#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Assessment of the clinical utility of four NGS panels in myeloid malignancies. Suggestions for NGS panel choice or design


Autoři: Almudena Aguilera-Diaz aff001;  Iria Vazquez aff002;  Beñat Ariceta aff003;  Amagoia Mañú aff003;  Zuriñe Blasco-Iturri aff003;  Sara Palomino-Echeverría aff003;  María José Larrayoz aff002;  Ramón García-Sanz aff004;  María Isabel Prieto-Conde aff004;  María del Carmen Chillón aff004;  Ana Alfonso-Pierola aff005;  Felipe Prosper aff001;  Marta Fernandez-Mercado aff001;  María José Calasanz aff002
Působiště autorů: Advanced Genomics Laboratory, Hemato-Oncology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain aff001;  Navarra Institute for Health Research (IdiSNA), Pamplona, Spain aff002;  Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain aff003;  Hematology Department, University Hospital of Salamanca, IBSAL and CIBERONC, Salamanca, Spain aff004;  Hematology Department, Clinica Universidad de Navarra (CUN), Pamplona, Spain aff005;  Biomedical Engineering Department, School of Engineering, University of Navarra, San Sebastian, Spain aff006;  Scientific Co-Director of CIMA LAB Diagnostics, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain aff007
Vyšlo v časopise: PLoS ONE 15(1)
Kategorie: Research Article
doi: https://doi.org/10.1371/journal.pone.0227986

Souhrn

The diagnosis of myeloid neoplasms (MN) has significantly evolved through the last few decades. Next Generation Sequencing (NGS) is gradually becoming an essential tool to help clinicians with disease management. To this end, most specialized genetic laboratories have implemented NGS panels targeting a number of different genes relevant to MN. The aim of the present study is to evaluate the performance of four different targeted NGS gene panels based on their technical features and clinical utility. A total of 32 patient bone marrow samples were accrued and sequenced with 3 commercially available panels and 1 custom panel. Variants were classified by two geneticists based on their clinical relevance in MN. There was a difference in panel’s depth of coverage. We found 11 discordant clinically relevant variants between panels, with a trend to miss long insertions. Our data show that there is a high risk of finding different mutations depending on the panel of choice, due both to the panel design and the data analysis method. Of note, CEBPA, CALR and FLT3 genes, remains challenging the use of NGS for diagnosis of MN in compliance with current guidelines. Therefore, conventional molecular testing might need to be kept in place for the correct diagnosis of MN for now.

Klíčová slova:

Acute myeloid leukemia – Cancer detection and diagnosis – Gene sequencing – Genomic libraries – Human genetics – Mutation detection – Next-generation sequencing – Prognosis


Zdroje

1. Korn Claudia and Simón Méndez-Ferrer. Myeloid malignancies and the microenvironment. Blood. 2018;129: 811–823. doi: 10.1182/blood-2016-09-670224 28064238

2. Arber DA, Orazi A, Hasserjian R, Borowitz MJ, Beau MM Le, Bloomfield CD, et al. The 2016 revision to the World Health Organization classi fi cation of myeloid neoplasms and acute leukemia. Blood. 2016;127: 2391–2406. doi: 10.1182/blood-2016-03-643544 27069254

3. Papaemmanuil E, Gerstung M, Malcovati L, Tauro S, Gundem G, Loo P Van, et al. CME Article Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood. 2013;122: 3616–3627. doi: 10.1182/blood-2013-08-518886 24030381

4. Haferlach T, Nagata Y, Grossmann V, Okuno Y, Bacher U, Nagae G, et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia. 2014;28: 241–247. doi: 10.1038/leu.2013.336 24220272

5. Mughal TI, Cross NCP, Padron E, Tiu R V., Savona M, Malcovati L, et al. An international MDS/MPN working group’s perspective and recommendations on molecular pathogenesis, diagnosis and clinical characterization of myelodysplastic/myeloproliferative neoplasms. Haematologica. 2015;100: 1117–1130. doi: 10.3324/haematol.2014.114660 26341525

6. Itzykson R, Kosmider O, Renneville A, Gelsi-Boyer V, Meggendorfer M, Morabito M, et al. Prognostic Score Including Gene Mutations in Chronic Myelomonocytic Leukemia. J Clin Oncol. 2013;31: 2428–2436. doi: 10.1200/JCO.2012.47.3314 23690417

7. Meggendorfer M, Bacher U, Alpermann T, Haferlach C, Kern W, Gambacorti-Passerini C, et al. SETBP1 mutations occur in 9% of MDS/MPN and in 4% of MPN cases and are strongly associated with atypical CML, monosomy 7, isochromosome i(17)(q10), ASXL1 and CBL mutations. Leukemia. 2013;27: 1852–1860. doi: 10.1038/leu.2013.133 23628959

8. Patnaik MM, Tefferi A. Cytogenetic and molecular abnormalities in chronic myelomonocytic leukemia. Blood Cancer J. 2016;6: 1–8. doi: 10.1038/bcj.2016.5 26849014

9. Mangaonkar AA, Lasho TL, Finke CM, Gangat N, Al-Kali A, Elliott MA, et al. Prognostic interaction between bone marrow morphology and SF3B1 and ASXL1 mutations in myelodysplastic syndromes with ring sideroblasts. Blood Cancer J. 2018;8: 1–4. doi: 10.1038/s41408-017-0043-6

10. Bejar R, Stevenson KE, Caughey B, Lindsley RC, Mar BG, Stojanov P, et al. Somatic Mutations Predict Poor Outcome in Patients With Myelodysplastic Syndrome After Hematopoietic Stem-Cell Transplantation. J Clin Oncol. 2014;32: 2691–2698. doi: 10.1200/JCO.2013.52.3381 25092778

11. Tefferi A, Lasho TL, Patnaik MM, Saeed L, Mudireddy M, Idossa D, et al. Targeted next-generation sequencing in myelodysplastic syndromes and prognostic interaction between mutations and IPSS-R. Am J Hematol. 2017;92: 1311–1317. doi: 10.1002/ajh.24901 28875545

12. Serratì S, De Summa S, Pilato B, Petriella D, Lacalamita R, Tommasi S, et al. Next-generation sequencing: advances and applications in cancer diagnosis. Onco Targets Ther. 2016;9: 7355–7365. doi: 10.2147/OTT.S99807 27980425

13. Rack KA, Berg E van den, Haferlach C, Beverloo HB, Costa D, Espinet B, et al. European recommendations and quality assurance for cytogenomic analysis of haematological neoplasms. Leukemia. 2019. doi: 10.1038/s41375-019-0378-z 30696948

14. Palomo L, Ibáñez M, Abáigar M, Vázquez I, Álvarez S, Cabezón M, et al. Spanish Guidelines for the use of targeted deep sequencing in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br J Haematol. 2019;October: bjh.16175. doi: 10.1111/bjh.16175 31621063

15. Robinson JT, Thorvaldsdóttir H, Winckler W, Guttman M, Lander ES, Getz G1, et al. Integrative genomics viewer. Nat Biotechnol. 2011;29: 24–26. doi: 10.1038/nbt.1754 21221095

16. Gombart AF, Hofmann W, Kawano S, Takeuchi S, Krug U, Kwok SH, et al. Mutations in the gene encoding the transcription factor CCAAT / enhancer binding protein α in myelodysplastic syndromes and acute myeloid leukemias. 2013;99: 1332–1340. doi: 10.1182/blood.V99.4.1332 11830484

17. Lin L-I, Chen C-Y, Lin D-T, Tsay W, Tang J-L, Yeh Y-C, et al. Characterization of CEBPA Mutations in Acute Myeloid Leukemia: Most Patients with CEBPA Mutations Have Biallelic Mutations and Show a Distinct Immunophenotype of the Leukemic Cells. Clin Cancer Res. 2005;11: 1372–1379. doi: 10.1158/1078-0432.CCR-04-1816 15746035

18. Nangalia J, Massie CE, Baxter EJ, Nice FL, Gundem G, Wedge DC, et al. Somatic CALR Mutations in Myeloproliferative Neoplasms with Nonmutated JAK2. N Engl J Med. 2013;369: 2391–2405. doi: 10.1056/NEJMoa1312542 24325359

19. Nakao M, Yokota S, Iwai T, Kaneko H, Horiike S, Kashima K, et al. Internal tandem duplication of the flt3 gene found in acute myeloid leukemia. Leukemia. 1996;10: 1911–1918. 8946930

20. Thomas M, Sukhai MA, Zhang T, Dolatshahi R, Harbi D, Garg S, et al. Integration of Technical, Bioinformatic, and Variant Assessment Approaches in the Validation of a Targeted Next-Generation Sequencing Panel for Myeloid Malignancies. Arch Pathol Lab Med. 2017;141: 759–775. doi: 10.5858/arpa.2016-0547-RA 28557600

21. Yamamoto Y, Kiyoi H, Nakano Y, Suzuki R, Kodera Y, Miyawaki S, et al. Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood. 2001;97: 2434–2439. doi: 10.1182/blood.v97.8.2434 11290608

22. Pellagatti A, Armstrong RN, Steeples V, Sharma E, Repapi E, Singh S, et al. Impact of spliceosome mutations on RNA splicing in myelodysplasia: Dysregulated genes/pathways and clinical associations. Blood. 2018;132: 1225–1240. doi: 10.1182/blood-2018-04-843771 29930011

23. Kim KH, Roberts CWM. Targeting EZH2 in cancer. Nat Med. 2016;22: 128–134. doi: 10.1038/nm.4036 26845405

24. Makishima H, Yoshizato T, Yoshida K, Sekeres MA, Radivoyevitch T, Suzuki H, et al. Dynamics of clonal evolution in myelodysplastic syndromes. Nat Genet. 2017;49: 204–212. doi: 10.1038/ng.3742 27992414

25. Murati A, Brecqueville M, Devillier R, Mozziconacci MJ, Gelsi-Boyer V, Birnbaum D. Myeloid malignancies: mutations, models and management. BMC Cancer. 2012;12. doi: 10.1186/1471-2407-12-304 22823977

26. Gill H, Leung A, Kwong Y-L. Molecular and Cellular Mechanisms of Myelodysplastic Syndrome: Implications on Targeted Therapy. Int J Mol Sci. 2016;17: 440. doi: 10.3390/ijms17040440 27023522

27. Lee JH, Jeong H, Choi JW, Oh H, Kim Y-S. Clinicopathologic significance of MYD88 L265P mutation in diffuse large B-cell lymphoma: a meta-analysis. Sci Rep. 2017;7: 1–8. doi: 10.1038/s41598-016-0028-x

28. Hu Y, Su H, Liu C, Wang Z, Huang L, Wang Q, et al. DEPTOR is a direct NOTCH1 target that promotes cell proliferation and survival in T-cell leukemia. Oncogene. 2017;36: 1038–1047. doi: 10.1038/onc.2016.275 27593934

29. Stenzinger A, Endris V, Pfarr N, Andrulis M, Jöhrens K, Klauschen F, et al. Targeted ultra-deep sequencing reveals recurrent and mutually exclusive mutations of cancer genes in blastic plasmacytoid dendritic cell neoplasm. Oncotarget. 2014;5: 6404–6413. doi: 10.18632/oncotarget.2223 25115387

30. Bello E, Pellagatti A, Shaw J, Mecucci C, Kušec R, Killick S, et al. CSNK1A1 mutations and gene expression analysis in myelodysplastic syndromes with del(5q). Br J Haematol. 2015;171: 210–214. doi: 10.1111/bjh.13563 26085061

31. Boudry-Labis E, Roche-Lestienne C, Nibourel O, Boissel N, Terre C, Perot C, et al. Neurofibromatosis-1 gene deletions and mutations in de novo adult acute myeloid leukemia. Am J Hematol. 2013;88: 306–311. doi: 10.1002/ajh.23403 23460398

32. Wong TN, Miller CA, Jotte MRM, Bagegni N, Baty JD, Schmidt AP, et al. Cellular stressors contribute to the expansion of hematopoietic clones of varying leukemic potential. Nat Commun. 2018;9: 1–10. doi: 10.1038/s41467-017-02088-w

33. Maslah N, Cassinat B, Verger E, Kiladjian JJ, Velazquez L. The role of LNK/SH2B3 genetic alterations in myeloproliferative neoplasms and other hematological disorders. Leukemia. 2017;31: 1661–1670. doi: 10.1038/leu.2017.139 28484264

34. Patnaik MM. The importance of FLT3 mutational analysis in acute myeloid leukemia. Leuk Lymphoma. 2018;59: 2273–2286. doi: 10.1080/10428194.2017.1399312 29164965

35. Bullinger L, Döhner K, Dohner H. Genomics of acute myeloid leukemia diagnosis and pathways. J Clin Oncol. 2017;35: 934–946. doi: 10.1200/JCO.2016.71.2208 28297624

36. Metzeler KH, Herold T, Rothenberg-Thurley M, Amler S, Sauerland MC, Görlich D, et al. Spectrum and prognostic relevance of driver gene mutations in acute myeloid leukemia. Blood. 2016;128: 686–698. doi: 10.1182/blood-2016-01-693879 27288520

37. Zoi K, Cross NCP. Genomics of myeloproliferative neoplasms. J Clin Oncol. 2017;35: 947–955. doi: 10.1200/JCO.2016.70.7968 28297629

38. Pardanani A, Lasho TL, Laborde RR, Elliott M, Hanson CA, Knudson RA, et al. CSF3R T618I is a highly prevalent and specific mutation in chronic neutrophilic leukemia. Leukemia. 2013;27: 1870–1873. doi: 10.1038/leu.2013.122 23604229

39. Perry AM, Attar EC. New Insights in AML Biology From Genomic Analysis. Semin Hematol. 2014;51: 282–297. doi: 10.1053/j.seminhematol.2014.08.005 25311741

40. Makishima H. Somatic SETBP1 mutations in myeloid neoplasms. Int J Hematol. 2017;105: 732–742. doi: 10.1007/s12185-017-2241-1 28447248

41. Lasho TL, Finke CM, Hanson CA, Jimma T, Knudson RA, Ketterling RP, et al. SF3B1 mutations in primary myelofibrosis: Clinical, histopathology and genetic correlates among 155 patients. Leukemia. 2012;26: 1135–1137. doi: 10.1038/leu.2011.320 22064353

42. Pellagatti A, Boultwood J. The molecular pathogenesis of the myelodysplastic syndromes. Eur J Haematol. 2015;95: 3–15. doi: 10.1111/ejh.12515 25645650

43. Terada K, Yamaguchi H, Ueki T, Usuki K, Kobayashi Y, Tajika K, et al. Usefulness of BCOR gene mutation as a prognostic factor in acute myeloid leukemia with intermediate cytogenetic prognosis. Genes Chromosom Cancer. 2018;57: 401–408. doi: 10.1002/gcc.22542 29663558

44. Crispino JD, Horwitz MS. GATA factor mutations in hematologic disease. Blood. 2017;129: 2103–2110. doi: 10.1182/blood-2016-09-687889 28179280

45. Song J, Hussaini M, Zhang H, Shao H, Qin D, Zhang X, et al. Comparison of the Mutational Profiles of Primary Myelofibrosis, Polycythemia Vera, and Essential Thrombocytosis. Am J Clin Pathol. 2017;147: 444–452. doi: 10.1093/ajcp/aqw222 28419183

46. Mori T, Nagata Y, Makishima H, Sanada M, Shiozawa Y, Kon A, et al. Somatic PHF6 mutations in 1760 cases with various myeloid neoplasms. Leukemia. 2016;30: 2270–2273. doi: 10.1038/leu.2016.212 27479181

47. Herbaux C, Duployez N, Badens C, Poret N, Gardin C, Decamp M, et al. Incidence of ATRX mutations in myelodysplastic syndromes, the value of microcytosis. Am J Hematol. 2015;90: 737–738. doi: 10.1002/ajh.24073 26017030

48. Wong CC, Martincorena I, Rust AG, Rashid M, Alifrangis C, Alexandrov LB, et al. Inactivating CUX1 mutations promote tumorigenesis. Nat Genet. 2014;46: 33–38. doi: 10.1038/ng.2846 24316979

49. Hirabayashi S, Wlodarski MW, Kozyra E, Niemeyer CM. Heterogeneity of GATA2-related myeloid neoplasms. Int J Hematol. 2017;106: 175–182. doi: 10.1007/s12185-017-2285-2 28643018

50. Jäger R, Gisslinger H, Passamonti F, Rumi E, Berg T, Gisslinger B, et al. Deletions of the transcription factor Ikaros in myeloproliferative neoplasms. Leukemia. 2010;24: 1290–1298. doi: 10.1038/leu.2010.99 20508609

51. Thol F, Bollin R, Gehlhaar M, Walter C, Dugas M, Suchanek KJ, et al. Mutations in the cohesin complex in acute myeloid leukemia: clinical and prognostic implications. Blood. 2014;123: 914–920. doi: 10.1182/blood-2013-07-518746 24335498

52. Kon A, Shih LY, Minamino M, Sanada M, Shiraishi Y, Nagata Y, et al. Recurrent mutations in multiple components of the cohesin complex in myeloid neoplasms. Nat Genet. 2013;45: 1232–1237. doi: 10.1038/ng.2731 23955599

53. Ha Jung-Sook and Jeon Dong-Seok. Possible new LNK mutations in myeloproliferative neoplasms. Am J Hematol. 2011;86: 866–868. doi: 10.1002/ajh.22107 21922527

54. Hurtado C, Erquiaga I, Aranaz P, Miguéliz I, García-Delgado M, Novo FJ, et al. LNK can also be mutated outside PH and SH2 domains in myeloproliferative neoplasms with and without V617FJAK2 mutation. Leuk Res. 2011;35: 1537–1539. doi: 10.1016/j.leukres.2011.07.009 21794913

55. Lasho TL, Tefferi A, Finke C, Pardanani A. Clonal hierarchy and allelic mutation segregation in a myelofibrosis patient with two distinct LNK mutations. Leukemia. 2011;25: 1056–1058. doi: 10.1038/leu.2011.45 21415853

56. Kantarjian HM, Keating MJ, Freireich EJ. Toward the potential cure of leukemias in the next decade. Cancer. 2018;124: 4301–4313. doi: 10.1002/cncr.31669 30291792

57. Stone RM. Which new agents will be incorporated into frontline therapy in acute myeloid leukemia? Best Pract Res Clin Haematol. 2017;30: 312–316. doi: 10.1016/j.beha.2017.09.006 29156201

58. Bejar R, Lord A, Stevenson K, Bar-Natan M, Pérez-Ladaga A, Zaneveld J, et al. TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients. Blood. 2014;124: 2705–2712. doi: 10.1182/blood-2014-06-582809 25224413

59. Metzeler KH, Walker A, Geyer S, Garzon R, Klisovic RB, Bloomfield D, et al. DNMT3A mutations and response to the hypomethylating agent decitabine in acute myeloid leukemia. Leukemia. Nature Publishing Group; 2012. doi: 10.1038/leu.2011.342 22124213

60. Luskin MR, Lee JW, Fernandez HF, Abdel-Wahab O, Bennett JM, Ketterling RP, et al. Benefit of high-dose daunorubicin in AML induction extends across cytogenetic and molecular groups. Blood. 2016;127: 1551–1558. doi: 10.1182/blood-2015-07-657403 26755712

61. Pabst T, Eyholzer M, Fos J, Mueller BU. Heterogeneity within AML with CEBPA mutations; Only CEBPA double mutations, but not single CEBPA mutations are associated with favourable prognosis. Br J Cancer. 2009;100: 1343–1346. doi: 10.1038/sj.bjc.6604977 19277035

62. Au CH, Wa A, Ho DN, Chan TL, Ma ESK. Clinical evaluation of panel testing by next-generation sequencing (NGS) for gene mutations in myeloid neoplasms. Diagn Pathol. 2016;11:11. doi: 10.1186/s13000-016-0456-8 26796102

63. Albitar A, Townsley D, Ma W, De Dios I, Funari V, Young NS, et al. Prevalence of somatic mutations in patients with aplastic anemia using peripheral blood cfDNA as compared with BM. Leukemia. 2018;32: 227–229. doi: 10.1038/leu.2017.271 28832022

64. Taylor J, Xiao W, Abdel-wahab O. Diagnosis and classification of hematologic malignancies on the basis of genetics. Blood. 2017;130: 410–424. doi: 10.1182/blood-2017-02-734541 28600336

65. Hattori K, Sakata-Yanagimoto M, Suehara Y, Yokoyama Y, Kato T, Kurita N, et al. Clinical significance of disease-specific MYD88 mutations in circulating DNA in primary central nervous system lymphoma. Cancer Sci. 2018;109: 225–230. doi: 10.1111/cas.13450 29151258

66. Qin SC, Xia Y, Miao Y, Zhu HY, Wu JZ, Fan L, et al. MYD88 mutations predict unfavorable prognosis in chronic lymphocytic leukemia patients with mutated IGHV gene. Blood Cancer J. 2017;7. doi: 10.1038/s41408-017-0014-y 29242635

67. Varettoni M, Arcaini L, Zibellini S, Boveri E, Rattotti S, Riboni R, et al. Prevalence and clinical significance of the MYD88 (L265P) somatic mutation in Waldenstrom’s macroglobulinemia and related lymphoid neoplasms. Blood. 2013;121: 2522–2528. doi: 10.1182/blood-2012-09-457101 23355535

68. Young AL, Challen GA, Birmann BM, Druley TE. Clonal haematopoiesis harbouring AML-associated mutations is ubiquitous in healthy adults. Nat Commun. 2016;7:12454. doi: 10.1038/ncomms12454

69. Hung SS, Meissner B, Chavez EA, Ben-Neriah S, Ennishi D, Jones MR, et al. Assessment of Capture and Amplicon-Based Approaches for the Development of a Targeted Next-Generation Sequencing Pipeline to Personalize Lymphoma Management. J Mol Diagnostics. 2018;20: 203–214. doi: 10.1016/j.jmoldx.2017.11.010 29429887

70. Kadri S, Zhen CJ, Wurst MN, Long BC, Jiang ZF, Wang YL, et al. Amplicon Indel Hunter Is a Novel Bioinformatics Tool to Detect Large Somatic Insertion/Deletion Mutations in Amplicon-Based Next-Generation Sequencing Data. J Mol Diagnostics. 2015;17: 635–643. doi: 10.1016/j.jmoldx.2015.06.005 26319364

71. Patel KP, Barkoh BA, Chen Z, Ma D, Reddy N, Medeiros LJ, et al. Diagnostic testing for IDH1 and IDH2 variants in acute myeloid leukemia an algorithmic approach using high-resolution melting curve analysis. J Mol Diagnostics. 2011;13: 678–686. doi: 10.1016/j.jmoldx.2011.06.004 21889610

72. Petrova L, Vrbacky F, Lanska M, Zavrelova A, Zak P, Hrochova K. IDH1 and IDH2 mutations in patients with acute myeloid leukemia: Suitable targets for minimal residual disease monitoring? Clin Biochem. 2018;61: 34–39. doi: 10.1016/j.clinbiochem.2018.08.012 30176240

73. Bannon SA, Dinardo CD. Hereditary predispositions to myelodysplastic syndrome. Int J Mol Sci. 2016;17: 838. doi: 10.3390/ijms17060838 27248996

74. Babushok D V, Bessler M, Olson TS. Genetic predisposition to myelodysplastic syndrome and acute myeloid leukemia in children and young adults. Leuk Lymphoma. 2016;57: 520–536. doi: 10.3109/10428194.2015.1115041 26693794

75. Pippucci T, Savoia A, Perrotta S, Pujol-Moix N, Noris P, Castegnaro G, et al. Mutations in the 5′ UTR of ANKRD26, the ankirin repeat domain 26 gene, cause an autosomal-dominant form of inherited thrombocytopenia, THC2. Am J Hum Genet. 2011;88: 115–120. doi: 10.1016/j.ajhg.2010.12.006 21211618

76. Marconi C, Canobbio I, Bozzi V, Pippucci T, Simonetti G, Melazzini F, et al. 5′UTR point substitutions and N-terminal truncating mutations of ANKRD26 in acute myeloid leukemia. J Hematol Oncol. 2017;10: 18. doi: 10.1186/s13045-016-0382-y 28100250

77. Schnittger S, Bacher U, Haferlach C, Alpermann T, Kern W, Haferlach T. Diversity of the Juxtamembrane and TKD I Mutations (Exons 13–15) in the FLT3 Gene with Regards to Mutant Load, Sequence, Length, Localization, and Correlation with Biological Data. Genes Chromosom Cancer. 2012;51: 910–924. doi: 10.1002/gcc.21975 22674490

78. Cabagnols X, Favale F, Pasquier F, Messaoudi K, Defour JP, Ianotto JC, et al. Presence of atypical thrombopoietin receptor (MPL) mutations in triple-negative essential thrombocythemia patients. Blood. 2016;127: 333–342. doi: 10.1182/blood-2015-07-661983 26450985

79. Milosevic Feenstra JD, Nivarthi H, Gisslinger H, Leroy E, Rumi E, Chachoua I, et al. Whole-exome sequencing identifies novel MPL and JAK2 mutations in triple-negative myeloproliferative neoplasms. Blood. 2016;127: 325–332. doi: 10.1182/blood-2015-07-661835 26423830

80. Dohner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Büchner T, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129: 424–447. doi: 10.1182/blood-2016-08-733196 27895058

81. Alonso CM, Llop M, Sargas C, Pedrola L, Panadero J, Hervás D, et al. Clinical Utility of a Next-Generation Sequencing Panel for Acute Myeloid Leukemia Diagnostics. J Mol Diagnostics. 2019;21: 228–240. doi: 10.1016/j.jmoldx.2018.09.009 30576870

82. Schranz K, Hubmann M, Harin E, Vosberg S, Herold T, Metzeler KH, et al. Clonal heterogeneity of FLT3-ITD detected by high-throughput amplicon sequencing correlates with adverse prognosis in acute myeloid leukemia. Oncotarget. 2018;9: 30128–30145. doi: 10.18632/oncotarget.25729 30046393

83. Bacher U, Shumilov E, Flach J, Porret N, Joncourt R, Wiedemann G, et al. Challenges in the introduction of next-generation sequencing (NGS) for diagnostics of myeloid malignancies into clinical routine use. Blood Cancer J. 2018;8: 113. doi: 10.1038/s41408-018-0148-6 30420667

84. Baker SC. Nex-Generation sequencing challenges. Genet Eng Biotechnol News. 2017;37: accessed on Feb 4, 2019. Available: https//www.genengnews.com/magazine/286/next-generation-sequencing-challenges


Článek vyšel v časopise

PLOS One


2020 Číslo 1
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

KOST
Koncepce osteologické péče pro gynekology a praktické lékaře
nový kurz
Autoři: MUDr. František Šenk

Sekvenční léčba schizofrenie
Autoři: MUDr. Jana Hořínková

Hypertenze a hypercholesterolémie – synergický efekt léčby
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Svět praktické medicíny 5/2023 (znalostní test z časopisu)

Imunopatologie? … a co my s tím???
Autoři: doc. MUDr. Helena Lahoda Brodská, Ph.D.

Všechny kurzy
Kurzy Podcasty Doporučená témata Časopisy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#