allergic diseases: role in patho- genesis and implications for therapy. Clin Exp Allergy. 2008;38(1):4-18.
6. Arock M, Valent P. Pathogenesis, classifica- tion and treatment of mastocytosis: state of the art in 2010 and future perspectives. Expert Rev Hematol. 2010;3(4):497-516.
7. Valent P, Akin C, Hartmann K, et al. Advances in the classification and treatment of mastocytosis: current status and outlook toward the future. Cancer Res. 2017;77 (6):1261-1270.
8. Pardanani A. Systemic mastocytosis in adults: 2017 update on diagnosis, risk strati- fication and management. Am J Hematol. 2016;91 (11):1146-1159.
9. Valent P, Horny HP, Escribano L, et al. Diagnostic criteria and classification of mas- tocytosis: a consensus proposal. Leuk Res. 2001;25(7):603-625.
10. ValentP,EscribanoL,Broesby-OlsenS,etal. Proposed diagnostic algorithm for patients with suspected mastocytosis: a proposal of the European Competence Network on Mastocytosis. Allergy. 2014;69(10):1267- 1274.
11. Butterfield JH, Weiler D, Dewald G, Gleich GJ. Establishment of an immature mast cell line from a patient with mast cell leukemia. Leuk Res. 1988;12(4):345-355.
12. SundstromM,VliagoftisH,KarlbergP,etal.
Functional and phenotypic studies of two variants of a human mast cell line with a dis- tinct set of mutations in the c-kit proto- oncogene. Immunology. 2003;108(1):89-97.
13. Kirshenbaum AS, Akin C, Wu Y, et al. Characterization of novel stem cell factor responsive human mast cell lines LAD 1 and 2 established from a patient with mast cell sarcoma/leukemia; activation following aggregation of FcepsilonRI or FcgammaRI. Leuk Res. 2003;27(8):677-682.
14. Laidlaw TM, Steinke JW, Tinana AM, et al. Characterization of a novel human mast cell line that responds to stem cell factor and expresses functional FcepsilonRI. J Allergy Clin Immunol. 2011;127(3):815-822.
15. Saleh R, Wedeh G, Herrmann H, et al. A new human mast cell line expressing a func- tional IgE receptor converts to tumorigenic growth by KIT D816V transfection. Blood. 2014;124(1):111-120.
16. Hoermann G, Blatt K, Greiner G, et al. CD52 is a molecular target in advanced systemic mastocytosis. FASEB J. 2014;28(8):3540- 3551.
17. Kanakura Y, Furitsu T, Tsujimura T, et al. Activating mutations of the c-kit proto- oncogene in a human mast cell leukemia cell line. Leukemia. 1994;8 (Suppl 1):S18-22.
18. Cruse G, Metcalfe DD, Olivera A. Functional deregulation of KIT: link to mast cell proliferative diseases and other neo- plasms. Immunol Allergy Clin North Am. 2014;34 (2):219-237.
19. Longley BJ, Tyrrell L, Lu SZ, et al. Somatic c- KIT activating mutation in urticaria pigmen- tosa and aggressive mastocytosis: establish- ment of clonality in a human mast cell neo- plasm. Nat Genet. 1996;12(3):312-314.
20. Bodemer C, Hermine O, Palmerini F, et al. Pediatric mastocytosis is a clonal disease associated with D816V and other activating c-KIT mutations. J Invest Dermatol. 2010;130(3):804815.
21. Arock M, Sotlar K, Akin C, et al. KIT muta- tion analysis in mast cell neoplasms: recom- mendations of the European Competence Network on Mastocytosis. Leukemia. 2015;29(6):1223-1232.
22. Harir N, Boudot C, Friedbichler K, et al. Oncogenic Kit controls neoplastic mast cell
growth through a Stat5/PI3-kinase signaling
cascade. Blood. 2008;112(6):2463-2473.
23. Bibi S, Arslanhan MD, Langenfeld F, et al. Co-operating STAT5 and AKT signaling pathways in chronic myeloid leukemia and mastocytosis: possible new targets of thera-
py. Haematologica. 2014;99(3):417-429.
24. Baumgartner C, Cerny-Reiterer S, Sonneck K, et al. Expression of activated STAT5 in neoplastic mast cells in systemic mastocyto- sis: subcellular distribution and role of the transforming oncoprotein KIT D816V. Am J
Pathol. 2009;175(6):2416-2429.
25. Voisset E, Lopez S, Dubreuil P, De Sepulveda
P. The tyrosine kinase FES is an essential effector of KITD816V proliferation signal. Blood. 2007;110(7):2593-2599.
26. Smrz D, Kim MS, Zhang S, et al. mTORC1 and mTORC2 differentially regulate home- ostasis of neoplastic and non-neoplastic human mast cells. Blood. 2011;118(26): 6803-6813.
27. Sun J, Pedersen M, Ronnstrand L. The D816V mutation of c-Kit circumvents a requirement for Src family kinases in c-Kit signal transduction. J Biol Chem. 2009;284(17):11039-11047.
28. Gleixner KV, Mayerhofer M, Cerny-Reiterer S, et al. KIT-D816V-independent oncogenic signaling in neoplastic cells in systemic mas- tocytosis: role of Lyn and Btk activation and disruption by dasatinib and bosutinib. Blood. 2011;118(7):1885-1898.
29. Zermati Y, De Sepulveda P, Feger F, et al. Effect of tyrosine kinase inhibitor STI571 on the kinase activity of wild-type and various mutated c-kit receptors found in mast cell neoplasms. Oncogene. 2003;22(5):660-664.
30. Alvarez-Twose I, Matito A, Morgado JM, et al. Imatinib in systemic mastocytosis: a phase IV clinical trial in patients lacking exon 17 KIT mutations and review of the lit- erature. Oncotarget. 2017;8(40):68950- 68963.
31. Traina F, Visconte V, Jankowska AM, et al. Single nucleotide polymorphism array lesions, TET2, DNMT3A, ASXL1 and CBL mutations are present in systemic mastocy- tosis. PLoS One. 2012;7(8):e43090.
32. Soucie E, Hanssens K, Mercher T, et al. In aggressive forms of mastocytosis, TET2 loss
haematologica | 2018; 103(11)
1769


































































































   27   28   29   30   31