Skip to main content Accessibility help
×
Hostname: page-component-7c8c6479df-ws8qp Total loading time: 0 Render date: 2024-03-29T11:14:05.273Z Has data issue: false hasContentIssue false

75 - EMS: the 8p11 myeloproliferative syndrome

from Part 3.6 - Molecular pathology: lymphoma and leukemia

Published online by Cambridge University Press:  05 February 2015

Donald H. C. Macdonald
Affiliation:
Department of Haematology, Imperial College, London, UK
Andreas Reiter
Affiliation:
Medizinische Klinik, Univers¨atsmedizin Mannheim, Germany
Nicholas C. P. Cross
Affiliation:
Wessex Regional Genetics Laboratory, University of Southampton, Salisbury District Hospital, Salisbury, UK
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Introduction

The “8p11 myeloproliferative syndrome” (EMS) is an extremely rare hematological malignancy characterized by disruption and constitutive activation of fibroblast growth-factor receptor type 1 (FGFR1; 1). The disease is also referred to as “stem-cell leukemia/lymphoma syndrome” (SCLL) or “myeloid and lymphoid neoplasms with FGFR1 abnormalities” (ICD-O code 9967/3; 2,3). Clinically, EMS is typically a biphenotypic disorder that may present as a myeloproliferative neoplasm, acute leukemia or lymphoblastic lymphoma, usually in conjunction with prominent eosinophilia. Although uncommon, EMS is of interest because of its stem-cell origin, marked genotype/phenotype correlations and diverse range of FGFR1 fusions, which all demonstrate a common pathogenic mechanism. Cell-line and animal studies have dissected the signaling pathways that are critical for transformation and may ultimately lead to molecularly targeted therapy.

Clinical features

Clinical and laboratory descriptions of more than 40 cases of EMS have been published and, although the clinical course is highly variable, some common features have emerged, as summarized in Table 75.1. The age range at onset is between 5 months and 84 years, with a median of 32 and a slight male to female predominance of 1.5:1. EMS may present as a myeloid and/or lymphoid malignancy; the myeloid presentation may be either a myeloproliferative neoplasm (MPN) or acute myeloid leukemia (AML), and the lymphoid presentation is typically either B-cell acute lymphoblastic leukemia (B-ALL) or T-cell lymphoblastic lymphoma (T-LBL).

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 809 - 817
Publisher: Cambridge University Press
Print publication year: 2013

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

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 ScholarPubMed
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.Google Scholar
Swerdlow, S, Campo, E, Harris, N, et al. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues, fourth edn. Lyon: IARC; 2008.Google Scholar
Goradia, A, Bayerl, M, Cornfield, D. The 8p11 myeloproliferative syndrome: review of literature and an illustrative case report. International Journal of Clinical and Experimental Pathology 2008;1:448–56.Google ScholarPubMed
Macdonald, D, Reiter, A, Cross, NC. The 8p11 myeloproliferative syndrome: a distinct clinical entity caused by constitutive activation of FGFR1. Acta Haematologica 2002;107:101–7.CrossRefGoogle ScholarPubMed
Vega, F, Medeiros, LJ, Davuluri, R, et al. t(8;13)-positive bilineal lymphomas: report of 6 cases. American Journal of Surgical Pathology 2008;32:14–20.CrossRefGoogle 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 and Cancer 2005;42:320–5.CrossRefGoogle Scholar
Anderson, MK, Hernandez-Hoyos, G, Diamond, RA, Rothenberg, EV. Precise developmental regulation of Ets family transcription factors during specification and commitment to the T cell lineage. Development 1999;126:3131–48.Google ScholarPubMed
Cross, NC, Reiter, A. Fibroblast growth factor receptor and platelet-derived growth factor receptor abnormalities in eosinophilic myeloproliferative disorders. Acta Haematologica 2008;119:199–206.CrossRefGoogle ScholarPubMed
Aguiar, RC, Macdonald, D, Mason, PJ, Cross, NC, Goldman, JM. Myeloproliferative disorder associated with 8p11 translocations. Blood 1995;86:834–5.Google ScholarPubMed
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. Nature Genetics 1996;14:33–41.CrossRefGoogle Scholar
Chaffanet, M, Mozziconacci, MJ, Fernandez, F, et al. A case of inv(8)(p11q24) associated with acute myeloid leukemia involves the MOZ and CBP genes in a masked t(8;16). Genes, Chromosomes and Cancer 1999;26:161–5.3.0.CO;2-6>CrossRefGoogle Scholar
Carapeti, M, Aguiar, RC, Watmore, AE, Goldman, JM, Cross, NC. Consistent fusion of MOZ and TIF2 in AML with inv(8)(p11q13). Cancer Genetics and Cytogenetics 1999;113:70–2.CrossRefGoogle Scholar
Agerstam, H, Lilljebjorn, H, Lassen, C, et al. Fusion gene-mediated truncation of RUNX1 as a potential mechanism underlying disease progression in the 8p11 myeloproliferative syndrome. Genes, Chromosomes and Cancer 2007;46:635–43.CrossRefGoogle ScholarPubMed
Macdonald, D, Cross, NC. Chronic myeloproliferative disorders: the role of tyrosine kinases in pathogenesis, diagnosis and therapy. Pathobiology 2007;74:81–8.CrossRefGoogle Scholar
Mason, JM, Morrison, DJ, Basson, MA, Licht, JD. Sprouty proteins: multifaceted negative-feedback regulators of receptor tyrosine kinase signaling. Trends in Cell Biology 2006;16:45–54.CrossRefGoogle ScholarPubMed
Ren, M, Cowell, JK. Constitutive Notch pathway activation in murine ZMYM2-FGFR1-induced T-cell lymphomas associated with atypical myeloproliferative disease. Blood 2011;117:6837–47.CrossRefGoogle ScholarPubMed
Demiroglu, A, Steer, EJ, Heath, C, et al. The t(8;22) in chronic myeloid leukemia fuses BCR to FGFR1: transforming activity and specific inhibition of FGFR1 fusion proteins. Blood 2001;98:3778–83.CrossRefGoogle Scholar
Murati, A, Arnoulet, C, Lafage-Pochitaloff, M, et al. Dual lympho-myeloproliferative disorder in a patient with t(8;22) with BCR-FGFR1 gene fusion. International Journal of Oncology 2005;26:1485–92.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
Vizmanos, JL, Hernandez, R, Vidal, MJ, et al. Clinical variability of patients with the t(6;8)(q27;p12) and FGFR1OP-FGFR1 fusion: two further cases. Hematology Journal 2004;5:534–7.CrossRefGoogle Scholar
Wakim, JJ, Tirado, CA, Chen, W, Collins, R. t(8;22)/BCR-FGFR1 myeloproliferative disorder presenting as B-acute lymphoblastic leukemia: report of a case treated with sorafenib and review of the literature. Leukemia Research 2011;35:e151–3.CrossRefGoogle Scholar
Baldazzi, C, Iacobucci, I, Luatti, S, et al. B-cell acute lymphoblastic leukemia as evolution of a 8p11 myeloproliferative syndrome with t(8;22)(p11;q11) and BCR-FGFR1 fusion gene. Leukemia Research 2010;34:e282–5.CrossRefGoogle Scholar
Kim, SY, Oh, B, She, CJ, et al. 8p11 Myeloproliferative syndrome with BCR-FGFR1 rearrangement presenting with T-lymphoblastic lymphoma and bone marrow stromal cell proliferation: a case report and review of the literature. Leukemia Research 2011;35:e30–4.CrossRefGoogle ScholarPubMed
Delaval, B, Lelievre, H, Birnbaum, D. Myeloproliferative disorders: the centrosome connection. Leukemia 2005;19:1739–44.CrossRefGoogle ScholarPubMed
Delaval, B, Letard, S, Lelievre, H, et al. Oncogenic tyrosine kinase of malignant hemopathy targets the centrosome. Cancer Research 2005;65:7231–40.CrossRefGoogle ScholarPubMed
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 and Cancer 2008;47:379–85.CrossRefGoogle Scholar
Ren, M, Qin, H, Ren, R, Tidwell, J, Cowell, JK. Src activation plays an important key role in lymphomagenesis induced by FGFR1 fusion kinases. Cancer Research 2011;71:7312–22.CrossRefGoogle ScholarPubMed
Gu, TL, Goss, VL, Reeves, C, et al. Phosphotyrosine profiling identifies the KG-1 cell line as a model for the study of FGFR1 fusions in acute myeloid leukemia. Blood 2006;108:4202–4.CrossRefGoogle Scholar
Heath, C, Cross, NC. Critical role of STAT5 activation in transformation mediated by ZNF198-FGFR1. Journal of Biological Chemistry 2004;279:6666–73.CrossRefGoogle ScholarPubMed
Dong, S, Kang, S, Gu, TL, et al. 14-3-3 Integrates prosurvival signals mediated by the AKT and MAPK pathways in ZNF198-FGFR1-transformed hematopoietic cells. Blood 2007;110:360–9.CrossRefGoogle ScholarPubMed
Kouhara, H, Hadari, YR, Spivak-Kroizman, T, et al. A lipid-anchored Grb2-binding protein that links FGF-receptor activation to the Ras/MAPK signaling pathway. Cell 1997;89:693–702.CrossRefGoogle ScholarPubMed
Roumiantsev, S, Krause, DS, Neumann, CA, et al. Distinct stem cell myeloproliferative/T lymphoma syndromes induced by ZNF198-FGFR1 and BCR-FGFR1 fusion genes from 8p11 translocations. Cancer Cell 2004;5:287–98.CrossRefGoogle ScholarPubMed
Agerstam, H, Jaras, M, Andersson, A, et al. Modeling the human 8p11-myeloproliferative syndrome in immunodeficient mice. Blood 2010;116:2103–11.CrossRefGoogle ScholarPubMed
Ren, M, Li, X, Cowell, JK. Genetic fingerprinting of the development and progression of T-cell lymphoma in a murine model of atypical myeloproliferative disorder initiated by the ZNF198-fibroblast growth factor receptor-1 chimeric tyrosine kinase. Blood 2009;114:1576–84.CrossRefGoogle Scholar
Chase, A, Grand, FH, Cross, NC. Activity of TKI258 against primary cells and cell lines with FGFR1 fusion genes associated with the 8p11 myeloproliferative syndrome. Blood 2007;110:3729–34.CrossRefGoogle ScholarPubMed
Chen, J, Deangelo, DJ, Kutok, JL, et al. PKC412 inhibits the zinc finger 198-fibroblast growth factor receptor 1 fusion tyrosine kinase and is active in treatment of stem cell myeloproliferative disorder. Proceedings of the National Academy of Sciences USA 2004;101:14 479–84.CrossRefGoogle ScholarPubMed
Chase, A, Bryant, C, Score, J, Cross, NC. Ponatinib as targeted therapy for FGFR1 fusions associated with the 8p11 myeloproliferative syndrome. Haematologica 2013;98:103–6.CrossRefGoogle ScholarPubMed
Lierman, E, Smits, S, Cools, J, et al. Ponatinib is active against imatinib-resistant mutants of FIP1L1-PDGFRA and KIT, and against FGFR1-derived fusion kinases. Leukemia 2012;26:1693–5.CrossRefGoogle ScholarPubMed
Ren, M, Qin, H, Ren, R, Cowell, JK. Ponatinib suppresses the development of myeloid and lymphoid malignancies associated with FGFR1 abnormalities. Leukemia 2012;27:32–40.CrossRefGoogle ScholarPubMed
Soler, G, Nusbaum, S, Varet, B, et al. LRRFIP1, a new FGFR1 partner gene associated with 8p11 myeloproliferative syndrome. Leukemia 2009;23:1359–61.CrossRefGoogle ScholarPubMed
Lourenco, GJ, Ortega, MM, Freitas, LL, et al. The rare t(6;8; q27;p11) translocation in a case of chronic myeloid neoplasm mimicking polycythemia vera. Leukemia and Lymphoma 2008;49:1832–5.CrossRefGoogle Scholar
Vannier, JP, Bizet, M, Bastard, C, et al. Simultaneous occurrence of a T-cell lymphoma and a chronic myelogenous leukemia with an unusual karyotype. Leukemia Research 1984;8:647–57.CrossRefGoogle Scholar
Sohal, J, Chase, A, Mould, S, et al. Identification of four new translocations involving FGFR1 in myeloid disorders. Genes, Chromosomes and Cancer 2001;32:155–63.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle Scholar
Mozziconacci, MJ, Carbuccia, N, Prebet, T, et al. Common features of myeloproliferative disorders with t(8;9)(p12;q33) and CEP110-FGFR1 fusion: report of a new case and review of the literature. Leukemia Research 2008;32:1304–8.CrossRefGoogle Scholar
Friedhoff, F, Rajendra, B, Moody, R, Alapatt, T. Novel reciprocal translocation between chromosomes 8 and 9 found in a patient with myeloproliferative disorder. Cancer Genetics and Cytogenetics 1983;9:391–4.CrossRefGoogle Scholar
Lewis, JP, Jenks, H, Lazerson, J. Philadelphia chromosome-negative chronic myelogenous leukemia in a child with t(8;9)(p11 or 12;q34). American Journal of Pediatric Hematology and Oncology 1983;5:265–9.Google Scholar
Oscier, DG, Mufti, GJ, Gardiner, A, Hamblin, TJ. Reciprocal translocation between chromosomes 8 and 9 in atypical chronic myeloid leukaemia. Journal of Medical Genetics 1985;22:398–401.CrossRefGoogle ScholarPubMed
Jotterand Bellomo, M, Muhlematter, D, Wicht, M, Delacretaz, F, Schmidt, PM. t(8;9)(p11;q32) in atypical chronic myeloid leukaemia: a new cytogenetic-clinicopathologic association? British Journal of Haematology 1992;81:307–8.CrossRefGoogle Scholar
Nakayama, H, Inamitsu, T, Ohga, S, et al. Chronic myelomonocytic leukaemia with t(8;9)(p11;q34) in childhood: an example of the 8p11 myeloproliferative disorder? British Journal of Haematology 1996;92:692–5.CrossRefGoogle Scholar
van den Berg, H, Kroes, W, van der Schoot, CE, et al. A young child with acquired t(8;9)(p11;q34): additional proof that 8p11 is involved in mixed myeloid/T lymphoid malignancies. Leukemia 1996;10:1252–3.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
Yamamoto, K, Kawano, H, Nishikawa, S, et al. A biphenotypic transformation of 8p11 myeloproliferative syndrome with CEP1/FGFR1 fusion gene. European Journal of Haematology 2006;77:349–54.CrossRefGoogle ScholarPubMed
Grand, EK, Grand, FH, Chase, AJ, et al. Identification of a novel gene, FGFR1OP2, fused to FGFR1 in 8p11 myeloproliferative syndrome. Genes, Chromosomes and Cancer 2004;40:78–83.CrossRefGoogle ScholarPubMed
Leslie, J, Barker, T, Glancy, M, Jennings, B, Pearson, J. t(8;13; p11;q12) translocation in a myeloproliferative disorder associated with a T-cell non-Hodgkin lymphoma. British Journal of Haematology 1994;86:876–8.CrossRefGoogle Scholar
Fagan, K, Hyde, S, Harrison, P. Translocation (8;13) and T-cell lymphoma: a case report. Cancer Genetics and Cytogenetics 1993;65:71–3.CrossRefGoogle ScholarPubMed
Abruzzo, LV, Jaffe, ES, Cotelingam, JD, et al. T-cell lymphoblastic lymphoma with eosinophilia associated with subsequent myeloid malignancy. American Journal of Surgical Pathology 1992;16:236–45.CrossRefGoogle ScholarPubMed
Still, IH, Chernova, O, Hurd, D, Stone, RM, Cowell, JK. Molecular characterization of the t(8;13)(p11;q12) translocation associated with an atypical myeloproliferative disorder: evidence for three discrete loci involved in myeloid leukemias on 8p11. Blood 1997;90:3136–41.Google Scholar
Chernova, O, Still, I, Kalaycio, M, Hoeltge, G, Cowell, JK. Characterization of the breakpoints in a t(8;13)(p11;q12) translocation from a patient with myeloproliferative disease using fluorescence in situ hybridization. Genes, Chromosomes and Cancer 1998;21:160–5.3.0.CO;2-V>CrossRefGoogle Scholar
Chaffanet, M, Popovici, C, Leroux, D, et al. t(6;8), t(8;9) and t(8;13) translocations associated with stem cell myeloproliferative disorders have close or identical breakpoints in chromosome region 8p11–12. Oncogene 1998;16:945–9.CrossRefGoogle ScholarPubMed
Michaux, L, Mecucci, C, Pereira Velloso, ER, et al. About the t(8;13)(p11;q12) clinico-pathologic entity. Blood 1996;87:1658–9.Google Scholar
Matsumoto, K, Morita, K, Takada, S, et al. A chronic myelogenous leukemia-like myeloproliferative disorder accompanied by T-cell lymphoblastic lymphoma with chromosome translocation t(8;13)(p11;q12): a Japanese case. International Journal of Hematology 1999;70:278–82.Google Scholar
JabbarAl-Obaidi, M, Rymes, N, White, P, et al. A fourth case of 8p11 myeloproliferative disorder transforming to B-lineage acute lymphoblastic leukaemia: a case report. Acta Haematologica 2002;107:98–100.CrossRefGoogle ScholarPubMed
Aguiar, RC, Chase, A, Coulthard, S, et al. Abnormalities of chromosome band 8p11 in leukemia: two clinical syndromes can be distinguished on the basis of MOZ involvement. Blood 1997;90:3130–5.Google ScholarPubMed
Wong, WS, Cheng, KC, Lau, KM, et al. Clonal evolution of 8p11 stem cell syndrome in a 14-year-old Chinese boy: a review of literature of t(8;13) associated myeloproliferative diseases. Leukemia Research 2007;31:235–8.CrossRefGoogle Scholar
Invernizzi, R, Benatti, C, Travaglino, E, et al. A further case of myeloproliferative syndrome with reciprocal translocation (8;13)(p11;q12). Haematologica 2004;89:239–41.Google Scholar
Sahin, F, Sercan, Z, Ertan, Y, et al. Rapid transformation of atypical myeloproliferative disorder with consistent t(8;13) to B-cell acute lymphoblastic leukemia: a case report. Hematology 2007;12:489–92.CrossRefGoogle Scholar
Rao, PH, Cesarman, G, Coleman, M, Acaron, S, Verma, RS. Cytogenetic evidence for extramedullary blast crisis with t(8;13)(q11;p11) in chronic myelomonocytic leukemia. Acta Haematologica 1992;88:201–3.CrossRefGoogle 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.CrossRefGoogle Scholar
Mugneret, F, Chaffanet, M, Maynadie, M, et al. The 8p12 myeloproliferative disorder. t(8;19)(p12;q13.3): a novel translocation involving the FGFR1 gene. British Journal of Haematology 2000;111:647–9.CrossRefGoogle Scholar
Pini, M, Gottardi, E, Scaravaglio, P, et al. A fourth case of BCR-FGFR1 positive CML-like disease with t(8;22) translocation showing an extensive deletion on the derivative chromosome 8p. Hematology Journal 2002;3:315–16.CrossRefGoogle Scholar
Fioretos, T, Panagopoulos, I, Lassen, C, et al. Fusion of the BCR and the fibroblast growth factor receptor-1 (FGFR1) genes as a result of t(8;22)(p11;q11) in a myeloproliferative disorder: the first fusion gene involving BCR but not ABL. Genes, Chromosomes and Cancer 2001;32:302–10.CrossRefGoogle 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
×