Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-x24gv Total loading time: 0 Render date: 2024-05-15T20:40:28.930Z Has data issue: false hasContentIssue false

76 - JAK2 and myeloproliferative neoplasms

from Part 3.6 - Molecular pathology: lymphoma and leukemia

Published online by Cambridge University Press:  05 February 2015

Ross L. Levine
Affiliation:
Human Oncology and Pathogenesis Program, Leukemia Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
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

Myeloproliferative neoplasms

Myeloproliferative neoplasms (MPNs) are clonal disorders of the myeloid lineage that clinically present as an excess of cells from one or more terminally differentiated myeloid compartments (Figure 76.1). Although there are a spectrum of MPNs involving all of the different myeloid compartments, the most common MPN are polcythemia vera (PV), essential thrombocytosis (ET), primary myelofibrosis (PMF), and chronic myeloid leukemia (CML). CML is characterized by neutrophilia and by the invariant presence of the BCR–ABL fusion tyrosine kinase, and is the subject of detailed discussion elsewhere in this volume. PV clinically presents with increased red blood-cell counts (hemoglobin and hematocrit), with variable involvement of the platelet and white blood-cell lineages. In contrast, patients with ET present with thrombocytosis without increased erythrocytosis. PMF is associated with a leukoerythroblastosis, splenomegaly, extra-medullary hematopoiesis, and systemic symptoms, and on bone-marrow exam PMF patients are found to have reticulin fibrosis. Over time, a subset of PV and ET patients progress myelofibrosis (MF); post-PV/ET MF is not distinguishable clinically or biologically from de novo PMF.

A significant proportion of MPN patients are diagnosed based on the identification of asymptomatic abnormalities on a complete blood count. A subset of patients at diagnosis, or with disease progression, develop symptomatic splenomegaly, constitutional symptoms, thrombosis, bleeding, or infection. From a clinical perspective, the most feared complications to occur over time are progressive bone-marrow failure or transformation to acute myeloid leukemia (AML), which are associated with an adverse overall prognosis (1). Therefore the current goals in MPN treatment are to ameliorate symptoms, and to prevent disease progression and transformation.

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 818 - 825
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

Mesa, RA, Li, CY, Ketterling, RP, et al. Leukemic transformation in myelofibrosis with myeloid metaplasia: a single-institution experience with 91 cases. Blood 2005;105:973–7.CrossRefGoogle ScholarPubMed
Adamson, JW, Fialkow, PJ, Murphy, S, Prchal, JF, Steinmann, L. Polycythemia vera: stem-cell and probable clonal origin of the disease. New England Journal of Medicine 1976;295:913–16.CrossRefGoogle ScholarPubMed
James, C, Ugo, V, Le Couédic, JP, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature 2005;434:1144–8.CrossRefGoogle ScholarPubMed
Baxter, EJ, Scott, LM, Campbell, PJ, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 2005;365:1054–61.CrossRefGoogle ScholarPubMed
Kralovics, R, Passamonti, F, Buser, AS, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. New England Journal of Medicine 2005;352:1779–90.CrossRefGoogle ScholarPubMed
Zhao, R, Xing, S, Li, Z, et al. Identification of an acquired JAK2 mutation in polycythemia vera. Journal of Biological Chemistry 2005;280:22 788–92.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;7:387–97.CrossRefGoogle ScholarPubMed
Kralovics, R, Guan, Y, Prchal, JT. Acquired uniparental disomy of chromosome 9p is a frequent stem cell defect in polycythemia vera. Experimental Hematology 2002;30:229–36.CrossRefGoogle ScholarPubMed
Dusa, A, Staerk, J, Elliot, J, et al. Substitution of pseudokinase domain residue Val-617 by large non-polar amino acids causes activation of JAK2. Journal of Biological Chemistry 2008;283:12 941–8.CrossRefGoogle ScholarPubMed
Scott, LM, Scott, MA, Campbell, PJ, Green, AR. Progenitors homozygous for the V617F mutation occur in most patients with polycythemia vera, but not essential thrombocythemia. Blood 2006;108:2435–7.CrossRefGoogle Scholar
Wernig, G, Mercher, T, Okabe, R, et al. Expression of Jak2V617F causes a polycythemia vera-like disease with associated myelofibrosis in a murine bone marrow transplant model. Blood 2006;107:4274–81.CrossRefGoogle Scholar
Lacout, C, Pisani, DF, Tulliez, M, et al. JAK2V617F expression in murine hematopoietic cells leads to MPD mimicking human PV with secondary myelofibrosis. Blood 2006;108:1652–60.CrossRefGoogle ScholarPubMed
Bumm, TG, Elsea, C, Corbin, AS, et al. Characterization of murine JAK2V617F-positive myeloproliferative disease. Cancer Research 2006;66:11 156–65.CrossRefGoogle ScholarPubMed
Zaleskas, VM, Krause, DS, Lazarides, K, et al. Molecular pathogenesis and therapy of polycythemia induced in mice by JAK2 V617F. PLoS One 2006;1:e18.CrossRefGoogle ScholarPubMed
Tiedt, R, Hoa-Shen, H, Sobas, MA, et al. Ratio of mutant JAK2-V617F to wild-type Jak2 determines the MPD phenotypes in transgenic mice. Blood 2008;111:3931–40.CrossRefGoogle ScholarPubMed
Xing, S, Wanting, TH, Zhao, W, et al. Transgenic expression of JAK2V617F causes myeloproliferative disorders in mice. Blood 2008;111:5109–17.CrossRefGoogle ScholarPubMed
Akada, H, Yan, D, Zou, H, et al. Conditional expression of heterozygous or homozygous Jak2V617F from its endogenous promoter induces a polycythemia vera-like disease. Blood 2010;115:3589–97.CrossRefGoogle ScholarPubMed
Mullally, A, Lane, SW, Ball, B, et al. Physiological Jak2V617F expression causes a lethal myeloproliferative neoplasm with differential effects on hematopoietic stem and progenitor cells. Cancer Cell 2010;17:584–96.CrossRefGoogle ScholarPubMed
Marty, C, Lacout, C, Martin, A, et al. Myeloproliferative neoplasm induced by constitutive expression of JAK2V617F in knock-in mice. Blood 2010;116:783–7.CrossRefGoogle ScholarPubMed
Li, J, Spensberger, D, Ahn, JS, et al. JAK2 V617F impairs hematopoietic stem cell function in a conditional knock-in mouse model of JAK2 V617F-positive essential thrombocythemia. Blood 2010;116:1528–38.CrossRefGoogle Scholar
Campbell, PJ, Scott, LM, Buck, G, et al. Definition of subtypes of essential thrombocythaemia and relation to polycythaemia vera based on JAK2 V617F mutation status: a prospective study. Lancet 2005;366:1945–53.CrossRefGoogle ScholarPubMed
Pardanani, AD, Levine, RL, Lasho, T, et al. MPL515 mutations in myeloproliferative and other myeloid disorders: a study of 1182 patients. Blood 2006;108:3472–6.CrossRefGoogle ScholarPubMed
Pikman, Y, Lee, BH, Mercher, T, et al. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Medicine 2006;3:e270.CrossRefGoogle ScholarPubMed
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;103:4198–200.CrossRefGoogle ScholarPubMed
Scott, LM, Tong, W, Levine, RL, et al. JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. New England Journal of Medicine 2007;356:459–68.CrossRefGoogle ScholarPubMed
Oh, ST, Simonds, EF, Jones, C, et al. Novel mutations in the inhibitory adaptor protein LNK drive JAK-STAT signaling in patients with myeloproliferative neoplasms. Blood 2010;116:988–92.CrossRefGoogle ScholarPubMed
Beer, PA, Jones, AV, Bench, AJ, et al. Clonal diversity in the myeloproliferative neoplasms: independent origins of genetically distinct clones. British Journal of Haematology 2009;144:904–8.CrossRefGoogle ScholarPubMed
Campbell, PJ, Baxter, EJ, Beer, PA, et al. Mutation of JAK2 in the myeloproliferative disorders: timing, clonality studies, cytogenetic associations, and role in leukemic transformation. Blood 2006;108:3548–55.CrossRefGoogle ScholarPubMed
Theocharides, A, Boissinot, M, Girodon, F, et al. Leukemic blasts in transformed JAK2-V617F-positive myeloproliferative disorders are frequently negative for the JAK2-V617F mutation. Blood 2007;110:375–9.CrossRefGoogle ScholarPubMed
Langemeijer, SM, Kuiper, RP, Berends, M, et al. Acquired mutations in TET2 are common in myelodysplastic syndromes. Nature Genetics 2009;41:838–42.CrossRefGoogle ScholarPubMed
Delhommeau, F, Dupont, S, Della Valle, V, et al. Mutation in TET2 in myeloid cancers. New England Journal of Medicine 2009;360:2289–301.CrossRefGoogle ScholarPubMed
Tahiliani, M, Koh, KP, Shen, Y, et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 2009;324:930–5.CrossRefGoogle ScholarPubMed
Ko, M, Huang, Y, Jankowska, AM, et al. Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2. Nature 2010;468:839–43.CrossRefGoogle ScholarPubMed
Figueroa, ME, Abdel-Wahab, O, Lu, C, et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell 2010;18:553–67.CrossRefGoogle Scholar
Moran-Crusio, K, Reavie, L, Shih, A, et al. Tet2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation. Cancer Cell 2011;20:11–24.CrossRefGoogle ScholarPubMed
Quivoron, C, Couronné, L, Della Valle, V, et al. TET2 inactivation results in pleiotropic hematopoietic abnormalities in mouse and is a recurrent event during human lymphomagenesis. Cancer Cell 2011;20:25–38.CrossRefGoogle ScholarPubMed
Carbuccia, N, Murati, A, Trouplin, V, et al. Mutations of ASXL1 gene in myeloproliferative neoplasms. Leukemia 2009;23:2183–6.CrossRefGoogle ScholarPubMed
Carbuccia, N, Trouplin, V, Gelsi-Boyer, V, et al. Mutual exclusion of ASXL1 and NPM1 mutations in a series of acute myeloid leukemias. Leukemia 2010;24:469–73.CrossRefGoogle Scholar
Gelsi-Boyer, V, Adélaïde, Trouplin V, et al. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. British Journal of Haematology 2009;145:788–800.CrossRefGoogle ScholarPubMed
Mardis, ER, Ding, L, Dooling, DJ, et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. New England Journal of Medicine 2009;361:1058–66.CrossRefGoogle ScholarPubMed
Ward, PS, Patel, J, Wise, DR, et al. The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell 2010;17:225–34.CrossRefGoogle ScholarPubMed
Marcucci, G, Maharry, K, Wu, YZ, et al. IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. Journal of Clinical Oncology 2010;28:2348–55.CrossRefGoogle ScholarPubMed
Gross, S, Cairns, RA, Minden, MD, et al. Cancer-associated metabolite 2-hydroxyglutarate accumulates in acute myelogenous leukemia with isocitrate dehydrogenase 1 and 2 mutations. Journal of Experimental Medicine 2010;207:339–44.CrossRefGoogle ScholarPubMed
Ernst, T, Chase, AJ, Score, J, et al. Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nature Genetics 2010;42:722–6.CrossRefGoogle ScholarPubMed
Nikoloski, G, Langemeijer, SM, Kuiper, RP, et al. Somatic mutations of the histone methyltransferase gene EZH2 in myelodysplastic syndromes. Nature Genetics 2010;42:665–7.CrossRefGoogle ScholarPubMed
Yan, XJ, Xu, J, Gu, ZH, et al. Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia. Nature Genetics 2011;43:309–15.CrossRefGoogle ScholarPubMed
Ley, TJ, Ding, L, Walter, MJ, et al. DNMT3A mutations in acute myeloid leukemia. New England Journal of Medicine 2010;363:2424–33.CrossRefGoogle ScholarPubMed
Abdel-Wahab, O, Manshouri, T, Patel, J, et al. Genetic analysis of transforming events that convert chronic myeloproliferative neoplasms to leukemias. Cancer Research 2010;70:447–52.CrossRefGoogle ScholarPubMed
Landgren, O, Goldin, LR, Kristinsson, SY, et al. Increased risks of polycythemia vera, essential thrombocythemia, and myelofibrosis among 24,577 first-degree relatives of 11,039 patients with myeloproliferative neoplasms in Sweden. Blood 2008;112:2199–204.CrossRefGoogle ScholarPubMed
Jones, AV, Chase, A, Silver, RT, et al. JAK2 haplotype is a major risk factor for the development of myeloproliferative neoplasms. Nature Genetics 2009;41:446–9.CrossRefGoogle ScholarPubMed
Kilpivaara, O, Mukherjee, S, Schram, AM, et al. A germline JAK2 SNP is associated with predisposition to the development of JAK2(V617F)-positive myeloproliferative neoplasms. Nature Genetics 2009;41:455–9.CrossRefGoogle ScholarPubMed
Olcaydu, D, Harutunyan, A, Jäger, R, et al. A common JAK2 haplotype confers susceptibility to myeloproliferative neoplasms. Nature Genetics 2009;41:450–4.CrossRefGoogle ScholarPubMed
Parganas, E, Wang, D, Stravopodis, D, et al. Jak2 is essential for signaling through a variety of cytokine receptors. Cell 1998;93:385–95.CrossRefGoogle ScholarPubMed
Boggon, TJ, Li, Y, Manley, PW, Eck, MJ. Crystal structure of the Jak3 kinase domain in complex with a staurosporine analog. Blood 2005;106:996–1002.CrossRefGoogle ScholarPubMed
Lucet, IS, Fantino, E, Styles, M, et al. The structural basis of Janus kinase 2 inhibition by a potent and specific pan-Janus kinase inhibitor. Blood 2006;107:176–83.CrossRefGoogle ScholarPubMed
Saharinen, P, Silvennoinen, O. The pseudokinase domain is required for suppression of basal activity of Jak2 and Jak3 tyrosine kinases and for cytokine-inducible activation of signal transduction. Journal of Biological Chemistry 2002;277:47 954–63.CrossRefGoogle ScholarPubMed
Bercovich, D, Ganmore, I, Scott, LM, et al. Mutations of JAK2 in acute lymphoblastic leukaemias associated with Down's syndrome. Lancet 2008;372:1484–92.CrossRefGoogle ScholarPubMed
Mullighan, CG, Zhang, J, Harvey, RC, et al. JAK mutations in high-risk childhood acute lymphoblastic leukemia. Proceedings of the National Academy of Sciences USA 2009;106:9414–18.CrossRefGoogle ScholarPubMed
Pardanani, A, Hood, J, Lasho, T, et al. TG101209, a small molecule JAK2-selective kinase inhibitor potently inhibits myeloproliferative disorder-associated JAK2V617F and MPLW515L/K mutations. Leukemia 2007;21:1658–68.CrossRefGoogle ScholarPubMed
Wernig, G, Kharas, MG, Okabe, R, et al. Efficacy of TG101348, a selective JAK2 inhibitor, in treatment of a murine model of JAK2V617F-induced polycythemia vera. Cancer Cell 2008;13:311–20.CrossRefGoogle ScholarPubMed
Hexner, EO, Serdikoff, C, Jan, M, et al. Lestaurtinib (CEP701) is a JAK2 inhibitor that suppresses JAK2/STAT5 signaling and the proliferation of primary erythroid cells from patients with myeloproliferative disorders. Blood 2008;111:5663–71.CrossRefGoogle ScholarPubMed
Hedvat, M, Huszar, D, Herrmann, A, et al. The JAK2 inhibitor AZD1480 potently blocks Stat3 signaling and oncogenesis in solid tumors. Cancer Cell 2009;16:487–97.CrossRefGoogle ScholarPubMed
Koppikar, P, Abdel-Wahab, O, Hedvat, C, et al. Efficacy of the JAK2 inhibitor INCB16562 in a murine model of MPLW515L-induced thrombocytosis and myelofibrosis. Blood 2010;115:2919–27.CrossRefGoogle Scholar
Verstovsek, S, Kantarjian, H, Mesa, RA, et al. Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis. New England Journal of Medicine 2010;363:1117–27.CrossRefGoogle ScholarPubMed
Pardanani, A, Gotlib, JR, Jamieson, C, et al. Safety and efficacy of TG101348, a selective JAK2 inhibitor, in myelofibrosis. Journal of Clinical Oncology 2011;29:789–96.CrossRefGoogle ScholarPubMed
Marubayashi, S, Koppikar, P, Taldone, T, et al. HSP90 is a therapeutic target in JAK2-dependent myeloproliferative neoplasms in mice and humans. Journal of Clinical Investigation 2010;120:3578–93.CrossRefGoogle ScholarPubMed
Wang, Y, Fiskus, W, Chong, DG, et al. Cotreatment with panobinostat and JAK2 inhibitor TG101209 attenuates JAK2V617F levels and signaling and exerts synergistic cytotoxic effects against human myeloproliferative neoplastic cells. Blood 2009;114:5024–33.CrossRefGoogle ScholarPubMed
Flex, E, Petrangeli, V, Stella, L, et al. Somatically acquired JAK1 mutations in adult acute lymphoblastic leukemia. Journal of Experimental Medicine 2008;205:751–8.CrossRefGoogle ScholarPubMed
Yoda, A, Yoda, Y, Chiaretti, S, et al. Functional screening identifies CRLF2 in precursor B-cell acute lymphoblastic leukemia. Proceedings of the National Academy of Sciences USA 2009;107:252–7.CrossRefGoogle ScholarPubMed
Shochat, C, Tal, N, Bandapalli, OR, et al. Gain-of-function mutations in interleukin-7 receptor-alpha (IL7R) in childhood acute lymphoblastic leukemias. Journal of Experimental Medicine 2011;208:901–8.CrossRefGoogle Scholar
Rebouissou, S, Amessou, M, Couchy, G, et al. Frequent in-frame somatic deletions activate gp130 in inflammatory hepatocellular tumours. Nature 2009;457:200–4.CrossRefGoogle ScholarPubMed
Gao, SP, Mark, KG, Leslie, K, et al. Mutations in the EGFR kinase domain mediate STAT3 activation via IL-6 production in human lung adenocarcinomas. Journal of Clinical Investigation 2007;117:3846–56.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
×