Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-ttngx Total loading time: 0 Render date: 2024-06-08T09:23:24.942Z Has data issue: false hasContentIssue false

Chapter 13 - Myeloid and Lymphoid Neoplasms Associated with Eosinophilia

Published online by Cambridge University Press:  12 November 2020

Jon van der Walt
Affiliation:
St Thomas’ Hospital, London
Attilio Orazi
Affiliation:
Texas Tech University
Daniel A. Arber
Affiliation:
University of Chicago
Get access

Summary

Myeloid and lymphoid neoplasms with eosinophilia (MLNE) and rearrangements of PDGFRA, PDGFRB and FGFR1 were recognized as a standalone category in the 2008 WHO classification. PCM1-JAK2 was added to this family as a new provisional entity in the 2016 WHO classification [1, 2]. The features shared by neoplasms in this category include a common presentation with eosinophilia or hypereosinophilia in peripheral blood and an increased number of eosinophilic forms in bone marrow (BM). Some cases present as acute leukaemia. Some cases may lack hypereosinophilia. The underlying mechanism is the overexpression of an aberrant tyrosine kinase as a result of a fusion gene, or rarely of a mutation, and a diagnosis and classification requires the demonstration of the specific gene fusions. The cell of origin is a mutated pluripotent stem cell that has the potential to involve myeloid, lymphoid or both lineages, concomitantly or sequentially, leading to clinically complex and heterogeneous manifestations. A common scenario is the presentation as a chronic myeloproliferative neoplasm (MPN), usually with eosinophilia followed within a variable time period and depending on the gene fusion involved, by a progression to acute myeloid leukaemia (AML) or mixed phenotype acute leukaemia (usually in the BM), and B- or T-lymphoblastic leukaemia/lymphoma (B-/T-ALL) in BM or in an extramedullary site. Thus it is critical to recognize the clinicopathologic features of these neoplasms, identify the molecular genetic lesions and classify them accordingly. An accurate diagnosis and classification have important therapeutic and prognostic implications.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2021

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

Bain, BJ, Horny, H-P, Arber, DA, et al. Myeloid/lymphoid neoplasms with eosinophilia and rearrangements of PDGFRA, PDGFRB or FGFR1, or with PCM1-JAK2. In Swerdlow, SH, Campo, E, Harris, NL, et al. (eds). WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues, Revised 4th edn. Lyon: International Agency for Research on Cancer (IARC); 2017:72–9.Google Scholar
Bain, BJ, Ahmad, S. Should myeloid and lymphoid neoplasms with PCM1-JAK2 and other rearrangements of JAK2 be recognized as specific entities? Br J Haematol. 2014;166:809–17.CrossRefGoogle ScholarPubMed
Griffin, JH, Leung, J, Bruner, RJ, Caligiuri, MA, Briesewitz, R. Discovery of a fusion kinase in EOL-1 cells and idiopathic hypereosinophilic syndrome. Proc Natl Acad Sci USA. 2003;100:7830–5.CrossRefGoogle ScholarPubMed
Gleich, GJ, Leiferman, KM, Pardanani, A, Tefferi, A, Butterfield, JH. Treatment of hypereosinophilic syndrome with imatinib mesilate. Lancet. 2002;359:1577–8.CrossRefGoogle ScholarPubMed
Cools, J, DeAngelo, DJ, Gotlib, J, et al. A tyrosine kinase created by fusion of the PDGFRA and FIP1L1 genes as a therapeutic target of imatinib in idiopathic hypereosinophilic syndrome. N Engl J Med. 2003;348:1201–14.Google Scholar
Trempat, P, Villalva, C, Laurent, G, et al. Chronic myeloproliferative disorders with rearrangement of the platelet-derived growth factor alpha receptor: a new clinical target for STI571/Glivec. Oncogene. 2003;22:5702–6.Google Scholar
Montano-Almendras, CP, Essaghir, A, Schoemans, H, et al. ETV6-PDGFRB and FIP1L1-PDGFRA stimulate human hematopoietic progenitor cell proliferation and differentiation into eosinophils: the role of nuclear factor-kappaB. Haematologica. 2012;97:1064–72.CrossRefGoogle ScholarPubMed
Gotlib, J, Cools, J. Five years since the discovery of FIP1L1-PDGFRA: what we have learned about the fusion and other molecularly defined eosinophilias. Leukemia 2008;22:19992010.Google Scholar
Rapanotti, MC, Caruso, R, Ammatuna, E, et al. Molecular characterization of paediatric idiopathic hypereosinophilia. Br J Haematol. 2010;151:440–6.Google Scholar
Safley, AM, Sebastian, S, Collins, TS, et al. Molecular and cytogenetic characterization of a novel translocation t(4;22) involving the breakpoint cluster region and platelet-derived growth factor receptor-alpha genes in a patient with atypical chronic myeloid leukemia. Genes Chromosomes Cancer. 2004;40:4450.CrossRefGoogle Scholar
Vandenberghe, P, Wlodarska, I, Michaux, L, et al. Clinical and molecular features of FIP1L1-PDFGRA (+) chronic eosinophilic leukemias. Leukemia. 2004;18:734–42.Google Scholar
Metzgeroth, G, Walz, C, Score, J, et al. Recurrent finding of the FIP1L1-PDGFRA fusion gene in eosinophilia-associated acute myeloid leukemia and lymphoblastic T-cell lymphoma. Leukemia. 2007;21:1183–8.Google Scholar
Schwaab, J, Jawhar, M, Naumann, N, et al. Diagnostic challenges in the work up of hypereosinophilia: pitfalls in bone marrow core biopsy interpretation. Ann Hematol. 2016;95:557–62.Google Scholar
Chen, D, Bachanova, V, Ketterling, RP, et al. A case of nonleukemic myeloid sarcoma with FIP1L1-PDGFRA rearrangement: an unusual presentation of a rare disease. Am J Surg Pathol. 2013;37:147–51.CrossRefGoogle ScholarPubMed
Yamamoto, M, Ikuta, K, Toki, Y, et al. Angioimmunoblastic T-cell lymphoma and hypereosinophilic syndrome with FIP1L1/PDGFRA fusion gene effectively treated with imatinib: a case report. Medicine. 2017;96:e8001.CrossRefGoogle ScholarPubMed
Score, J, Curtis, C, Waghorn, K, et al. Identification of a novel imatinib responsive KIF5B-PDGFRA fusion gene following screening for PDGFRA overexpression in patients with hypereosinophilia. Leukemia. 2006;20:827–32.CrossRefGoogle ScholarPubMed
Walz, C, Curtis, C, Schnittger, S, et al. Transient response to imatinib in a chronic eosinophilic leukemia associated with ins(9;4)(q33;q12q25) and a CDK5RAP2-PDGFRA fusion gene. Genes Chromosomes Cancer. 2006;45:950–6.Google Scholar
Chalmers, ZR, Ali, SM, Ohgami, RS, et al. Comprehensive genomic profiling identifies a novel TNKS2-PDGFRA fusion that defines a myeloid neoplasm with eosinophilia that responded dramatically to imatinib therapy. Blood Cancer J. 2015;5:e278.CrossRefGoogle ScholarPubMed
Yoshida, M, Tamagawa, N, Nakao, T, et al. Imatinib non-responsive chronic eosinophilic leukemia with ETV6-PDGFRA fusion gene. Leuk Lymphoma. 2015;56:768–9.Google Scholar
Sugimoto, Y, Sada, A, Shimokariya, Y, et al. A novel FOXP1-PDGFRA fusion gene in myeloproliferative neoplasm with eosinophilia. Cancer Genet. 2015;208:508–12.Google Scholar
Curtis, CE, Grand, FH, Musto, P, et al. Two novel imatinib-responsive PDGFRA fusion genes in chronic eosinophilic leukaemia. Br J Haematol. 2007;138:7781.CrossRefGoogle ScholarPubMed
Curtis, CE, Grand, FH, Waghorn, K, et al. A novel ETV6-PDGFRB fusion transcript missed by standard screening in a patient with an imatinib responsive chronic myeloproliferative disease. Leukemia. 2007;21:1839–41.CrossRefGoogle Scholar
Zhou, J, Papenhausen, P, Shao, H. Therapy-related acute myeloid leukemia with eosinophilia, basophilia, t(4;14)(q12;q24) and PDGFRA rearrangement: a case report and review of the literature. Int J Clin Exp Pathol. 2015;8:5812–20.Google Scholar
Baxter, EJ, Hochhaus, A, Bolufer, P, et al. The t(4;22)(q12;q11) in atypical chronic myeloid leukaemia fuses BCR to PDGFRA. Hum Mol Genet. 2002;11:1391–7.Google Scholar
Crescenzi, B, Chase, A, Starza, RL, et al. FIP1L1-PDGFRA in chronic eosinophilic leukemia and BCR-ABL1 in chronic myeloid leukemia affect different leukemic cells. Leukemia. 2007;21:397402.Google Scholar
Yigit, N, Wu, WW, Subramaniyam, S, Mathew, S, Geyer, JT. BCR-PDGFRA fusion in a T lymphoblastic leukemia/lymphoma. Cancer Genet. 2015;208:404–7.Google Scholar
La Starza, R, Specchia, G, Cuneo, A, et al. The hypereosinophilic syndrome: fluorescence in situ hybridization detects the del(4)(q12)-FIP1L1/PDGFRA but not genomic rearrangements of other tyrosine kinases. Haematologica. 2005;90:596601.Google Scholar
Pardanani, A, Ketterling, RP, Li, CY, et al. FIP1L1-PDGFRA in eosinophilic disorders: prevalence in routine clinical practice, long-term experience with imatinib therapy, and a critical review of the literature. Leuk Res. 2006;30:965–70.Google Scholar
Lierman, E, Michaux, L, Beullens, E, et al. FIP1L1-PDGFRalpha D842V, a novel panresistant mutant, emerging after treatment of FIP1L1-PDGFRalpha T674I eosinophilic leukemia with single agent sorafenib. Leukemia. 2009;23:845–51.Google Scholar
Qu, SQ, Qin, TJ, Xu, ZF, et al. Long-term outcomes of imatinib in patients with FIP1L1/PDGFRA associated chronic eosinophilic leukemia: experience of a single center in China. Oncotarget. 2016;7:33229–36.CrossRefGoogle ScholarPubMed
Baccarani, M, Cilloni, D, Rondoni, M, et al. The efficacy of imatinib mesylate in patients with FIP1L1-PDGFRalpha-positive hypereosinophilic syndrome. Results of a multicenter prospective study. Haematologica. 2007;92:1173–9.Google Scholar
Naumann, N, Schwaab, J, Metzgeroth, G, et al. Fusion of PDGFRB to MPRIP, CPSF6, and GOLGB1 in three patients with eosinophilia-associated myeloproliferative neoplasms. Genes Chromosomes Cancer. 2015;54:762–70.Google Scholar
Gosenca, D, Kellert, B, Metzgeroth, G, et al. Identification and functional characterization of imatinib-sensitive DTD1-PDGFRB and CCDC88C-PDGFRB fusion genes in eosinophilia-associated myeloid/lymphoid neoplasms. Genes Chromosomes Cancer. 2014;53:411–21.Google Scholar
Kim, HG, Jang, JH, Koh, EH. TRIP11-PDGFRB fusion in a patient with a therapy-related myeloid neoplasm with t(5;14)(q33;q32) after treatment for acute promyelocytic leukemia. Mol Cytogenet. 2014;7:103.Google Scholar
Walz, C, Haferlach, C, Hanel, A, et al. Identification of a MYO18A-PDGFRB fusion gene in an eosinophilia-associated atypical myeloproliferative neoplasm with a t(5;17)(q33–34;q11.2). Genes Chromosomes Cancer. 2009;48:179–83.Google Scholar
Zou, YS, Hoppman, NL, Singh, ZN, et al. Novel t(5;11)(q32;q13.4) with NUMA1-PDGFRB fusion in a myeloid neoplasm with eosinophilia with response to imatinib mesylate. Cancer Genet. 2017;212–13:3844.Google Scholar
Hidalgo-Curtis, C, Apperley, JF, Stark, A, et al. Fusion of PDGFRB to two distinct loci at 3p21 and a third at 12q13 in imatinib-responsive myeloproliferative neoplasms. Br J Haematol. 2010;148:268–73.CrossRefGoogle Scholar
Hidalgo-Curtis, C, Chase, A, Drachenberg, M, et al. The t(1;9)(p34;q34) and t(8;12)(p11;q15) fuse pre-mRNA processing proteins SFPQ (PSF) and CPSF6 to ABL and FGFR1. Genes Chromosomes Cancer. 2008;47:379–85.Google Scholar
Walz, C, Metzgeroth, G, Haferlach, C, et al. Characterization of three new imatinib-responsive fusion genes in chronic myeloproliferative disorders generated by disruption of the platelet-derived growth factor receptor beta gene. Haematologica. 2007;92:163–9.Google Scholar
Saultz, JN, Kaffenberger, BH, Taylor, M, Heerema, NA, Klisovic, R. Novel chromosome 5 inversion associated with PDGFRB rearrangement in hypereosinophilic syndrome. JAMA Dermatol. 2016;152:1391–3.Google Scholar
Rosati, R, La Starza, R, Luciano, L, et al. TPM3/PDGFRB fusion transcript and its reciprocal in chronic eosinophilic leukemia. Leukemia. 2006;20:1623–4.CrossRefGoogle ScholarPubMed
Wilkinson, K, Velloso, ER, Lopes, LF, et al. Cloning of the t(1;5)(q23;q33) in a myeloproliferative disorder associated with eosinophilia: involvement of PDGFRB and response to imatinib. Blood. 2003;102:4187–90.Google Scholar
Gallagher, G, Horsman, DE, Tsang, P, Forrest, DL. Fusion of PRKG2 and SPTBN1 to the platelet-derived growth factor receptor beta gene (PDGFRB) in imatinib-responsive atypical myeloproliferative disorders. Cancer Genet Cytogenet. 2008;181:4651.Google Scholar
Campregher, PV, Halley, NDS, Vieira, GA, et al. Identification of a novel fusion TBL1XR1-PDGFRB in a patient with acute myeloid leukemia harboring the DEK-NUP214 fusion and clinical response to dasatinib. Leuk Lymphoma. 2017;58:2969–72.Google Scholar
Ross, DM, Altamura, HK, Hahn, CN, et al. Delayed diagnosis leading to accelerated-phase chronic eosinophilic leukemia due to a cytogenetically cryptic, imatinib-responsive TNIP1-PDFGRB fusion gene. Leukemia. 2016;30:1402–5.CrossRefGoogle ScholarPubMed
Winkelmann, N, Hidalgo-Curtis, C, Waghorn, K, et al. Recurrent CEP85L-PDGFRB fusion in patient with t(5;6) and imatinib-responsive myeloproliferative neoplasm with eosinophilia. Leuk Lymphoma. 2013;54:1527–31.Google Scholar
Ross, TS, Bernard, OA, Berger, R, Gilliland, DG. Fusion of Huntingtin interacting protein 1 to platelet-derived growth factor beta receptor (PDGFbetaR) in chronic myelomonocytic leukemia with t(5;7)(q33;q11.2). Blood. 1998;91:4419–26.Google Scholar
Medves, S, Duhoux, FP, Ferrant, A, et al. KANK1, a candidate tumor suppressor gene, is fused to PDGFRB in an imatinib-responsive myeloid neoplasm with severe thrombocythemia. Leukemia. 2010;24:1052–5.CrossRefGoogle Scholar
Schwaller, J, Anastasiadou, E, Cain, D, et al. H4(D10S170), a gene frequently rearranged in papillary thyroid carcinoma, is fused to the platelet-derived growth factor receptor beta gene in atypical chronic myeloid leukemia with t(5;10)(q33;q22). Blood. 2001;97:3910–18.CrossRefGoogle Scholar
Erben, P, Gosenca, D, Muller, MC, et al. Screening for diverse PDGFRA or PDGFRB fusion genes is facilitated by generic quantitative reverse transcriptase polymerase chain reaction analysis. Haematologica. 2010;95:738–44.Google Scholar
Bidet, A, Chollet, C, Gardembas, M, et al. Molecular monitoring of patients with ETV6-PDGFRB rearrangement: implications for therapeutic adaptation. Br J Haematol. 2018;182(1):148–52.Google Scholar
Vizmanos, JL, Novo, FJ, Roman, JP, et al. NIN, a gene encoding a CEP110-like centrosomal protein, is fused to PDGFRB in a patient with a t(5;14)(q33;q24) and an imatinib-responsive myeloproliferative disorder. Cancer Res. 2004;64:2673–6.CrossRefGoogle Scholar
Levine, RL, Wadleigh, M, Sternberg, DW, et al. KIAA1509 is a novel PDGFRB fusion partner in imatinib-responsive myeloproliferative disease associated with a t(5;14)(q33;q32). Leukemia. 2005;19:2730.Google Scholar
Grand, FH, Burgstaller, S, Kuhr, T, et al. p53-Binding protein 1 is fused to the platelet-derived growth factor receptor beta in a patient with a t(5;15)(q33;q22) and an imatinib-responsive eosinophilic myeloproliferative disorder. Cancer Res. 2004;64:7216–19.CrossRefGoogle Scholar
La Starza, R, Rosati, R, Roti, G, et al. A new NDE1/PDGFRB fusion transcript underlying chronic myelomonocytic leukaemia in Noonan Syndrome. Leukemia. 2007;21:830–3.Google Scholar
Magnusson, MK, Meade, KE, Brown, KE, et al. Rabaptin-5 is a novel fusion partner to platelet-derived growth factor beta receptor in chronic myelomonocytic leukemia. Blood. 2001;98:2518–25.Google Scholar
Morerio, C, Acquila, M, Rosanda, C, et al. HCMOGT-1 is a novel fusion partner to PDGFRB in juvenile myelomonocytic leukemia with t(5;17)(q33;p11.2). Cancer Res. 2004;64:2649–51.Google Scholar
Sheng, G, Zeng, Z, Pan, J, et al. Multiple MYO18A-PDGFRB fusion transcripts in a myeloproliferative neoplasm patient with t(5;17)(q32;q11). Mol Cytogenet. 2017;10:4.Google Scholar
Greco, A, Roccato, E, Miranda, C, et al. Growth-inhibitory effect of STI571 on cells transformed by the COL1A1/PDGFB rearrangement. Int J Cancer. 2001;92:354–60.Google Scholar
Patnaik, MM, Lasho, TL, Finke, CM, Pardanani, A, Tefferi, A. Targeted next generation sequencing of PDGFRB rearranged myeloid neoplasms with monocytosis. Am J Hematol. 2016;91:E12–14.Google Scholar
Bell, GC, Padron, E. Detection of a PDGFRB fusion in refractory CMML without eosinophilia: a case for broad spectrum tumor profiling. Leuk Res Rep. 2015;4:70–1.Google Scholar
Schwab, C, Ryan, SL, Chilton, L, et al. EBF1-PDGFRB fusion in pediatric B-cell precursor acute lymphoblastic leukemia (BCP-ALL): genetic profile and clinical implications. Blood. 2016;127:2214–18.Google Scholar
Weston, BW, Hayden, MA, Roberts, KG, et al. Tyrosine kinase inhibitor therapy induces remission in a patient with refractory EBF1-PDGFRB-positive acute lymphoblastic leukemia. J Clin Oncol. 2013;31:e413–16.Google Scholar
Zhang, G, Zhang, Y, Wu, J, Chen, Y, Ma, Z. Acute lymphoblastic leukemia patient with variant ATF7IP/PDGFRB fusion and favorable response to tyrosine kinase inhibitor treatment: a case report. Am J Case Rep. 2017;18:1204–8.CrossRefGoogle ScholarPubMed
Gorello, P, La Starza, R, Brandimarte, L, et al. A PDGFRB-positive acute myeloid malignancy with a new t(5;12)(q33;p13.3) involving the ERC1 gene. Leukemia. 2008;22:216–18.Google Scholar
Gong, SL, Guo, MQ, Tang, GS, et al. Fusion of platelet-derived growth factor receptor beta to CEV14 gene in chronic myelomonocytic leukemia: a case report and review of the literature. Oncol Lett. 2016;11:770–4.Google Scholar
Kobayashi, K, Mitsui, K, Ichikawa, H, et al. ATF7IP as a novel PDGFRB fusion partner in acute lymphoblastic leukaemia in children. Br J Haematol. 2014;165:836–41.Google Scholar
Roberts, KG, Li, Y, Payne-Turner, D, et al. Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia. N Engl J Med. 2014;371:1005–15.Google Scholar
Ondrejka, SL, Jegalian, AG, Kim, AS, et al. PDGFRB-rearranged T-lymphoblastic leukemia/lymphoma occurring with myeloid neoplasms: the missing link supporting a stem cell origin. Haematologica. 2014;99:e148–51.Google Scholar
Arefi, M, Garcia, JL, Penarrubia, MJ, et al. Incidence and clinical characteristics of myeloproliferative neoplasms displaying a PDGFRB rearrangement. Eur J Haematol. 2012;89:3741.Google Scholar
Reiter, A, Gotlib, J. Myeloid neoplasms with eosinophilia. Blood. 2017;129:704–14.Google Scholar
Malfuson, JV, Konopacki, J, Fagot, T, et al. Therapy-related myeloproliferative neoplasm with ETV6-PDGFRB rearrangement following treatment of acute promyelocytic leukemia. Ann Hematol. 2011;90:1477–9.Google Scholar
David, M, Cross, NC, Burgstaller, S, et al. Durable responses to imatinib in patients with PDGFRB fusion gene-positive and BCR-ABL-negative chronic myeloproliferative disorders. Blood. 2007;109:61–4.Google Scholar
Steer, EJ, Cross, NC. Myeloproliferative disorders with translocations of chromosome 5q31–35: role of the platelet-derived growth factor receptor beta. Acta Haematol. 2002;107:113–22.Google Scholar
Lierman, E, Cools, J. TV6 and PDGFRB: a license to fuse. Haematologica. 2007;92:145–7.Google Scholar
Jawhar, M, Naumann, N, Schwaab, J, et al. Imatinib in myeloid/lymphoid neoplasms with eosinophilia and rearrangement of PDGFRB in chronic or blast phase. Ann Hematol. 2017;96:1463–70.Google Scholar
Jawhar, M, Naumann, N, Knut, M, et al. Cytogenetically cryptic ZMYM2-FLT3 and DIAPH1-PDGFRB gene fusions in myeloid neoplasms with eosinophilia. Leukemia. 2017;31:2271–3.Google Scholar
Galimberti, S, Ferreri, MI, Simi, P, et al. Platelet-derived growth factor beta receptor (PDGFRB) gene is rearranged in a significant percentage of myelodysplastic syndromes with normal karyotype. Br J Haematol. 2009;147:763–6.Google Scholar
Cheah, CY, Burbury, K, Apperley, JF, et al. Patients with myeloid malignancies bearing PDGFRB fusion genes achieve durable long-term remissions with imatinib. Blood. 2014;123:3574–7.Google Scholar
Apperley, JF, Gardembas, M, Melo, JV, et al. Response to imatinib mesylate in patients with chronic myeloproliferative diseases with rearrangements of the platelet-derived growth factor receptor beta. N Engl J Med. 2002;347:481–7.Google Scholar
Abruzzo, LV, Jaffe, ES, Cotelingam, JD, et al. T-cell lymphoblastic lymphoma with eosinophilia associated with subsequent myeloid malignancy. Am J Surg Pathol. 1992;16:236–45.CrossRefGoogle ScholarPubMed
Jackson, CC, Medeiros, LJ, Miranda, RN. 8p11 myeloproliferative syndrome: a review. Hum Pathol. 2010;41:461–76.Google Scholar
Vega, F, Medeiros, LJ, Bueso-Ramos, CE, Arboleda, P, Miranda, RN. Hematolymphoid neoplasms associated with rearrangements of PDGFRA, PDGFRB, and FGFR1. Am J Clin Pathol. 2015;144:377–92.Google Scholar
Strati, P, Tang, G, Duose, DY, et al. Myeloid/lymphoid neoplasms with FGFR1 rearrangement. Leuk Lymphoma. 2017:1–5.CrossRefGoogle Scholar
Miranda, RN, Medeiros, LJ. Blastic T/myeloid neoplasm associated with ZMYM2-FGFR1. In Medeiros, LJ, Miranda, RN, (eds) Diagnostic Pathology: Lymph Nodes and Extranodal Lymphomas, 2nd edn. Salt Lake City, UT: Elsevier; 2017:802–11.Google Scholar
Vega, F, Medeiros, LJ, Davuluri, R, et al. t(8;13)-positive bilineal lymphomas: report of 6 cases. Am J Surg Pathol. 2008;32:1420.Google Scholar
Macdonald, D, Aguiar, RC, Mason, PJ, Goldman, JM, Cross, NC. A new myeloproliferative disorder associated with chromosomal translocations involving 8p11: a review. Leukemia. 1995;9:1628–30.Google Scholar
Inhorn, RC, Aster, JC, Roach, SA, et al. A syndrome of lymphoblastic lymphoma, eosinophilia, and myeloid hyperplasia/malignancy associated with t(8;13)(p11;q11): description of a distinctive clinicopathologic entity. Blood. 1995;85:1881–7.CrossRefGoogle Scholar
Kim, WS, Park, SG, Park, G, et al. 8p11 Myeloproliferative syndrome with t(1;8)(q25;p11.2): a case report and review of the literature. Acta Haematol. 2015;133:101–5.Google Scholar
Gervais, C, Dano, L, Perrusson, N, et al. A translocation t(2;8)(q12;p11) fuses FGFR1 to a novel partner gene, RANBP2/NUP358, in a myeloproliferative/myelodysplastic neoplasm. Leukemia. 2013;27:1186–8.CrossRefGoogle Scholar
Soler, G, Nusbaum, S, Varet, B, et al. LRRFIP1, a new FGFR1 partner gene associated with 8p11 myeloproliferative syndrome. Leukemia. 2009;23:1359–61.Google Scholar
Popovici, C, Zhang, B, Gregoire, MJ, et al. The t(6;8)(q27;p11) translocation in a stem cell myeloproliferative disorder fuses a novel gene, FOP, to fibroblast growth factor receptor 1. Blood. 1999;93:1381–9.Google Scholar
Belloni, E, Trubia, M, Gasparini, P, et al. 8p11 Myeloproliferative syndrome with a novel t(7;8) translocation leading to fusion of the FGFR1 and TIF1 genes. Genes Chromosomes Cancer. 2005;42:320–5.Google Scholar
Wasag, B, Lierman, E, Meeus, P, Cools, J, Vandenberghe, P. The kinase inhibitor TKI258 is active against the novel CUX1-FGFR1 fusion detected in a patient with T-lymphoblastic leukemia/lymphoma and t(7;8)(q22;p11). Haematologica. 2011;96:922–6.Google Scholar
Guasch, G, Mack, GJ, Popovici, C, et al. FGFR1 is fused to the centrosome-associated protein CEP110 in the 8p12 stem cell myeloproliferative disorder with t(8;9)(p12;q33). Blood. 2000;95:1788–96.Google Scholar
Sohal, J, Chase, A, Mould, S, et al. Identification of four new translocations involving FGFR1 in myeloid disorders. Genes Chromosomes Cancer. 2001;32:155–63.Google Scholar
Walz, C, Chase, A, Schoch, C, et al. The t(8;17)(p11;q23) in the 8p11 myeloproliferative syndrome fuses MYO18A to FGFR1. Leukemia. 2005;19:1005–9.Google Scholar
Lewis, JP, Jenks, H, Lazerson, J. Philadelphia chromosome-negative chronic myelogenous leukemia in a child with t(8;9)(p11 or 12;q34). Am J Pediatr Hematol Oncol. 1983;5:265–9.Google Scholar
Duckworth, CB, Zhang, L, Li, S. Systemic mastocytosis with associated myeloproliferative neoplasm with t(8;19)(p12;q13.1) and abnormality of FGFR1: report of a unique case. Int J Clin Exp Pathol. 2014;7:801–7.Google Scholar
Montenegro-Garreaud, X, Miranda, RN, Reynolds, A, et al. Myeloproliferative neoplasms with t(8;22)(p11.2;q11.2)/BCR-FGFR1: a meta-analysis of 20 cases shows cytogenetic progression with B-lymphoid blast phase. Hum Pathol. 2017;65:147–56.Google Scholar
Grand, EK, Grand, FH, Chase, AJ, et al. Identification of a novel gene, FGFR1OP2, fused to FGFR1 in 8p11 myeloproliferative syndrome. Genes Chromosomes Cancer. 2004;40:7883.CrossRefGoogle ScholarPubMed
Wang, W, Tang, G, Kadia, T, et al. Cytogenetic evolution associated with disease progression in hematopoietic neoplasms with t(8;22)(p11;q11)/BCR-FGFR1 rearrangement. J Natl Compr Canc Netw. 2016;14:708–11.Google Scholar
Popovici, C, Adelaide, J, Ollendorff, V, et al. Fibroblast growth factor receptor 1 is fused to FIM in stem-cell myeloproliferative disorder with t(8;13). Proc Natl Acad Sci USA. 1998;95:5712–17.Google Scholar
Qiu, XH, Li, F, Cao, HQ, et al. Activity of fibroblast growth factor receptor inhibitors TKI258, ponatinib and AZD4547 against TPRFGFR1 fusion. Mol Med Rep. 2017;15:1024–30.Google Scholar
Reiter, A, Walz, C, Watmore, A, et al. The t(8;9)(p22;p24) is a recurrent abnormality in chronic and acute leukemia that fuses PCM1 to JAK2. Cancer Res. 2005;65:2662–7.Google Scholar
He, R, Greipp, PT, Rangan, A, et al. BCR-JAK2 fusion in a myeloproliferative neoplasm with associated eosinophilia. Cancer Genet. 2016;209:223–8.Google Scholar
Rumi, E, Milosevic, JD, Selleslag, D, et al. Efficacy of ruxolitinib in myeloid neoplasms with PCM1-JAK2 fusion gene. Ann Hematol. 2015;94:1927–8.Google Scholar
Catovsky, D, Bernasconi, C, Verdonck, PJ, et al. The association of eosinophilia with lymphoblastic leukaemia or lymphoma: a study of seven patients. Br J Haematol. 1980;45:523–34.Google Scholar
Bank, I, Amariglio, N, Reshef, A, et al. The hypereosinophilic syndrome associated with CD4+CD3-helper type 2 (Th2) lymphocytes. Leuk Lymphoma. 2001;42:123–33.Google Scholar
Walker, S, Wang, C, Walradt, T, et al. Identification of a gain-of-function STAT3 mutation (p.Y640F) in lymphocytic variant hypereosinophilic syndrome. Blood. 2016;127:948–51.Google Scholar
Salas-Coronas, J, Cabezas-Fernandez, MT, Vazquez-Villegas, J, et al. Evaluation of eosinophilia in immigrants in Southern Spain using tailored screening and treatment protocols: a prospective study. Travel Med Infect Dis. 2015;13:315–21.Google Scholar
Wang, SA, Tam, W, Tsai, AG, et al. Targeted next-generation sequencing identifies a subset of idiopathic hypereosinophilic syndrome with features similar to chronic eosinophilic leukemia, not otherwise specified. Mod Pathol. 2016;29:854–64.Google Scholar
Wang, SA, Hasserjian, RP, Tam, W, et al. Bone marrow morphology is a strong discriminator between chronic eosinophilic leukemia, not otherwise specified and reactive idiopathic hypereosinophilic syndrome. Haematologica. 2017;102:1352–60.Google Scholar
Pardanani, A, Lim, KH, Lasho, TL, et al. Prognostically relevant breakdown of 123 patients with systemic mastocytosis associated with other myeloid malignancies. Blood. 2009;114:3769–72.CrossRefGoogle ScholarPubMed
Schmitt-Graeff, AH, Erben, P, Schwaab, J, et al. The FIP1L1-PDGFRA fusion gene and the KIT D816V mutation are coexisting in a small subset of myeloid/lymphoid neoplasms with eosinophilia. Blood. 2014;123:595–7.Google Scholar
Roberts, KG. The biology of Philadelphia chromosome-like ALL. Best Pract Res Clin Haematol. 2017;30:212–21.Google Scholar
Tasian, SK, Loh, ML, Hunger, SP. Philadelphia chromosome-like acute lymphoblastic leukemia. Blood. 2017;130:2064–72.Google Scholar
Roberts, KG, Gu, Z, Payne-Turner, D, et al. High frequency and poor outcome of Philadelphia chromosome-like acute lymphoblastic leukemia in adults. J Clin Oncol. 2017;35:394401.Google Scholar
Jain, N, Roberts, KG, Jabbour, E, et al. Ph-like acute lymphoblastic leukemia: a high-risk subtype in adults. Blood. 2017;129:572–81.Google Scholar
Reshmi, SC, Harvey, RC, Roberts, KG, et al. Targetable kinase gene fusions in high-risk B-ALL: a study from the Children’s Oncology Group. Blood. 2017;129:3352–61.Google Scholar
Poitras, JL, Dal Cin, P, Aster, JC, Deangelo, DJ, Morton, CC. Novel SSBP2-JAK2 fusion gene resulting from a t(5;9)(q14.1;p24.1) in pre-B acute lymphocytic leukemia. Genes Chromosomes Cancer. 2008;47:884–9.Google Scholar
Chase, A, Bryant, C, Score, J, et al. Ruxolitinib as potential targeted therapy for patients with JAK2 rearrangements. Haematologica. 2013;98:404–8.CrossRefGoogle ScholarPubMed
Su, RJ, Jonas, BA, Welborn, J, Gregg, JP, Chen, M. Chronic eosinophilic leukemia, NOS with t(5;12)(q31;p13)/ETV6-ACSL6 gene fusion: a novel variant of myeloid proliferative neoplasm with eosinophilia. Hum Pathol. 2016;5:69.Google Scholar
De Luca-Johnson, J, Ninfea, JI, Pearson, L, et al. Myeloid neoplasms with t(5;12) and ETV6-ACSL6 gene fusion, potential mimickers of myeloid neoplasm with PDGFRB rearrangement: case report with imatinib therapy and review of the literature. Case Rep Med. 2016;2016:8324791.Google Scholar
Chung, A, Hou, Y, Ohgami, RS, et al. A novel TRIP11-FLT3 fusion in a patient with a myeloid/lymphoid neoplasm with eosinophilia. Cancer Genet. 2017;216–217:1015.Google Scholar
Borrow, J, Stanton, VP Jr., Andresen, JM, et al. The translocation t(8;16)(p11;p13) of acute myeloid leukaemia fuses a putative acetyltransferase to the CREB-binding protein. Nat Genet. 1996;14:3341.Google Scholar
Carapeti, M, Aguiar, RC, Goldman, JM, Cross, NC. A novel fusion between MOZ and the nuclear receptor coactivator TIF2 in acute myeloid leukemia. Blood. 1998;91:3127–33.Google Scholar
Kitabayashi, I, Aikawa, Y, Yokoyama, A, et al. Fusion of MOZ and p300 histone acetyltransferases in acute monocytic leukemia with a t(8;22)(p11;q13) chromosome translocation. Leukemia. 2001;15:8994.Google Scholar
Murati, A, Adelaide, J, Gelsi-Boyer, V, et al. t(5;12)(q23–31;p13) with ETV6-ACSL6 gene fusion in polycythemia vera. Leukemia. 2006;20:1175–8.Google Scholar
Bain, BJ, Gilliland, DG, Horny, H-P, Vardiman, JW. Myeloid and lymphoid neoplasms with eosinophilia and abnormalities of PDGFRA, PDGFRB or FGFR1. In Swerdlow, SH, Campo, E, Harris, NL, et al. (eds) WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. Lyon: International Agency for Research on Cancer (IARC); 2008:6873.Google Scholar
Crane, MM, Chang, CM, Kobayashi, MG, Weller, PF. Incidence of myeloproliferative hypereosinophilic syndrome in the United States and an estimate of all hypereosinophilic syndrome incidence. J Allergy Clin Immunol. 2010;126:179–81.Google Scholar
Helbig, G, Soja, A, Bartkowska-Chrobok, A, Kyrcz-Krzemien, S. Chronic eosinophilic leukemia-not otherwise specified has a poor prognosis with unresponsiveness to conventional treatment and high risk of acute transformation. Am J Hematol. 2012;87:643–5.Google Scholar
Weide, R, Rieder, H, Mehraein, Y, et al. Chronic eosinophilic leukaemia (CEL): a distinct myeloproliferative disease. Br J Haematol. 1997;96:117–23.Google Scholar
Ogbogu, PU, Bochner, BS, Butterfield, JH, et al. Hypereosinophilic syndrome: a multicenter, retrospective analysis of clinical characteristics and response to therapy. J Allergy Clin Immunol. 2009;124:1319–25 e3.Google Scholar
Lefebvre, C, Bletry, O, Degoulet, P, et al. [Prognostic factors of hypereosinophilic syndrome. Study of 40 cases]. Ann Med Interne (Paris). 1989;140:253–7.Google Scholar
Khoury, P, Makiya, M, Klion, AD. Clinical and biological markers in hypereosinophilic syndromes. Front Med (Lausanne). 2017;4:240.Google Scholar
Simon, HU, Plotz, SG, Dummer, R, Blaser, K. Abnormal clones of T cells producing interleukin-5 in idiopathic eosinophilia. N Engl J Med. 1999;341:1112–20.Google Scholar
Bain, BJ, Gilliland, DG, Vardiman, JW, Horny, H-P. Chronic eosinophilic leukemia, not otherwise specified. In Swerdlow, SH, Campo, E, Lee Harris, N, et al. (eds) WHO Classification of Tumors of Haematopoietic and Lymphoid Tissues: Lyon: IARC Press; 2008:51–3.Google Scholar
Bain, BJ, Horny, HP, Hasserjian, RP, Orazi, A. Chronic Eosinophilic Leukaemia, NOS. Lyons: IARC Press; 2017.Google Scholar
Schwaab, J, Umbach, R, Metzgeroth, G, et al. KIT D816V and JAK2 V617F mutations are seen recurrently in hypereosinophilia of unknown significance. Am J Hematol. 2015;90:774–7.Google Scholar
Bacher, U, Reiter, A, Haferlach, T, et al. A combination of cytomorphology, cytogenetic analysis, fluorescence in situ hybridization and reverse transcriptase polymerase chain reaction for establishing clonality in cases of persisting hypereosinophilia. Haematologica. 2006;91:817–20.Google Scholar
Lee, JS, Seo, H, Im, K, et al. Idiopathic hypereosinophilia is clonal disorder? Clonality identified by targeted sequencing. PLoS One. 2017;12:e0185602.Google Scholar
Cross, NCP, Hoade, Y, Tapper, WJ, et al. Recurrent activating STAT5B N642H mutation in myeloid neoplasms with eosinophilia. Leukemia. 2018.Google Scholar
Cargo, CA, Rowbotham, N, Evans, PA, et al. Targeted sequencing identifies patients with preclinical MDS at high risk of disease progression. Blood. 2015;126:2362–5.Google Scholar
Steensma, DP, Bejar, R, Jaiswal, S, et al. Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood. 2015;126:916.Google Scholar
Carruthers, MN, Park, S, Slack, GW, et al. IgG4-related disease and lymphocyte-variant hypereosinophilic syndrome: a comparative case series. Eur J Haematol. 2017;98:378–87.Google Scholar
Bain, BJ, Gilliland, DG, Horny, H-P, Vardiman, JW. Myeloid and lymphoid neoplasms with eosinophilia and abnormalities of PDGFRA, PDGFRB or FGFR1. In Swerdlow, SH, Campo, E, Harris, NL, et al. (eds) WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. Lyon: International Agency for Research on Cancer (IARC); 2008:6873.Google Scholar
Crane, MM, Chang, CM, Kobayashi, MG, Weller, PF. Incidence of myeloproliferative hypereosinophilic syndrome in the United States and an estimate of all hypereosinophilic syndrome incidence. J Allergy Clin Immunol. 2010;126:179–81.Google Scholar
Helbig, G, Soja, A, Bartkowska-Chrobok, A, Kyrcz-Krzemien, S. Chronic eosinophilic leukemia-not otherwise specified has a poor prognosis with unresponsiveness to conventional treatment and high risk of acute transformation. Am J Hematol. 2012;87:643–5.Google Scholar
Weide, R, Rieder, H, Mehraein, Y, et al. Chronic eosinophilic leukaemia (CEL): a distinct myeloproliferative disease. Br J Haematol. 1997;96:117–23.Google Scholar
Ogbogu, PU, Bochner, BS, Butterfield, JH, et al. Hypereosinophilic syndrome: a multicenter, retrospective analysis of clinical characteristics and response to therapy. J Allergy Clin Immunol. 2009;124:1319–25 e3.Google Scholar
Lefebvre, C, Bletry, O, Degoulet, P, et al. [Prognostic factors of hypereosinophilic syndrome. Study of 40 cases]. Ann Med Interne (Paris). 1989;140:253–7.Google Scholar
Khoury, P, Makiya, M, Klion, AD. Clinical and biological markers in hypereosinophilic syndromes. Front Med (Lausanne). 2017;4:240.Google Scholar
Simon, HU, Plotz, SG, Dummer, R, Blaser, K. Abnormal clones of T cells producing interleukin-5 in idiopathic eosinophilia. N Engl J Med. 1999;341:1112–20.Google Scholar
Bain, BJ, Gilliland, DG, Vardiman, JW, Horny, H-P. Chronic eosinophilic leukemia, not otherwise specified. In Swerdlow, SH, Campo, E, Lee Harris, N, et al. (eds) WHO Classification of Tumors of Haematopoietic and Lymphoid Tissues: Lyon: IARC Press; 2008:51–3.Google Scholar
Bain, BJ, Horny, HP, Hasserjian, RP, Orazi, A. Chronic Eosinophilic Leukaemia, NOS. Lyons: IARC Press; 2017.Google Scholar
Schwaab, J, Umbach, R, Metzgeroth, G, et al. KIT D816V and JAK2 V617F mutations are seen recurrently in hypereosinophilia of unknown significance. Am J Hematol. 2015;90:774–7.Google Scholar
Bacher, U, Reiter, A, Haferlach, T, et al. A combination of cytomorphology, cytogenetic analysis, fluorescence in situ hybridization and reverse transcriptase polymerase chain reaction for establishing clonality in cases of persisting hypereosinophilia. Haematologica. 2006;91:817–20.Google Scholar
Lee, JS, Seo, H, Im, K, et al. Idiopathic hypereosinophilia is clonal disorder? Clonality identified by targeted sequencing. PLoS One. 2017;12:e0185602.Google Scholar
Cross, NCP, Hoade, Y, Tapper, WJ, et al. Recurrent activating STAT5B N642H mutation in myeloid neoplasms with eosinophilia. Leukemia. 2018.Google Scholar
Cargo, CA, Rowbotham, N, Evans, PA, et al. Targeted sequencing identifies patients with preclinical MDS at high risk of disease progression. Blood. 2015;126:2362–5.Google Scholar
Steensma, DP, Bejar, R, Jaiswal, S, et al. Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood. 2015;126:916.Google Scholar
Carruthers, MN, Park, S, Slack, GW, et al. IgG4-related disease and lymphocyte-variant hypereosinophilic syndrome: a comparative case series. Eur J Haematol. 2017;98:378–87.Google Scholar
Bain, BJ, Gilliland, DG, Horny, H-P, Vardiman, JW. Myeloid and lymphoid neoplasms with eosinophilia and abnormalities of PDGFRA, PDGFRB or FGFR1. In Swerdlow, SH, Campo, E, Harris, NL, et al. (eds) WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. Lyon: International Agency for Research on Cancer (IARC); 2008:6873.Google Scholar
Crane, MM, Chang, CM, Kobayashi, MG, Weller, PF. Incidence of myeloproliferative hypereosinophilic syndrome in the United States and an estimate of all hypereosinophilic syndrome incidence. J Allergy Clin Immunol. 2010;126:179–81.Google Scholar
Helbig, G, Soja, A, Bartkowska-Chrobok, A, Kyrcz-Krzemien, S. Chronic eosinophilic leukemia-not otherwise specified has a poor prognosis with unresponsiveness to conventional treatment and high risk of acute transformation. Am J Hematol. 2012;87:643–5.Google Scholar
Weide, R, Rieder, H, Mehraein, Y, et al. Chronic eosinophilic leukaemia (CEL): a distinct myeloproliferative disease. Br J Haematol. 1997;96:117–23.Google Scholar
Ogbogu, PU, Bochner, BS, Butterfield, JH, et al. Hypereosinophilic syndrome: a multicenter, retrospective analysis of clinical characteristics and response to therapy. J Allergy Clin Immunol. 2009;124:1319–25 e3.Google Scholar
Lefebvre, C, Bletry, O, Degoulet, P, et al. [Prognostic factors of hypereosinophilic syndrome. Study of 40 cases]. Ann Med Interne (Paris). 1989;140:253–7.Google Scholar
Khoury, P, Makiya, M, Klion, AD. Clinical and biological markers in hypereosinophilic syndromes. Front Med (Lausanne). 2017;4:240.Google Scholar
Simon, HU, Plotz, SG, Dummer, R, Blaser, K. Abnormal clones of T cells producing interleukin-5 in idiopathic eosinophilia. N Engl J Med. 1999;341:1112–20.Google Scholar
Bain, BJ, Gilliland, DG, Vardiman, JW, Horny, H-P. Chronic eosinophilic leukemia, not otherwise specified. In Swerdlow, SH, Campo, E, Lee Harris, N, et al. (eds) WHO Classification of Tumors of Haematopoietic and Lymphoid Tissues: Lyon: IARC Press; 2008:51–3.Google Scholar
Bain, BJ, Horny, HP, Hasserjian, RP, Orazi, A. Chronic Eosinophilic Leukaemia, NOS. Lyons: IARC Press; 2017.Google Scholar
Schwaab, J, Umbach, R, Metzgeroth, G, et al. KIT D816V and JAK2 V617F mutations are seen recurrently in hypereosinophilia of unknown significance. Am J Hematol. 2015;90:774–7.Google Scholar
Bacher, U, Reiter, A, Haferlach, T, et al. A combination of cytomorphology, cytogenetic analysis, fluorescence in situ hybridization and reverse transcriptase polymerase chain reaction for establishing clonality in cases of persisting hypereosinophilia. Haematologica. 2006;91:817–20.Google Scholar
Lee, JS, Seo, H, Im, K, et al. Idiopathic hypereosinophilia is clonal disorder? Clonality identified by targeted sequencing. PLoS One. 2017;12:e0185602.Google Scholar
Cross, NCP, Hoade, Y, Tapper, WJ, et al. Recurrent activating STAT5B N642H mutation in myeloid neoplasms with eosinophilia. Leukemia. 2018.Google Scholar
Cargo, CA, Rowbotham, N, Evans, PA, et al. Targeted sequencing identifies patients with preclinical MDS at high risk of disease progression. Blood. 2015;126:2362–5.Google Scholar
Steensma, DP, Bejar, R, Jaiswal, S, et al. Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood. 2015;126:916.Google Scholar
Carruthers, MN, Park, S, Slack, GW, et al. IgG4-related disease and lymphocyte-variant hypereosinophilic syndrome: a comparative case series. Eur J Haematol. 2017;98:378–87.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×