Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-wg55d Total loading time: 0 Render date: 2024-05-16T11:08:42.681Z Has data issue: false hasContentIssue false

12 - Myeloproliferative neoplasms

from Section 2 - Neoplastic hematopathology

Published online by Cambridge University Press:  03 May 2011

Robert B. Lorsbach
Affiliation:
University of Arkansas for Medical Sciences
Maria A. Proytcheva
Affiliation:
Northwestern University Medical School, Illinois
Get access

Summary

Introduction

Although hematologic malignancies are collectively the most common neoplasms of childhood, comprising approximately one-fourth to one-third of all cases of pediatric cancer in the USA, the myeloproliferative neoplasms, formerly termed chronic myeloproliferative disorders, are distinctly rare in the pediatric setting [1, 2]. Among the seven entities recognized by the 2008 WHO classification of myeloproliferative neoplasms (Table 12.1), chronic myelogenous leukemia and mastocytosis are the only ones encountered with any regularity in the pediatric population. Primary myelofibrosis, polycythemia vera, and essential thrombocythemia are very uncommon in children, and chronic neutrophilic leukemia and chronic eosinophilic leukemia are extraordinarily rare in this population, the description of which is limited to an occasional case report [3]. Several pediatric “myeloproliferative” disorders are not truly neoplastic, and as such they have a pathogenesis distinct from their bona fide neoplastic counterparts in adults. The application to these pediatric disorders of the same terminology employed for adult myeloproliferative neoplasms, which are by definition clonal and neoplastic, creates confusion with regard to their pathogenesis, clinical behavior, and appropriate treatment. Our discussion of pediatric myeloproliferative neoplasms will focus almost exclusively on those disorders seen in children, with emphasis on the distinctive aspects of the pediatric forms of these disorders, as well as conditions which can mimic true pediatric myeloproliferative neoplasms. Myeloproliferative neoplasms which are exceedingly rare in children (e.g., chronic neutrophilic leukemia) will not be discussed.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2011

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

Ries, LAG, Smith, MA, Gurney, JG, et al. (eds.). Cancer Incidence and Survival Among Children and Adolescents: United States SEER Program 1975–1995, NIH Pub. No. 99–4649. Bethesda, MD: National Cancer Institute, SEER Program; 1999.Google Scholar
Coebergh, JW, Reedijk, AM, Vries, E, et al. Leukaemia incidence and survival in children and adolescents in Europe during 1978–1997. Report from the Automated Childhood Cancer Information System project. European Journal of Cancer. 2006;42:2019–2036.CrossRefGoogle ScholarPubMed
Hasle, H, Olesen, G, Kerndrup, G, Philip, P, Jacobsen, N. Chronic neutrophil leukaemia in adolescence and young adulthood. British Journal of Haematology. 1996;94:628–630.CrossRefGoogle ScholarPubMed
Bennett, JH. Case of hypertrophy of the spleen and liver in which death took place from suppuration of the blood. Edinburgh Medical and Surgical Journal. 1845;64:413–423.Google Scholar
Nowell, PC, Hungerford, DA. A minute chromosome in human chronic granulocytic leukemia. Science. 1960;132:1497.Google Scholar
Rowley, JD. A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining. Nature. 1973;243:290–293.CrossRefGoogle ScholarPubMed
Shtivelman, E, Lifshitz, B, Gale, RP, Canaani, E. Fused transcript of abl and bcr genes in chronic myelogenous leukaemia. Nature. 1985;315:550–554.CrossRefGoogle ScholarPubMed
Grosveld, G, Verwoerd, T, van Agthoven, T, et al. The chronic myelocytic cell line K562 contains a breakpoint in bcr and produces a chimeric bcr/c-abl transcript. Molecular and Cellular Biology. 1986;6:607–616.CrossRefGoogle ScholarPubMed
Druker, BJ, Talpaz, M, Resta, DJ, et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. The New England Journal of Medicine. 2001;344:1031–1037.CrossRefGoogle ScholarPubMed
Hervy, M, Hoffman, L, Beckerle, MC. From the membrane to the nucleus and back again: bifunctional focal adhesion proteins. Current Opinion in Cell Biology. 2006;18:524–532.CrossRefGoogle ScholarPubMed
Pendergast, AM. The Abl family kinases: mechanisms of regulation and signaling. Advances in Cancer Research. 2002;85:51–100.CrossRefGoogle ScholarPubMed
Schwartzberg, PL, Stall, AM, Hardin, JD, et al. Mice homozygous for the ablm1 mutation show poor viability and depletion of selected B and T cell populations. Cell. 1991;65:1165–1175.CrossRefGoogle ScholarPubMed
Tybulewicz, VL, Crawford, CE, Jackson, PK, Bronson, RT, Mulligan, RC. Neonatal lethality and lymphopenia in mice with a homozygous disruption of the c-abl proto-oncogene. Cell. 1991;65:1153–1163.CrossRefGoogle ScholarPubMed
Dorsch, M, Goff, SP. Increased sensitivity to apoptotic stimuli in c-abl-deficient progenitor B-cell lines. Proceedings of the National Academy of Sciences of the United States of America. 1996;93:13131–13136.CrossRefGoogle ScholarPubMed
Zipfel, PA, Grove, M, Blackburn, K, et al. The c-Abl tyrosine kinase is regulated downstream of the B cell antigen receptor and interacts with CD19. Journal of Immunology. 2000;165:6872–6879.CrossRefGoogle Scholar
Cho, YJ, Cunnick, JM, Yi, SJ, et al. Abr and Bcr, two homologous Rac GTPase-activating proteins, control multiple cellular functions of murine macrophages. Molecular and Cellular Biology. 2007;27:899–911.CrossRefGoogle ScholarPubMed
Daley, GQ, Etten, RA, Baltimore, D. Induction of chronic myelogenous leukemia in mice by the P210bcr/abl gene of the Philadelphia chromosome. Science. 1990;247:824–830.CrossRefGoogle ScholarPubMed
Pear, WS, Miller, JP, Xu, L, et al. Efficient and rapid induction of a chronic myelogenous leukemia-like myeloproliferative disease in mice receiving P210 bcr/abl-transduced bone marrow. Blood. 1998;92:3780–3792.Google ScholarPubMed
Zhang, X, Ren, R. Bcr-Abl efficiently induces a myeloproliferative disease and production of excess interleukin-3 and granulocyte-macrophage colony-stimulating factor in mice: a novel model for chronic myelogenous leukemia. Blood. 1998;92:3829–3840.Google ScholarPubMed
Pane, F, Intrieri, M, Quintarelli, C, et al. BCR/ABL genes and leukemic phenotype: from molecular mechanisms to clinical correlations 1. Oncogene. 2002;21:8652–8667.CrossRefGoogle Scholar
Goldman, JM, Melo, JV. Chronic myeloid leukemia – advances in biology and new approaches to treatment. The New England Journal of Medicine. 2003;349:1451–1464.CrossRefGoogle Scholar
Pane, F, Frigeri, F, Sindona, M, et al. Neutrophilic-chronic myeloid leukemia: a distinct disease with a specific molecular marker (BCR/ABL with C3/A2 junction). Blood. 1996;88:2410–2414.Google Scholar
Wilson, G, Frost, L, Goodeve, A, et al. BCR-ABL transcript with an e19a2 (c3a2) junction in classical chronic myeloid leukemia. Blood. 1997;89:3064.Google ScholarPubMed
Mittre, H, Leymarie, P, Macro, M, Leporrier, M. A new case of chronic myeloid leukemia with c3/a2 BCR/ABL junction. Is it really a distinct disease? Blood. 1997;89:4239–4241.Google ScholarPubMed
Melo, JV, Barnes, DJ. Chronic myeloid leukaemia as a model of disease evolution in human cancer. Nature Reviews. Cancer. 2007;7:441–453.CrossRefGoogle ScholarPubMed
Mejia-Arangure, JM, Bonilla, M, Lorenzana, R, et al. Incidence of leukemias in children from El Salvador and Mexico City between 1996 and 2000: population-based data. BMC Cancer. 2005;5:33.CrossRefGoogle ScholarPubMed
Castro-Malaspina, H, Schaison, G, Briere, J, et al. Philadelphia chromosome-positive chronic myelocytic leukemia in children. Survival and prognostic factors. Cancer. 1983;52:721–727.3.0.CO;2-X>CrossRefGoogle ScholarPubMed
Chang, YH, Lu, MY, Jou, ST, et al. Forty-seven children suffering from chronic myeloid leukemia at a center over a 25-year period. Pediatric Hematology and Oncology. 2003;20:505–515.CrossRefGoogle Scholar
Millot, F, Traore, P, Guilhot, J, et al. Clinical and biological features at diagnosis in 40 children with chronic myeloid leukemia. Pediatrics 2005;116:140–143.CrossRefGoogle ScholarPubMed
Dow, LW, Raimondi, SC, Culbert, SJ, et al. Response to alpha-interferon in children with Philadelphia chromosome-positive chronic myelocytic leukemia. Cancer. 1991;68:1678–1684.3.0.CO;2-J>CrossRefGoogle ScholarPubMed
Fioretos, T, Heim, S, Garwicz, S, Ludvigsson, J, Mitelman, F. Molecular analysis of Philadelphia-positive childhood chronic myeloid leukemia. Leukemia. 1992;6:723–725.Google ScholarPubMed
Kantarjian, HM, Keating, MJ, Talpaz, M, et al. Chronic myelogenous leukemia in blast crisis. Analysis of 242 patients. The American Journal of Medicine. 1987;83:445–454.CrossRefGoogle ScholarPubMed
Cervantes, F, Villamor, N, Esteve, J, et al. ‘Lymphoid’ blast crisis of chronic myeloid leukaemia is associated with distinct clinicohaematological features. British Journal of Haematology. 1998;100:123–128.CrossRefGoogle ScholarPubMed
Khalidi, HS, Brynes, RK, Medeiros, LJ, et al. The immunophenotype of blast transformation of chronic myelogenous leukemia: a high frequency of mixed lineage phenotype in “lymphoid” blasts and a comparison of morphologic, immunophenotypic, and molecular findings. Modern Pathology. 1998;11:1211–1221.Google Scholar
Lorand-Metze, I, Vassallo, J, Souza, CA. Histological and cytological heterogeneity of bone marrow in Philadelphia-positive chronic myelogenous leukaemia at diagnosis. British Journal of Haematology. 1987;67:45–49.CrossRefGoogle ScholarPubMed
Dickstein, JI, Vardiman, JW. Hematopathologic findings in the myeloproliferative disorders. Seminars in Oncology. 1995;22:355–373.Google ScholarPubMed
Thiele, J, Kvasnicka, HM, Schmitt-Graeff, A, et al. Bone marrow features and clinical findings in chronic myeloid leukemia – a comparative, multicenter, immunohistological and morphometric study on 614 patients. Leukemia & Lymphoma. 2000;36:295–308.CrossRefGoogle ScholarPubMed
Albrecht, M. “Gaucher-Zellen” bei chronisch myeloischer Leukamie. Blut. 1966;13:169–179.CrossRefGoogle Scholar
Dosik, H, Rosner, F, Sawitsky, A. Acquired lipidosis: Gaucher-like cells and “blue cells” in chronic granulocytic leukemia. Seminars in Hematology. 1972;9:309–316.Google Scholar
Kelsey, PR, Geary, CG. Sea-blue histiocytes and Gaucher cells in bone marrow of patients with chronic myeloid leukaemia. Journal of Clinical Pathology. 1988;41:960–962.CrossRefGoogle ScholarPubMed
Anastasi, J, Musvee, T, Roulston, D, et al. Pseudo-Gaucher histiocytes identified up to 1 year after transplantation for CML are BCR/ABL-positive. Leukemia 1998;12:233–237.CrossRefGoogle ScholarPubMed
Kantarjian, HM, Bueso-Ramos, CE, Talpaz, M, et al. Significance of myelofibrosis in early chronic-phase, chronic myelogenous leukemia on imatinib mesylate therapy. Cancer. 2005;104:777–780.CrossRefGoogle ScholarPubMed
Buesche, G, Ganser, A, Schlegelberger, B, et al. Marrow fibrosis and its relevance during imatinib treatment of chronic myeloid leukemia. Leukemia. 2007;21:2420–2427.CrossRefGoogle ScholarPubMed
Beham-Schmid, C, Apfelbeck, U, Sill, H, et al. Treatment of chronic myelogenous leukemia with the tyrosine kinase inhibitor STI571 results in marked regression of bone marrow fibrosis. Blood 2002;99:381–383.CrossRefGoogle ScholarPubMed
Braziel, RM, Launder, TM, Druker, BJ, et al. Hematopathologic and cytogenetic findings in imatinib mesylate-treated chronic myelogenous leukemia patients: 14 months' experience. Blood 2002;100:435–441.CrossRefGoogle ScholarPubMed
Hasserjian, RP, Boecklin, F, Parker, S, et al. ST1571 (imatinib mesylate) reduces bone marrow cellularity and normalizes morphologic features irrespective of cytogenetic response. American Journal of Clinical Pathology. 2002;117:360–367.CrossRefGoogle ScholarPubMed
Frater, JL, Tallman, MS, Variakojis, D, et al. Chronic myeloid leukemia following therapy with imatinib mesylate (Gleevec). Bone marrow histopathology and correlation with genetic status. American Journal of Clinical Pathology. 2003;119:833–841.CrossRefGoogle ScholarPubMed
Thiele, J, Kvasnicka, HM, Schmitt-Graeff, A, et al. Bone marrow changes in chronic myelogenous leukaemia after long-term treatment with the tyrosine kinase inhibitor STI571: an immunohistochemical study on 75 patients. Histopathology. 2005;46:540–550.CrossRefGoogle ScholarPubMed
Lokeshwar, N, Kumar, L, Kumari, M. Severe bone marrow aplasia following imatinib mesylate in a patient with chronic myelogenous leukemia. Leukemia & Lymphoma. 2005;46:781–784.CrossRefGoogle Scholar
Srinivas, U, Pillai, LS, Kumar, R, Pati, HP, Saxena, R. Bone marrow aplasia – a rare complication of imatinib therapy in CML patients. American Journal of Hematology. 2007;82:314–316.CrossRefGoogle ScholarPubMed
Bain, B, Catovsky, D, O'Brien, M, Spiers, AS, Richards, GH. Megakaryoblastic transformation of chronic granulocytic leukaemia. An electron microscopy and cytochemical study. Journal of Clinical Pathology. 1977;30:235–242.CrossRefGoogle Scholar
Ekblom, M, Borgström, G, Willebrand, E, et al. Erythroid blast crisis in chronic myelogenous leukemia. Blood. 1983;62:591–596.Google ScholarPubMed
Villeval, JL, Cramer, P, Lemoine, F, et al. Phenotype of early erythroblastic leukemias. Blood. 1986;68:1167–1174.Google ScholarPubMed
Hernandez, JM, Gonzalez-Sarmiento, R, Martin, C, et al. Immunophenotypic, genomic and clinical characteristics of blast crisis of chronic myelogenous leukaemia. British Journal of Haematology. 1991;79:408–414.CrossRefGoogle ScholarPubMed
Urbano-Ispizua, A, Cervantes, F, Matutes, E, et al. Immunophenotypic characteristics of blast crisis of chronic myeloid leukaemia: correlations with clinico-biological features and survival. Leukemia. 1993;7:1349–1354.Google Scholar
Kosugi, N, Tojo, A, Shinzaki, H, Nagamura-Inoue, T, Asano, S. The preferential expression of CD7 and CD34 in myeloid blast crisis in chronic myeloid leukemia. Blood. 2000;95:2188–2189.Google ScholarPubMed
Kantarjian, H, O'Brien, S, Cortes, J, et al. Sudden onset of the blastic phase of chronic myelogenous leukemia: patterns and implications. Cancer. 2003;98:81–85.CrossRefGoogle ScholarPubMed
Morimoto, A, Ogami, A, Chiyonobu, T, et al. Early blastic transformation following complete cytogenetic response in a pediatric chronic myeloid leukemia patient treated with imatinib mesylate. Journal of Pediatric Hematology/Oncology. 2004;26:320–322.CrossRefGoogle Scholar
Avery, S, Nadal, E, Marin, D, et al. Lymphoid transformation in a CML patient in complete cytogenetic remission following treatment with imatinib. Leukemia Research. 2004;28(Suppl 1): S75–S77.CrossRefGoogle Scholar
Alimena, G, Breccia, M, Latagliata, R, et al. Sudden blast crisis in patients with Philadelphia chromosome-positive chronic myeloid leukemia who achieved complete cytogenetic remission after imatinib therapy. Cancer. 2006;107:1008–1013.CrossRefGoogle ScholarPubMed
Jabbour, E, Kantarjian, H, O'Brien, S, et al. Sudden blastic transformation in patients with chronic myeloid leukemia treated with imatinib mesylate. Blood. 2006;107:480–482.CrossRefGoogle ScholarPubMed
Kim, AS, Goldstein, SC, Luger, S, Deerlin, VM, Bagg, A. Sudden extramedullary T-lymphoblastic blast crisis in chronic myelogenous leukemia: a nonrandom event associated with imatinib?American Journal of Clinical Pathology. 2008;129:639–648.CrossRefGoogle ScholarPubMed
Ganesan, TS, Rassool, F, Guo, AP, et al. Rearrangement of the bcr gene in Philadelphia chromosome-negative chronic myeloid leukemia. Blood 1986;68:957–960.Google ScholarPubMed
Shtalrid, M, Talpaz, M, Blick, M, et al. Philadelphia-negative chronic myelogenous leukemia with breakpoint cluster region rearrangement: molecular analysis, clinical characteristics, and response to therapy. Journal of Clinical Oncology. 1988;6:1569–1575.CrossRefGoogle ScholarPubMed
Morel, F, Herry, A, Le Bris, MJ, et al. Contribution of fluorescence in situ hybridization analyses to the characterization of masked and complex Philadelphia chromosome translocations in chronic myelocytic leukemia. Cancer Genetics and Cytogenetics. 2003;147:115–120.CrossRefGoogle Scholar
Aoun, P, Wiggins, M, Pickering, D, et al. Interphase fluorescence in situ hybridization studies for the detection of 9q34 deletions in chronic myelogenous leukemia: a practical approach to clinical diagnosis. Cancer Genetics and Cytogenetics. 2004;154:138–143.CrossRefGoogle ScholarPubMed
Costa, D, Carrio, A, Madrigal, I, et al. Studies of complex Ph translocations in cases with chronic myelogenous leukemia and one with acute lymphoblastic leukemia. Cancer Genetics and Cytogenetics. 2006;166:89–93.CrossRefGoogle ScholarPubMed
Hughes, T, Deininger, M, Hochhaus, A, et al. Monitoring CML patients responding to treatment with tyrosine kinase inhibitors: review and recommendations for harmonizing current methodology for detecting BCR-ABL transcripts and kinase domain mutations and for expressing results. Blood. 2006;108:28–37.CrossRefGoogle ScholarPubMed
Baccarani, M, Saglio, G, Goldman, J, et al. Evolving concepts in the management of chronic myeloid leukemia: recommendations from an expert panel on behalf of the European LeukemiaNet. Blood. 2006;108:1809–1820.CrossRefGoogle Scholar
Kantarjian, H, Schiffer, C, Jones, D, Cortes, J. Monitoring the response and course of chronic myeloid leukemia in the modern era of BCR-ABL tyrosine kinase inhibitors: practical advice on the use and interpretation of monitoring methods. Blood. 2008;111:1774–1780.CrossRefGoogle ScholarPubMed
Derderian, PM, Kantarjian, HM, Talpaz, M, et al. Chronic myelogenous leukemia in the lymphoid blastic phase: characteristics, treatment response, and prognosis. The American Journal of Medicine. 1993;94:69–74.CrossRefGoogle ScholarPubMed
Johansson, B, Fioretos, T, Mitelman, F. Cytogenetic and molecular genetic evolution of chronic myeloid leukemia. Acta Haematologica. 2002;107:76–94.CrossRefGoogle ScholarPubMed
Castaigne, S, Berger, R, Jolly, V, et al. Promyelocytic blast crisis of chronic myelocytic leukemia with both t(9;22) and t(15;17) in M3 cells. Cancer. 1984;54:2409–2413.3.0.CO;2-V>CrossRefGoogle Scholar
Hogge, , Misawa, S, Schiffer, CA, Testa, JR. Promyelocytic blast crisis in chronic granulocytic leukemia with 15;17 translocation. Leukemia Research. 1984;8:1019–1023.CrossRefGoogle ScholarPubMed
Yin, CC, Medeiros, LJ, Glassman, AB, Lin, P. t(8;21)(q22;q22) in blast phase of chronic myelogenous leukemia. American Journal of Clinical Pathology. 2004;121:836–842.CrossRefGoogle Scholar
Wu, Y, Slovak, ML, Snyder, DS, Arber, DA. Coexistence of inversion 16 and the Philadelphia chromosome in acute and chronic myeloid leukemias: report of six cases and review of literature. American Journal of Clinical Pathology. 2006;125:260–266.CrossRefGoogle ScholarPubMed
Merzianu, M, Medeiros, LJ, Cortes, J, et al. inv(16)(p13q22) in chronic myelogenous leukemia in blast phase: a clinicopathologic, cytogenetic, and molecular study of five cases. American Journal of Clinical Pathology. 2005;124:807–814.CrossRefGoogle ScholarPubMed
Chan, KW, Kaikov, Y, Wadsworth, LD. Thrombocytosis in childhood: a survey of 94 patients. Pediatrics. 1989;84:1064–1067.Google ScholarPubMed
Heath, HW, Pearson, HA. Thrombocytosis in pediatric outpatients. The Journal of Pediatrics. 1989;114:805–807.CrossRefGoogle ScholarPubMed
Vora, AJ, Lilleyman, JS. Secondary thrombocytosis. Archives of Disease in Childhood. 1993;68:88–90.CrossRefGoogle ScholarPubMed
Yohannan, MD, Higgy, KE, al-Mashhadani, SA, Santhosh-Kumar, CR. Thrombocytosis. Etiologic analysis of 663 patients. Clinical Pediatrics. 1994;33:340–343.CrossRefGoogle ScholarPubMed
Heng, JT, Tan, AM. Thrombocytosis in childhood. Singapore Medical Journal. 1998;39:485–487.Google ScholarPubMed
Chen, HL, Chiou, SS, Sheen, JM, et al. Thrombocytosis in children at one medical center of southern Taiwan. Acta Paediatrica Taiwanica. 1999;40:309–313.Google ScholarPubMed
Matsubara, K, Fukaya, T, Nigami, H, et al. Age-dependent changes in the incidence and etiology of childhood thrombocytosis. Acta Haematologica. 2004;111:132–137.CrossRefGoogle ScholarPubMed
Hasle, H. Incidence of essential thrombocythaemia in children. British Journal of Haematology. 2000;110:751.CrossRefGoogle ScholarPubMed
Jensen, MK, de Nully, BP, Nielsen, OJ, Hasselbalch, HC. Incidence, clinical features and outcome of essential thrombocythaemia in a well defined geographical area. European Journal of Haematology. 2000;65:132–139.CrossRefGoogle Scholar
Dame, C, Sutor, AH. Primary and secondary thrombocytosis in childhood. British Journal of Haematology. 2005;129:165–177.CrossRefGoogle ScholarPubMed
Gassas, A, Doyle, JJ, Weitzman, S, et al. A basic classification and a comprehensive examination of pediatric myeloproliferative syndromes. Journal of Pediatric Hematology/Oncology. 2005;27:192–196.CrossRefGoogle Scholar
Choi, ES, Nichol, JL, Hokom, MM, Hornkohl, AC, Hunt, P. Platelets generated in vitro from proplatelet-displaying human megakaryocytes are functional. Blood. 1995;85:402–413.Google ScholarPubMed
Cramer, EM, Norol, F, Guichard, J, et al. Ultrastructure of platelet formation by human megakaryocytes cultured with the Mpl ligand. Blood. 1997;89:2336–2346.Google ScholarPubMed
Junt, T, Schulze, H, Chen, Z, et al. Dynamic visualization of thrombopoiesis within bone marrow. Science. 2007;317:1767–1770.CrossRefGoogle ScholarPubMed
Stuart, MJ, Murphy, S, Oski, FA. A simple nonradioisotope technic for the determination of platelet life-span. The New England Journal of Medicine. 1975;292:1310–1313.CrossRefGoogle ScholarPubMed
Sinzinger, H, Fitscha, P, Peskar, BA. Platelet half-life, plasma thromboxane B2 and circulating endothelial cells in peripheral vascular disease. Angiology. 1986;37:112–118.CrossRefGoogle ScholarPubMed
Fritz, E, Ludwig, H, Scheithauer, W, Sinzinger, H. Shortened platelet half-life in multiple myeloma. Blood. 1986;68:514–520.Google ScholarPubMed
Shivdasani, RA. Molecular and transcriptional regulation of megakaryocyte differentiation. Stem Cells. 2001;19:397–407.CrossRefGoogle ScholarPubMed
Deutsch, VR, Tomer, A. Megakaryocyte development and platelet production. British Journal of Haematology. 2006;134:453–466.CrossRefGoogle ScholarPubMed
Goldfarb, AN. Transcriptional control of megakaryocyte development. Oncogene. 2007;26:6795–6802.CrossRefGoogle ScholarPubMed
Ballmaier, M, Germeshausen, M, Schulze, H, et al. c-mpl mutations are the cause of congenital amegakaryocytic thrombocytopenia. Blood 2001;97:139–146.CrossRefGoogle ScholarPubMed
Ihara, K, Ishii, E, Eguchi, M, et al. Identification of mutations in the c-mpl gene in congenital amegakaryocytic thrombocytopenia. Proceedings of the National Academy of Sciences of the United States of America. 1999;96:3132–3136.CrossRefGoogle ScholarPubMed
Mehaffey, MG, Newton, AL, Gandhi, MJ, Crossley, M, Drachman, JG. X-linked thrombocytopenia caused by a novel mutation of GATA-1. Blood. 2001;98:2681–2688.CrossRefGoogle ScholarPubMed
Nichols, KE, Crispino, JD, Poncz, M, et al. Familial dyserythropoietic anaemia and thrombocytopenia due to an inherited mutation in GATA1. Nature Genetics. 2000;24:266–270.CrossRefGoogle Scholar
Song, WJ, Sullivan, MG, Legare, RD, et al. Haploinsufficiency of CBFA2 causes familial thrombocytopenia with propensity to develop acute myelogenous leukaemia. Nature Genetics. 1999;23:166–175.CrossRefGoogle ScholarPubMed
Tonelli, R, Scardovi, AL, Pession, A, et al. Compound heterozygosity for two different amino-acid substitution mutations in the thrombopoietin receptor (c-mpl gene) in congenital amegakaryocytic thrombocytopenia (CAMT). Human Genetics. 2000;107:225–233.CrossRefGoogle Scholar
Oudenrijn, S, Bruin, M, Folman, CC, et al. Mutations in the thrombopoietin receptor, Mpl, in children with congenital amegakaryocytic thrombocytopenia. British Journal of Haematology. 2000;110:441–448.CrossRefGoogle ScholarPubMed
Kondo, T, Okabe, M, Sanada, M, et al. Familial essential thrombocythemia associated with one-base deletion in the 5′-untranslated region of the thrombopoietin gene. Blood. 1998;92:1091–1096.Google ScholarPubMed
Wiestner, A, Schlemper, RJ, Maas, AP, Skoda, RC. An activating splice donor mutation in the thrombopoietin gene causes hereditary thrombocythaemia. Nature Genetics. 1998;18:49–52.CrossRefGoogle ScholarPubMed
Ghilardi, N, Wiestner, A, Kikuchi, M, Ohsaka, A, Skoda, RC. Hereditary thrombocythaemia in a Japanese family is caused by a novel point mutation in the thrombopoietin gene. British Journal of Haematology. 1999;107:310–316.CrossRefGoogle Scholar
Ding, J, Komatsu, H, Wakita, A, et al. Familial essential thrombocythemia associated with a dominant-positive activating mutation of the c-MPL gene, which encodes for the receptor for thrombopoietin. Blood. 2004;103:4198–4200.CrossRefGoogle ScholarPubMed
Onishi, M, Mui, AL, Morikawa, Y, et al. Identification of an oncogenic form of the thrombopoietin receptor MPL using retrovirus-mediated gene transfer. Blood. 1996;88:1399–1406.Google ScholarPubMed
Moliterno, AR, Williams, DM, Gutierrez-Alamillo, LI, et al. Mpl Baltimore: a thrombopoietin receptor polymorphism associated with thrombocytosis. Proceedings of the National Academy of Sciences of the United States of America. 2004;101:11444–11447.CrossRefGoogle ScholarPubMed
Tecuceanu, N, Dardik, R, Rabizadeh, E, Raanani, P, Inbal, A. A family with hereditary thrombocythaemia and normal genes for thrombopoietin and c-Mpl. British Journal of Haematology. 2006;135:348–351.CrossRefGoogle ScholarPubMed
Teofili, L, Giona, F, Martini, M, et al. The revised WHO diagnostic criteria for Ph-negative myeloproliferative diseases are not appropriate for the diagnostic screening of childhood polycythemia vera and essential thrombocythemia. Blood. 2007;110:3384–3386.CrossRefGoogle Scholar
Veselovska, J, Pospisilova, D, Pekova, S, et al. Most pediatric patients with essential thrombocythemia show hypersensitivity to erythropoietin in vitro, with rare JAK2 V617F-positive erythroid colonies. Leukemia Research. 2008;32:369–377.CrossRefGoogle ScholarPubMed
Mertens, C, Darnell, JE Jr. SnapShot: JAK-STAT signaling. Cell. 2007;131:612.CrossRefGoogle ScholarPubMed
Tefferi, A. JAK and MPL mutations in myeloid malignancies. Leukemia & Lymphoma. 2008;49:388–397.CrossRefGoogle ScholarPubMed
Neubauer, H, Cumano, A, Muller, M, et al. Jak2 deficiency defines an essential developmental checkpoint in definitive hematopoiesis. Cell. 1998;93:397–409.CrossRefGoogle ScholarPubMed
Parganas, E, Wang, D, Stravopodis, D, et al. Jak2 is essential for signaling through a variety of cytokine receptors. Cell. 1998;93:385–395.CrossRefGoogle ScholarPubMed
James, C, Ugo, V, Couedic, JP, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature. 2005;434:1144–1148.CrossRefGoogle ScholarPubMed
Levine, RL, Wadleigh, M, Cools, J, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 2005;7:387–397.CrossRefGoogle ScholarPubMed
Baxter, EJ, Scott, LM, Campbell, PJ, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet. 2005;365:1054–1061.CrossRefGoogle ScholarPubMed
Kralovics, R, Passamonti, F, Buser, AS, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. The New England Journal of Medicine. 2005;352:1779–1790.CrossRefGoogle ScholarPubMed
Khwaja, A. The role of Janus kinases in haemopoiesis and haematological malignancy. British Journal of Haematology. 2006;134:366–384.CrossRefGoogle ScholarPubMed
Levine, RL, Pardanani, A, Tefferi, A, Gilliland, DG. Role of JAK2 in the pathogenesis and therapy of myeloproliferative disorders. Nature Reviews. Cancer. 2007;7:673–683.CrossRefGoogle ScholarPubMed
El-Moneim, AA, Kratz, CP, Böll, S, et al. Essential versus reactive thrombocythemia in children: retrospective analyses of 12 cases. Pediatric Blood & Cancer. 2007;49:52–55.CrossRefGoogle ScholarPubMed
Randi, ML, Putti, MC, Scapin, M, et al. Pediatric patients with essential thrombocythemia are mostly polyclonal and V617FJAK2 negative. Blood. 2006;108:3600–3602.CrossRefGoogle ScholarPubMed
Teofili, L, Giona, F, Martini, M, et al. Markers of myeloproliferative diseases in childhood polycythemia vera and essential thrombocythemia. Journal of Clinical Oncology. 2007;25:1048–1053.CrossRefGoogle ScholarPubMed
Randi, ML, Putti, MC, Pacquola, E, et al. Normal thrombopoietin and its receptor (c-mpl) genes in children with essential thrombocythemia. Pediatric Blood & Cancer 2005;44:47–50.CrossRefGoogle ScholarPubMed
Roy, NB, Treacy, M, Kench, P. Childhood essential thrombocythaemia. British Journal of Haematology. 2005;129:567.CrossRefGoogle ScholarPubMed
Steensma, DP. JAK2 V617F in myeloid disorders: molecular diagnostic techniques and their clinical utility: a paper from the 2005 William Beaumont Hospital Symposium on Molecular Pathology. The Journal of Molecular Diagnostics. 2006;8:397–411.CrossRefGoogle Scholar
Olsen, RJ, Tang, Z, Farkas, DH, Bernard, DW, Zu, Y, Chang, CC. Detection of the JAK2(V617F) mutation in myeloproliferative disorders by melting curve analysis using the LightCycler system. Archives of Pathology & Laboratory Medicine. 2006;130:997–1003.Google ScholarPubMed
Chapelle, A, Träskelin, AL, Juvonen, E. Truncated erythropoietin receptor causes dominantly inherited benign human erythrocytosis. Proceedings of the National Academy of Sciences of the United States of America. 1993;90:4495–4499.CrossRefGoogle ScholarPubMed
Ang, SO, Chen, H, Hirota, K, et al. Disruption of oxygen homeostasis underlies congenital Chuvash polycythemia. Nature Genetics. 2002;32:614–621.CrossRefGoogle ScholarPubMed
Pastore, YD, Jelinek, J, Ang, S, et al. Mutations in the VHL gene in sporadic apparently congenital polycythemia. Blood. 2003;101:1591–1595.CrossRefGoogle ScholarPubMed
Percy, MJ, Zhao, Q, Flores, A, et al. A family with erythrocytosis establishes a role for prolyl hydroxylase domain protein 2 in oxygen homeostasis. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:654–659.CrossRefGoogle ScholarPubMed
Percy, MJ, Furlow, PW, Beer, PA, et al. A novel erythrocytosis-associated PHD2 mutation suggests the location of a HIF binding groove. Blood. 2007;110:2193–2196.CrossRefGoogle ScholarPubMed
Gordeuk, VR, Stockton, DW, Prchal, JT. Congenital polycythemias/erythrocytoses. Haematologica. 2005;90:109–116.Google ScholarPubMed
Percy, MJ, Lee, FS. Familial erythrocytosis: molecular links to red blood cell control. Haematologica. 2008;93:963–967.CrossRefGoogle ScholarPubMed
Lee, FS. Genetic causes of erythrocytosis and the oxygen-sensing pathway. Blood Reviews. 2008;22:321–332.CrossRefGoogle ScholarPubMed
Park, MJ, Shimada, A, Asada, H, et al. JAK2 mutation in a boy with polycythemia vera, but not in other pediatric hematologic disorders. Leukemia. 2006;20:1453–1454.CrossRefGoogle Scholar
Teofili, L, Foa, R, Giona, F, Larocca, LM. Childhood polycythemia vera and essential thrombocythemia: does their pathogenesis overlap with that of adult patients? Haematologica. 2008;93:169–172.CrossRefGoogle ScholarPubMed
Kvasnicka, HM, Thiele, J. Classification of Ph-negative chronic myeloproliferative disorders – morphology as the yardstick of classification. Pathobiology 2007;74:63–71.CrossRefGoogle ScholarPubMed
Cooperberg, AA, Singer, OP. Reversible myelofibrosis due to vitamin D deficiency rickets. Canadian Medical Association Journal. 1966;94:392–395.Google ScholarPubMed
Evans, DI. Acute myelofibrosis in children with Down's syndrome. Archives of Disease in Childhood. 1975;50:458–462.CrossRefGoogle ScholarPubMed
Schlackman, N, Green, AA, Naiman, JL. Myelofibrosis in children with chronic renal insufficiency. The Journal of Pediatrics. 1975;87:720–724.CrossRefGoogle ScholarPubMed
Ueda, K, Kawaguchi, Y, Kodama, M, et al. Primary myelofibrosis with myeloid metaplasia and cytogenetically abnormal clones in 2 children with Down's syndrome. Scandinavian Journal of Haematology. 1981;27:152–158.CrossRefGoogle ScholarPubMed
Yetgin, S, Ozsoylu, S. Myeloid metaplasia in vitamin D deficiency rickets. Scandinavian Journal of Haematology. 1982;28:180–185.CrossRefGoogle ScholarPubMed
Pantazis, CG, McKie, VC, Sabio, H, Davis, PC, Allsbrook, WC. Down's syndrome and acute myelofibrosis. Time study of DNA content during the progression to leukemia. Cancer. 1988;61:2239–2243.3.0.CO;2-5>CrossRefGoogle Scholar
Balkan, C, Ersoy, B, Nese, N. Myelofibrosis associated with severe vitamin D deficiency rickets. The Journal of International Medical Research. 2005;33:356–359.CrossRefGoogle ScholarPubMed
Mallouh, AA, Sa'di, AR. Agnogenic myeloid metaplasia in children. American Journal of Diseases of Children. 1992;146:965–967.Google ScholarPubMed
Cetingul, N, Yener, E, Oztop, S, Nisli, G, Soydan, S. Agnogenic myeloid metaplasia in childhood: a report of two cases and efficiency of intravenous high dose methylprednisolone treatment. Acta Paediatrica Japonica. 1994;36:697–700.CrossRefGoogle ScholarPubMed
Bonduel, M, Sciuccati, G, Torres, AF, Pierini, A, Gallo, G. Familial idiopathic myelofibrosis and multiple hemangiomas. American Journal of Hematology. 1998;59:175–177.3.0.CO;2-8>CrossRefGoogle ScholarPubMed
Altura, RA, Head, DR, Wang, WC. Long-term survival of infants with idiopathic myelofibrosis. British Journal of Haematology. 2000;109:459–462.CrossRefGoogle ScholarPubMed
Sah, A, Minford, A, Parapia, . Spontaneous remission of juvenile idiopathic myelofibrosis. British Journal of Haematology. 2001;112:1083.CrossRefGoogle ScholarPubMed
Walia, M, Mehta, R, Paul, P, et al. Idiopathic myelofibrosis with generalized periostitis in a 4-year-old girl. Journal of Pediatric Hematology/Oncology. 2005;27:278–282.CrossRefGoogle Scholar
Sekhar, M, Prentice, HG, Popat, U, et al. Idiopathic myelofibrosis in children. British Journal of Haematology. 1996;93:394–397.CrossRefGoogle ScholarPubMed
Sieff, CA, Malleson, P. Familial myelofibrosis. Archives of Disease in Childhood. 1980;55:888–893.CrossRefGoogle ScholarPubMed
Rossbach, HC. Familial infantile myelofibrosis as an autosomal recessive disorder: preponderance among children from Saudi Arabia. Pediatric Hematology and Oncology. 2006;23:453–454.CrossRefGoogle ScholarPubMed
Castro-Malaspina, H, Gay, RE, Jhanwar, SC, et al. Characteristics of bone marrow fibroblast colony-forming cells (CFU-F) and their progeny in patients with myeloproliferative disorders. Blood. 1982;59:1046–1054.Google ScholarPubMed
Jacobson, RJ, Salo, A, Fialkow, PJ. Agnogenic myeloid metaplasia: a clonal proliferation of hematopoietic stem cells with secondary myelofibrosis. Blood. 1978;51:189–194.Google ScholarPubMed
Greenberg, BR, Woo, L, Veomett, IC, Payne, CM, Ahmann, FR. Cytogenetics of bone marrow fibroblastic cells in idiopathic chronic myelofibrosis. British Journal of Haematology. 1987;66:487–490.CrossRefGoogle ScholarPubMed
Lau, SO, Ramsay, NK, Smith, CM, McKenna, R, Kersey, JH. Spontaneous resolution of severe childhood myelofibrosis. The Journal of Pediatrics. 1981;98:585–588.CrossRefGoogle ScholarPubMed
Shankar, S, Choi, JK, Dermody, TS, et al. Pulmonary hypertension complicating bone marrow transplantation for idiopathic myelofibrosis. Journal of Pediatric Hematology/Oncology. 2004;26:393–397.CrossRefGoogle ScholarPubMed
Wallis, JP, Reid, MM. Bone marrow fibrosis in childhood acute lymphoblastic leukaemia. Journal of Clinical Pathology. 1989;42:1253–1254.CrossRefGoogle ScholarPubMed
Noren-Nystrom, U, Roos, G, Bergh, A, et al. Bone marrow fibrosis in childhood acute lymphoblastic leukemia correlates to biological factors, treatment response and outcome. Leukemia. 2008;22:504–510.CrossRefGoogle ScholarPubMed
Stephan, JL, Galambrun, C, Dutour, A, Freycon, F. Myelofibrosis: an unusual presentation of vitamin D-deficient rickets. European Journal of Pediatrics. 1999;158:828–829.Google ScholarPubMed
Gruner, BA, DeNapoli, TS, Elshihabi, S, et al. Anemia and hepatosplenomegaly as presenting features in a child with rickets and secondary myelofibrosis. Journal of Pediatric Hematology/Oncology. 2003;25:813–815.CrossRefGoogle Scholar
Uysal, Z, Ileri, T, Azik, F, et al. Reversible myelofibrosis associated with hemophagocytic lymphohistiocytosis. Pediatric Blood & Cancer. 2007;49:108–109.CrossRefGoogle ScholarPubMed
Aydinok, Y. Myelofibrosis in a child with EBV-associated hemophagocytic lymphohistiocytosis. Pediatric Blood & Cancer. 2008;51:311.CrossRefGoogle Scholar
Friedman, GK, Hammers, Y, Reddy, V, Pressey, JG. Myelofibrosis in a patient with familial hemophagocytic lymphohistiocytosis. Pediatric Blood & Cancer. 2008;50:1260–1262.CrossRefGoogle Scholar
Nettleship, T, Tay, W. Rare forms of urticaria. British Medical Journal. 1869;2:323–330.Google Scholar
Ehrlich, P. Beitrage zur Theorie und Praxis der Histologischen Farbung. Thesis. Leipzig University; 1878.
Unna, P. Beitrage zur Anatomie und Pathogenese der urticaria simplex und pigmentosa. Monatschrift der praktischen. Dermatologie. 1887;6:9–18.Google Scholar
Touraine, A, Solente, G, Renault, P. Urticaire pigmentaire avec reaction splenique et myelinique. Bulletin de la Société Française de Dermatologie et de Syphiligraphie. 1933;40:1691.Google Scholar
Ellis, JM. Urticaria pigmentosa; a report of a case with autopsy. Archives of Pathology. 1949;48:426–435.Google ScholarPubMed
Kitamura, Y, Go, S, Hatanaka, K. Decrease of mast cells in W/Wv mice and their increase by bone marrow transplantation. Blood. 1978;52:447–452.Google ScholarPubMed
Kitamura, Y, Yokoyama, M, Matsuda, H, Ohno, T, Mori, KJ. Spleen colony-forming cell as common precursor for tissue mast cells and granulocytes. Nature. 1981;291:159–160.CrossRefGoogle ScholarPubMed
Kirshenbaum, AS, Kessler, SW, Goff, JP, Metcalfe, DD. Demonstration of the origin of human mast cells from CD34+ bone marrow progenitor cells. Journal of Immunology. 1991;146:1410–1415.Google ScholarPubMed
Orfao, A, Garcia-Montero, AC, Sanchez, L, Escribano, L. Recent advances in the understanding of mastocytosis: the role of KIT mutations. British Journal of Haematology. 2007;138:12–30.CrossRefGoogle ScholarPubMed
Galli, SJ, Kitamura, Y. Genetically mast-cell-deficient W/Wv and Sl/Sld mice. Their value for the analysis of the roles of mast cells in biologic responses in vivo. The American Journal of Pathology. 1987;127:191–198.Google ScholarPubMed
Geissler, EN, Ryan, MA, Housman, . The dominant-white spotting (W) locus of the mouse encodes the c-kit proto-oncogene. Cell. 1988;55:185–192.CrossRefGoogle ScholarPubMed
Chabot, B, Stephenson, DA, Chapman, VM, Besmer, P, Bernstein, A. The proto-oncogene c-kit encoding a transmembrane tyrosine kinase receptor maps to the mouse W locus. Nature. 1988;335:88–89.CrossRefGoogle ScholarPubMed
Copeland, NG, Gilbert, DJ, Cho, BC, et al. Mast cell growth factor maps near the steel locus on mouse chromosome 10 and is deleted in a number of steel alleles. Cell. 1990;63:175–183.CrossRefGoogle Scholar
Huang, E, Nocka, K, Beier, DR, et al. The hematopoietic growth factor KL is encoded by the Sl locus and is the ligand of the c-kit receptor, the gene product of the W locus. Cell. 1990;63:225–233.CrossRefGoogle Scholar
Zsebo, KM, Williams, DA, Geissler, EN, et al. Stem cell factor is encoded at the Sl locus of the mouse and is the ligand for the c-kit tyrosine kinase receptor. Cell. 1990;63:213–224.CrossRefGoogle Scholar
Furitsu, T, Tsujimura, T, Tono, T, et al. Identification of mutations in the coding sequence of the proto-oncogene c-kit in a human mast cell leukemia cell line causing ligand-independent activation of c-kit product. The Journal of Clinical Investigation. 1993;92:1736–1744.CrossRefGoogle Scholar
Tsujimura, T, Furitsu, T, Morimoto, M, et al. Ligand-independent activation of c-kit receptor tyrosine kinase in a murine mastocytoma cell line P-815 generated by a point mutation. Blood. 1994;83:2619–2626.Google Scholar
Nagata, H, Worobec, AS, Oh, CK, et al. Identification of a point mutation in the catalytic domain of the protooncogene c-kit in peripheral blood mononuclear cells of patients who have mastocytosis with an associated hematologic disorder. Proceedings of the National Academy of Sciences of the United States of America. 1995;92:10560–10564.CrossRefGoogle ScholarPubMed
Longley, BJ, Tyrrell, L, Lu, SZ, et al. Somatic c-KIT activating mutation in urticaria pigmentosa and aggressive mastocytosis: establishment of clonality in a human mast cell neoplasm. Nature Genetics. 1996;12:312–314.CrossRefGoogle Scholar
Longley, BJ, Reguera, MJ, Ma, Y. Classes of c-KIT activating mutations: proposed mechanisms of action and implications for disease classification and therapy. Leukemia Research. 2001;25:571–576.CrossRefGoogle ScholarPubMed
Longley, BJ Jr., Metcalfe, DD, Tharp, M, et al. Activating and dominant inactivating c-KIT catalytic domain mutations in distinct clinical forms of human mastocytosis. Proceedings of the National Academy of Sciences of the United States of America. 1999;96:1609–1614.CrossRefGoogle ScholarPubMed
Fritsche-Polanz, R, Jordan, JH, Feix, A, et al. Mutation analysis of C-KIT in patients with myelodysplastic syndromes without mastocytosis and cases of systemic mastocytosis. British Journal of Haematology. 2001;113:357–364.CrossRefGoogle ScholarPubMed
Buttner, C, Henz, BM, Welker, P, Sepp, NT, Grabbe, J. Identification of activating c-kit mutations in adult-, but not in childhood-onset indolent mastocytosis: a possible explanation for divergent clinical behavior. The Journal of Investigative Dermatology. 1998;111:1227–1231.CrossRefGoogle Scholar
Arceci, RJ, Longley, BJ, Emanuel, PD. Atypical cellular disorders. Hematology/the Education Program of the American Society of Hematology. 2002;297–314.Google ScholarPubMed
Yanagihori, H, Oyama, N, Nakamura, K, Kaneko, F. c-kit mutations in patients with childhood-onset mastocytosis and genotype-phenotype correlation. The Journal of Molecular Diagnostics. 2005;7:252–257.CrossRefGoogle ScholarPubMed
Kettelhut, BV, Metcalfe, DD. Pediatric mastocytosis. The Journal of Investigative Dermatology. 1991;96:15S–18S.CrossRefGoogle ScholarPubMed
Wolff, K, Komar, M, Petzelbauer, P. Clinical and histopathological aspects of cutaneous mastocytosis. Leukemia Research. 2001;25:519–528.CrossRefGoogle ScholarPubMed
Kettelhut, BV, Parker, RI, Travis, WD, Metcalfe, DD. Hematopathology of the bone marrow in pediatric cutaneous mastocytosis. A study of 17 patients. American Journal of Clinical Pathology. 1989;91:558–562.CrossRefGoogle ScholarPubMed
Patnaik, MM, Rindos, M, Kouides, PA, Tefferi, A, Pardanani, A. Systemic mastocytosis: a concise clinical and laboratory review. Archives of Pathology & Laboratory Medicine. 2007;131:784–791.Google ScholarPubMed
Schwartz, LB. Clinical utility of tryptase levels in systemic mastocytosis and associated hematologic disorders. Leukemia Research. 2001;25:553–562.CrossRefGoogle ScholarPubMed
Schwartz, LB, Min, HK, Ren, S, et al. Tryptase precursors are preferentially and spontaneously released, whereas mature tryptase is retained by HMC-1 cells, Mono-Mac-6 cells, and human skin-derived mast cells. Journal of Immunology. 2003;170:5667–5673.CrossRefGoogle ScholarPubMed
Kanthawatana, S, Carias, K, Arnaout, R, et al. The potential clinical utility of serum alpha-protryptase levels. The Journal of Allergy and Clinical Immunology. 1999;103:1092–1099.CrossRefGoogle ScholarPubMed
Sperr, WR, Jordan, JH, Fiegl, M, et al. Serum tryptase levels in patients with mastocytosis: correlation with mast cell burden and implication for defining the category of disease. International Archives of Allergy and Immunology. 2002;128:136–141.CrossRefGoogle Scholar
Parker, RI. Hematologic aspects of mastocytosis: I: Bone marrow pathology in adult and pediatric systemic mast cell disease. The Journal of Investigative Dermatology. 1991;96:47S–51S.CrossRefGoogle ScholarPubMed
Horny, HP, Parwaresch, MR, Lennert, K. Bone marrow findings in systemic mastocytosis. Human Pathology. 1985;16:808–814.CrossRefGoogle ScholarPubMed
Brunning, RD, McKenna, RW, Rosai, J, Parkin, JL, Risdall, R. Systemic mastocytosis. Extracutaneous manifestations. The American Journal of Surgical Pathology. 1983;7:425–438.CrossRefGoogle ScholarPubMed
Chott, A, Guenther, P, Huebner, A, et al. Morphologic and immunophenotypic properties of neoplastic cells in a case of mast cell sarcoma. The American Journal of Surgical Pathology. 2003;27:1013–1019.CrossRefGoogle Scholar
Brcic, L, Vuletic, LB, Stepan, J, et al. Mast-cell sarcoma of the tibia. Journal of Clinical Pathology. 2007;60:424–425.CrossRefGoogle ScholarPubMed
Escribano, L, Diaz-Agustin, B, López, A, et al. Immunophenotypic analysis of mast cells in mastocytosis: When and how to do it. Proposals of the Spanish Network on Mastocytosis (REMA). Cytometry. Part B, Clinical Cytometry. 2004;58:1–8.CrossRefGoogle Scholar
Escribano, L, Garcia Montero, AC, Nunez, R, Orfao, A. Flow cytometric analysis of normal and neoplastic mast cells: role in diagnosis and follow-up of mast cell disease. Immunology and Allergy Clinics of North America. 2006;26:535–547.CrossRefGoogle ScholarPubMed
Irani, AM, Bradford, TR, Kepley, CL, Schechter, NM, Schwartz, LB. Detection of MCT and MCTC types of human mast cells by immunohistochemistry using new monoclonal anti-tryptase and anti-chymase antibodies. The Journal of Histochemistry and Cytochemistry. 1989;37:1509–1515.CrossRefGoogle ScholarPubMed
Walls, AF, Jones, DB, Williams, JH, Church, MK, Holgate, ST. Immunohistochemical identification of mast cells in formaldehyde-fixed tissue using monoclonal antibodies specific for tryptase. The Journal of Pathology. 1990;162:119–126.CrossRefGoogle ScholarPubMed
Li, WV, Kapadia, SB, Sonmez-Alpan, E, Swerdlow, SH. Immunohistochemical characterization of mast cell disease in paraffin sections using tryptase, CD68, myeloperoxidase, lysozyme, and CD20 antibodies. Modern Pathology. 1996;9:982–988.Google ScholarPubMed
Buckley, MG, McEuen, AR, Walls, AF. The detection of mast cell subpopulations in formalin-fixed human tissues using a new monoclonal antibody specific for chymase. The Journal of Pathology. 1999;189:138–143.3.0.CO;2-H>CrossRefGoogle ScholarPubMed
Natkunam, Y, Rouse, RV. Utility of paraffin section immunohistochemistry for C-KIT (CD117) in the differential diagnosis of systemic mast cell disease involving the bone marrow. The American Journal of Surgical Pathology. 2000;24:81–91.CrossRefGoogle ScholarPubMed
Hoyer, JD, Grogg, KL, Hanson, CA, Gamez, JD, Dogan, A. CD33 detection by immunohistochemistry in paraffin-embedded tissues: a new antibody shows excellent specificity and sensitivity for cells of myelomonocytic lineage. American Journal of Clinical Pathology. 2008;129:316–323.CrossRefGoogle ScholarPubMed
Escribano, L, Orfao, A, Villarrubia, J, et al. Expression of lymphoid-associated antigens in mast cells: report of a case of systemic mast cell disease. British Journal of Haematology. 1995;91:941–943.CrossRefGoogle ScholarPubMed
Escribano, L, Orfao, A, Diaz-Agustin, B, et al. Indolent systemic mast cell disease in adults: immunophenotypic characterization of bone marrow mast cells and its diagnostic implications. Blood. 1998;91:2731–2736.Google ScholarPubMed
Jordan, JH, Walchshofer, S, Jurecka, W, et al. Immunohistochemical properties of bone marrow mast cells in systemic mastocytosis: evidence for expression of CD2, CD117/Kit, and bcl-x(L). Human Pathology. 2001;32:545–552.CrossRefGoogle Scholar
Sotlar, K, Horny, HP, Simonitsch, I, et al. CD25 indicates the neoplastic phenotype of mast cells: a novel immunohistochemical marker for the diagnosis of systemic mastocytosis (SM) in routinely processed bone marrow biopsy specimens. The American Journal of Surgical Pathology. 2004;28:1319–1325.CrossRefGoogle Scholar
Krokowski, M, Sotlar, K, Krauth, MT, et al. Delineation of patterns of bone marrow mast cell infiltration in systemic mastocytosis: value of CD25, correlation with subvariants of the disease, and separation from mast cell hyperplasia. American Journal of Clinical Pathology. 2005;124:560–568.CrossRefGoogle ScholarPubMed
Hahn, HP, Hornick, JL. Immunoreactivity for CD25 in gastrointestinal mucosal mast cells is specific for systemic mastocytosis. The American Journal of Surgical Pathology. 2007;31:1669–1676.CrossRefGoogle ScholarPubMed
Hollmann, TJ, Brenn, T, Hornick, JL. CD25 expression on cutaneous mast cells from adult patients presenting with urticaria pigmentosa is predictive of systemic mastocytosis. The American Journal of Surgical Pathology. 2008;32:139–145.CrossRefGoogle ScholarPubMed
Baumgartner, C, Sonneck, K, Krauth, MT, et al. Immunohistochemical assessment of CD25 is equally sensitive and diagnostic in mastocytosis compared to flow cytometry. European Journal of Clinical Investigation. 2008;38:326–335.CrossRefGoogle ScholarPubMed
Zuluaga, TT, Hsieh, FH, Bodo, J, Dong, HY, Hsi, ED. Detection of phospho-STAT5 in mast cells: a reliable phenotypic marker of systemic mast cell disease that reflects constitutive tyrosine kinase activation. British Journal of Haematology. 2007;139:31–40.CrossRefGoogle Scholar
Sundram, UN, Natkunam, Y. Mast cell tryptase and microphthalmia transcription factor effectively discriminate cutaneous mast cell disease from myeloid leukemia cutis. Journal of Cutaneous Pathology. 2007;34:289–295.CrossRefGoogle ScholarPubMed
Yang, F, Tran, TA, Carlson, JA, et al. Paraffin section immunophenotype of cutaneous and extracutaneous mast cell disease: comparison to other hematopoietic neoplasms. The American Journal of Surgical Pathology. 2000;24:703–709.CrossRefGoogle ScholarPubMed
Ma, Y, Zeng, S, Metcalfe, DD, et al. The c-KIT mutation causing human mastocytosis is resistant to STI571 and other KIT kinase inhibitors; kinases with enzymatic site mutations show different inhibitor sensitivity profiles than wild-type kinases and those with regulatory-type mutations. Blood. 2002;99:1741–1744.CrossRefGoogle ScholarPubMed
Frost, MJ, Ferrao, PT, Hughes, TP, Ashman, LK. Juxtamembrane mutant V560GKit is more sensitive to Imatinib (STI571) compared with wild-type c-kit whereas the kinase domain mutant D816VKit is resistant. Molecular Cancer Therapeutics. 2002;1:1115–1124.Google ScholarPubMed
Akin, C, Brockow, K, D'Ambrosio, C, et al. Effects of tyrosine kinase inhibitor STI571 on human mast cells bearing wild-type or mutated c-kit. Experimental Hematology. 2003;31:686–692.CrossRefGoogle ScholarPubMed
Gleixner, KV, Mayerhofer, M, Sonneck, K, et al. Synergistic growth-inhibitory effects of two tyrosine kinase inhibitors, dasatinib and PKC412, on neoplastic mast cells expressing the D816V-mutated oncogenic variant of KIT. Haematologica. 2007;92:1451–1459.CrossRefGoogle ScholarPubMed
Sotlar, K, Escribano, L, Landt, O, et al. One-step detection of c-kit point mutations using peptide nucleic acid-mediated polymerase chain reaction clamping and hybridization probes. The American Journal of Pathology. 2003;162:737–746.CrossRefGoogle ScholarPubMed
Corless, CL, Harrell, P, Lacouture, M, et al. Allele-specific polymerase chain reaction for the imatinib-resistant KIT D816V and D816F mutations in mastocytosis and acute myelogenous leukemia. The Journal of Molecular Diagnostics. 2006;8:604–612.CrossRefGoogle ScholarPubMed
Tan, A, Westerman, D, McArthur, GA, et al. Sensitive detection of KIT D816V in patients with mastocytosis. Clinical Chemistry. 2006;52:2250–2257.CrossRefGoogle ScholarPubMed
Garcia-Montero, AC, Jara-Acevedo, M, Teodosio, C, et al. KIT mutation in mast cells and other bone marrow hematopoietic cell lineages in systemic mast cell disorders: a prospective study of the Spanish Network on Mastocytosis (REMA) in a series of 113 patients. Blood. 2006;108:2366–2372.CrossRefGoogle Scholar
Valent, P, Akin, C, Escribano, L, et al. Standards and standardization in mastocytosis: consensus statements on diagnostics, treatment recommendations and response criteria. European Journal of Clinical Investigation. 2007;37:435–453.CrossRefGoogle ScholarPubMed
Schumacher, JA, Elenitoba-Johnson, KS, Lim, MS. Detection of the c-kit D816V mutation in systemic mastocytosis by allele-specific PCR. Journal of Clinical Pathology. 2008;61:109–114.CrossRefGoogle ScholarPubMed

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
×