Acessibilidade / Reportar erro

Molecular biology of Philadelphia-negative myeloproliferative neoplasms

Abstract

Myeloproliferative neoplasms are clonal diseases of hematopoietic stem cells characterized by myeloid hyperplasia and increased risk of developing acute myeloid leukemia. Myeloproliferative neoplasms are caused, as any other malignancy, by genetic defects that culminate in the neoplastic phenotype. In the past six years, since the identification of JAK2V617F, we have experienced a substantial increase in our knowledge about the genetic mechanisms involved in the genesis of myeloproliferative neoplasms. Mutations described in several genes have revealed a considerable degree of molecular homogeneity between different subtypes of myeloproliferative neoplasms. At the same time, the molecular differences between each subtype have become clearer. While mutations in several genes, such as JAK2, myeloproliferative leukemia (MPL) and LNK have been validated in functional assays or animal models as causative mutations, the roles of other recurring mutations in the development of disease, such as TET2 and ASXL1 remain to be elucidated. In this review we will examine the most prevalent recurring gene mutations found in myeloproliferative neoplasms and their molecular consequences.

polycythemia vera; primary myelofibrosis; thrombocythemia, essential; leukemia, myeloid, acute; myeloproliferative disorders; janus kinase 2


REVIEW ARTICLE

Molecular biology of Philadelphia-negative myeloproliferative neoplasms

Paulo Vidal Campregher; Fábio Pires de Souza Santos; Guilherme Fleury Perini; Nelson Hamerschlak

Hematology Department, Hospital Israelita Albert Einstein - HIAE, São Paulo, SP, Brazil

Corresponding author Corresponding author: Paulo Vidal Campregher Hospital Israelita Albert Einstein. Laboratório de Técnicas Especializadas Avenida Albert Einstein, 627/701Bloco D, 1º andar 05652-900 - São Paulo, SP, Brazil Phone: 55 11 5475-2000 paulo.campregher@einstein.br

ABSTRACT

Myeloproliferative neoplasms are clonal diseases of hematopoietic stem cells characterized by myeloid hyperplasia and increased risk of developing acute myeloid leukemia. Myeloproliferative neoplasms are caused, as any other malignancy, by genetic defects that culminate in the neoplastic phenotype. In the past six years, since the identification of JAK2V617F, we have experienced a substantial increase in our knowledge about the genetic mechanisms involved in the genesis of myeloproliferative neoplasms. Mutations described in several genes have revealed a considerable degree of molecular homogeneity between different subtypes of myeloproliferative neoplasms. At the same time, the molecular differences between each subtype have become clearer. While mutations in several genes, such as JAK2, myeloproliferative leukemia (MPL) and LNK have been validated in functional assays or animal models as causative mutations, the roles of other recurring mutations in the development of disease, such as TET2 and ASXL1 remain to be elucidated. In this review we will examine the most prevalent recurring gene mutations found in myeloproliferative neoplasms and their molecular consequences.

Keywords: polycythemia vera; primary myelofibrosis; thrombocythemia, essential; leukemia, myeloid, acute; myeloproliferative disorders; janus kinase 2

Introduction

Myeloproliferative neoplasms (MNs) are clonal disorders of hematopoietic stem cells characterized by increased proliferation of myeloid cells and an increased risk of developing acute myeloid leukemia (AML).(1)

Traditionally, MNs have been diagnosed and classified according to which cell type (erythrocytes, platelets or granulocytes) predominates in the peripheral blood and to the bone marrow histology. Since the discovery that the Janus Kinase 2 (JAK2) V617F mutation is present in more than 60% of MN patients,(2-5) the identification of this and other equivalent mutations, such as JAK2 exon 12(6) and myeloproliferative leukemia (MPL) exon 10 mutations,(7) has become an essential step in the diagnosis of these disorders. While the absence of these mutations does not exclude the diagnosis, its presence in the context of myeloid proliferation confirms the diagnosis of MNs.

Regarding the specificity of these mutations for the diagnosis of specific subtypes of MNs, polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF), the picture is very different from that of chronic myelogenous leukemia (CML), in which the presence of the hybrid gene BRC-ABL1 establishes the diagnosis. With the exception of JAK2 exon 12 mutations, which are specific for PV, JAK2V617F can occur in PV, ET and PMF, among other diseases and MPL mutations occur both in ET and PMF.

We now know that other genes are mutated in patients with MN and the physiologic consequences of each one of these mutations are starting to be unveiled. The most relevant mutations identified so far can be broadly classified into three main groups:

1 - Mutations associated with constitutive Signal Transducer and Activators of Transcription (STAT)3/5 activation.

2 - Mutations associated with transcriptional regulation.

3 - Mutations associated with progression to AML.

In this review we will examine the advances in understanding the molecular biology of the BCR-ABL1 negative MNs: PV, PMF and ET and the practical implications of these developments.

Other MNs such as mast cell disease, chronic neutrophilic leukemia and chronic eosinophilic leukemia will not be discussed here.

Mutations associated with constitutive STAT3/5 activation

The JAK-STAT signaling pathway is essential for normal hematopoiesis.(8) This pathway is "turned on" after activation of cell receptors by its ligands, establishing the link between extracellular stimuli and the cellular effects of numerous growth factors, cytokines, interferon, etc.

Upon binding of the ligand, conformational changes in the receptor lead to JAK and STAT phosphorylation. Once phosphorylated, STATs form homodimers and translocate to the nucleus where they will promote transcription of specific genes.(9)

Constitutive STAT3 or STAT5 activation is the hallmark of most, if not all, patients with MNs.(10) Although STAT mutations are rare in MN patients, activating mutations in STAT activators (JAK2 and MPL) or inactivating mutations in STAT inhibitors [LNK and Cas-Br-M ecotropicretroviral transforming sequence (CBL)] have been documented in most patients with MN.

Janus Kinase 2 (JAK2)

JAK2 is a non-receptor tyrosine kinase downstream of a vast number of cytokine receptors, such as erythropoietin (EPO), MPL and granulocyte-macrophage colony-stimulating factor receptor (GM-CSFR).(11) JAK2V617F is the most prevalent mutation found in BCR-ABL1 negative MNs; it is present in roughly 95%, 50% and 60% of PV, ET and PMF patients, respectively.(12) JAK2V617F affects the regulatory pseudokinase domain leading to constitutional activation of JAK2 and, as a consequence, permanently activated STAT3/5. JAK2V617F induces myeloproliferative disease in mice confirming its role as a causal mutation.(13)

While homozygous JAK2V617F mutations are frequently found in PV and PMF patients, most TE patients are heterozygous.(14) This finding contributes to the idea that PMF could represent an advanced stage MNs, with ET and PV representing early stages of the same disease.(15)

Other mutations in the JAK2 gene have been described, occurring predominantly in exon 12.(16) These mutations are specific to PV patients and are mutually exclusive with JAK2V617F.(17)

Due to its high prevalence, the detection of JAK2 mutations has become an essential step in the diagnosis of myeloproliferative disorders. Regarding the prognostic importance of JAK2 mutations, the findings are not conclusive yet; most studies have not found differences in survival or progression to AML between patients with and without JAK2 mutations.(18,19) Intriguingly, a low JAK2V617F allele burden has been associated with worse survival in PMF.(20,21)

Myeloproliferative leukemia virus oncogene (MPL)

MPL is the gene encoding the thrombopoietin receptor. Exon 10 MPL mutations are found in about 6% and 3% of patients with PMF and ET, respectively, and are not mutually exclusive with JAK2V617F. (7,22) No MPL mutations have been described in PV patients. Expression of MPLW515L in cell lines resulted in constitutive phosphorylation of JAK2, STAT3, STAT5, AKT and extracellular signal-regulated kinase (ERK).(23) In a murine model assay, the presence of MPLW515L induced a PMF-like disease, confirming the role of MPL mutations as causative agents in MNs.(23)

Cas-Br-M ecotropicretroviral transforming sequence

(CBL)

The CBL protein can act either as a positive or negative regulator of tyrosine kinase signaling. It can bind to activated receptors, acting as an adaptor protein, recruiting downstream molecules, but it also has E3 ubiquitin ligase activity, 'marking' active kinases for degradation, such as FLT3, KIT and MPL.(24,25) CBL mutations are present in 6% of PMF patients(26) and also in other myeloid malignancies.(27,28) In most cases the mutation is homozygous due to uniparental disomy. To the extent of our knowledge, CBF mutations have not been described in chronic phase ET or PV.

Transfection of CBL mutants was sufficient to induce an oncogenic phenotype in fibroblast cell lines. Additionally, mice with the CBL null phenotype developed a disease characterized by splenomegaly and augmented hematopoietic progenitor pool, suggesting a role for this gene in the development of MNs.(29) Additionally, mutated CBL inhibited E3 ubiquitin ligase activity of wild type CBL in transfected fibroblasts, revealed a dominant negative role for the mutated protein and not only a classical tumor suppressor effect.(29)

SH2B adaptor protein 3 (SH2B3)/LNK

LNK is an adaptor protein that acts as a negative regulator of cytokine signaling. The LNK protein specifically interacts with JAK2 (wild type or mutant V617F), MPL (wild type or mutant W515L) inhibiting the downstream activation of STAT.(30-32) LNK mutations are found in roughly 13% of patients with blast phase MNs(33) but are rarely found in chronic phase ET, PMF(34) and JAK2 wild type erythrocytosis.(35) The few mutations described affect predominantly a hot spot at exon 2 and are heterozygous.(33,34) Mutant LNK has diminished/abolished capacity to inhibit JAK2 signaling in thrombopoietin (TPO)-dependent cell lines,(34) showing that the mutation causes a protein loss of function, making LNK a tumor suppressor gene. Primary samples from patients harboring LNK mutations presented increased STAT3/5 activation, suggesting that the loss of function caused by LNK is analogous to JAK2 or MPL gain of function mutations.(35) Mice homozygous for LNK deficiency display a phenotype characterized by splenomegaly, extramedullary hematopoiesis and increased numbers of hematopoietic precursors. Heterozygous mice display a similar but milder phenotype consistent with a haploinsufficiency model.(36)

Mutations associated with transcriptional regulation

Gene transcription is a complex cellular function that relies on the coordinated functioning of different classes of proteins, such as RNA polymerases, transcription factors, histone deacetylases and DNA methyltransferases.(37) Recently, recurring mutations in genes involved in transcriptional regulation have been described in myeloid diseases.(38-40) In MNs, the most prevalent mutations in this group of genes are found in additional sex combs-like 1 (ASXL1), enhancer of zeste homolog 2 (EZH2), TET2 and IDH1/2.

Additional sex combs-like 1 (ASXL1)

ASXL1 is a member of the polycomb group of proteins involved in transcriptional repression and activation. ASXL1 represses retinoic acid receptor-mediated transcription via its N-terminal region by the modulation of histone 3 demethylation.(41) Mutations in ASXL1 are mainly heterozygous, nonsense and frameshift, occurring predominantly in exon 12 and disrupting the C-terminal region of the protein. ASXL1 mutations are found in 32%, 36%, 50% and 18% of patients with PMF, post-PV MF, post-ET MF and post-MN AML, respectively,(42,43) but only rarely found in ET and PV.(42,44) If the mutations generate loss or gain of function is still unknown. The prognostic significance of ASXL1 mutations is not established in the literature.(43) In animal models, loss of function of ASXL1 expression caused only mild defects in hematopoiesis, suggesting it may have a modulating, rather than causal effect in MNs.(45)

Enhancer of zeste homolog 2 (EZH2)

EZH2, like ASXL1, is a member of the polycomb group of proteins. It is part of a protein complex (called polycomb repressive complex 2) which is a histone 3 methyltransferase associated with transcriptional repression.(46) EZH2 mutations in myeloid diseases can be homozygous or heterozygous, nonsense or frameshift, resulting, in most cases, in truncation of the protein.(40) Additionally, it has been shown that EZH2 mutations in myeloid malignancies cause absence of histone 3 methylation at Lysine 27, revealing a loss-of-function phenotype compatible with a tumor suppressor function for EZH2.(40) Mutations in EZH2 are present in 13% and 3% of patients with MF (primary or secondary) and PV, respectively.(40) Moreover, the presence of EZH2 mutations has recently been associated with decreased overall survival in patients with PMF.(20)

Tet oncogene family member 2 (TET2)

TET2 encodes a ketoglutarate-dependent methylcytosine dioxygenase that converts methylcytosine to 5-hydroxylmethylcytosine (5-HMC).(47) 5-HMC is widely distributed in all tissues, but its precise function is unknown.(48) There is some evidence suggesting that 5-HMC could be either an intermediate step in the process of cytosine demethylation(49) or a modification associated with gene de-repression, without going through demethylation.(50) TET2 mutations occur over all exons and can be missense or nonsense substitutions, splice site, insertions or deletions resulting in frameshift or stop codons. Most mutations are expected to result in loss of protein function thus classifying TET2 as a tumor suppressor gene. Additionally, the mutations can be heterozygous, homozygous or compound heterozygous.(47,51) The prevalences of TET2 mutations are 5%, 16%, 17% and 17% in ET, PV, MF and blastic phase MNs, respectively.(52) TET2 knockout mice present reductions in 5-HMC levels and develop myeloid malignancies resembling MNs, myelodysplastic syndromes and AML.(53) The presence of TET2 mutations has added no prognostic significance in MNs in most studies.(54)

Isocitrate dehydrogenase 1 and 2 (IDH1/2)

IDH1 and 2 are enzymes that participate in the citric acid cycle catalyzing the conversion of isocitrate to a-ketoglutarate. Mutations in either IDH1 or IDH2 are present in: 0.8%, 1.9%, 4.2% and 11% to 21% of ET, PV, PMF and blast-phase MNs, respectively. IDH1 and IDH2 mutations are mutually exclusive and affect three specific arginine residues: IDH1-R132, IDH2-R140 and IDH2-R172.(55,56) It has been shown that IDH1/2 mutations are associated to lower leukemia-free survival and overall survival in patients with JAK2V617F PMF.(57) In addition, the documentation of chronic phase IDH mutations in two patients that progressed to AML suggests that IDH may be used as a surrogate marker for AML progression.(57) Mutated IDH1/2 have a neo-enzymatic activity. Instead of converting isocitrate to a-ketoglutarate, the mutated enzyme converts a-ketoglutarate to 2-hydroxyglutarate, resulting in a reduction of a-ketoglutarate (essential for TET2 activity) and an accumulation of 2-hydroxyglutarate in the neoplastic cells.(58-60) Moreover, recent evidence suggests that 2-hydroxyglutarate is an inhibitor of a-ketoglutarate.(61) In conclusion, these findings suggest a mechanistic link between IDH and TET2, in which IDH mutations would cause (like TET2 mutations) decreased TET2 activity and may explain the low incidence of concurrent mutations in IDH and TET2.(55,61,62)

Mutations associated with disease progression to AML

Mutations associated with disease progression are, by definition, found more frequently in post-MN AML and rarely in chronic phase disease. It has yet to be determined if these mutations happen before disease acceleration, contributing to it, or only at the time of progression to AML. If the former proves to be the case in large cohorts, these mutations could serve as surrogate markers for transformation and consequently could help in the indication of more aggressive treatment strategies such as hematopoietic stem cell transplantation. The genes whose mutations are associated with blastic phase MNs are RUNX1, TP53, IKZF1, LNK and IDH1/2. Since LNK and IDH1/2 have already been discussed, we will now focus on the former three.

Runt-related transcription factor 1 (RUNX1)

The protein encoded by this gene is the alpha subunit of the transcription factor called core binding factor (CBF). CBF is essential for hematopoiesis and its disruption by either translocations or point mutations is a frequent event in AML and MDS.(63) It has been shown that RUNX1 C-terminal mutations impair the DNA-damage repair response.(64) RUNX1 mutations in chronic phase MNs are rare(63,65) but its acquisition in blastic phase of patients with wild type RUNX1 has been documented in 27% (5 out of 18) of patients.(65) In another study, 37.5% (6 out of 16) of post-MN AML patients were positive for RUNX1 mutations.(66) The mutations are mostly located in the RUNT domain, are heterozygous and are probably associated with loss of function.(66)

Tumor protein p53 (TP53)

TP53 encodes a tumor suppressor gene that has a central role in the cellular response to stress. It can activate DNA repair, induce cell cycle arrest, apoptosis and senescence after genomic damage. TP53 is one of the most frequently mutated genes in a range of cancers. TP53 mutations are rarely found in chronic phase MNs, but they have been found in 20% (4 out of 16) of post-MN AML patients.(66) The mutations cause loss of function of the protein and in all cases both alleles were affected. In another study, TP53 was found mutated in 27.3% and 3.1% of post-MN AML and chronic phase (heterozygous mutations) patients, respectively.(67) In both studies, TP53 mutations were documented in the chronic phase before progression to AML, suggesting that it may be useful as a predictor of progression to AML. Additionally, amplifications of chromosome 1q have been documented in 0.32% and 18.18% of patients with chronic phase and post-MN AML.(67) The amplified region harbored MDM4, an inhibitor of p53, amplified in other malignancies.(68,69) In this study, TP53 mutations and 1q amplifications were mutually exclusive, suggesting a similar role for both genetic changes. In total, 45.5% of the patients with post-MN AML had an abnormality in either TP53 or chromosome 1q.(67)

IKAROS family zinc finger 1 (IKZF1)

IKZF1 encodes a transcription factor associated with chromatin remodeling, essential for normal lymphopoiesis. Mutations in IKZF1 have been described in lymphoid malignancies, with deletions being particularly frequent in Ph+ acute lymphoblastic leukemia (ALL).(70) IKZF1 deletions are present in less than 1% and 20% of chronic phase and post-MN AML, respectively.(71) The absence of IKZF1 deletions in the chronic phase of three patients with AML suggests that IKZF1 mutations may be a late event in the progression to AML and therefore not useful as a surrogate marker.(71)

Discussion

Medicine is evolving towards the complete molecular characterization of diseases. In few years, all the DNA-associated changes, both genetic or epigenetic, causing diseases will be characterized and consequently used for diagnostic, prognostic stratification or therapeutic purposes. With the advent of the new generation of DNA sequencers, the rate of discovery of new mutations has been greatly accelerated, and our understanding regarding the molecular events associated with distinct phenotypes has followed the same trend. It is reasonable to believe that in the near future, with the identification of other mutations, we will be able to detect the multiple molecular events associated with each disease in the majority of patients. The challenge now is to define disease entities that are better associated with prognosis and response to treatment than the classic histopathological classification could predict. Regarding MNs, the use of RUNX1, TP53 and IDH mutations will probably be included soon in the clinical evaluation, since they may predict transformation to AML and therefore the need for more aggressive treatment.(57,65-67,72) Additionally, mutated IDH1 and 2 are good candidates for targeted therapy, since both are neo-enzymes with definite functions.

In summary, the molecular events causing MNs and the progression to AML are just starting to be unveiled. The complete characterization of all steps involved in this process will substantially change disease classification and certainly lead to better treatment strategies.

References

1. Swerdlow SH, Campo, E, Harris NL, Jaffe ES, Pileri SA, Stein H, et al. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 5th ed. Geneva, WHO; 2008. (IARC WHO Classification of Tumours n. 2).

2. James C, Ugo V, Le Couedic JP, Staerk J, Delhommeau F, Lacout C, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature. 2005;434(7037):1144-8.

3. Levine RL, Wadleigh M, Cools J, Ebert BL, Wernig G, Huntly BJ, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 2005;7(4):387-97.

4. Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, Swanton S, Vassiliou GS, Bench AJ, Boyd EM, Curtin EM, Scott MA, Erber WN, Green AR; Cancer Genome Project. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet. 2005;365(9464):1054-61. Erratum in: Lancet. 2005;366(9480):122.

5. Kralovics R, Passamonti F, Buser AS, Teo SS, Tiedt R, Passweg JR, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med. 2005;352(17):1779-90. Comment in: N Engl J Med. 2005;353(13):1416-7; author reply 1416-7. N Engl J Med. 2005;352(17):1744-6.

6. Martinez-Aviles L, Besses C, Alvarez-Larran A, Cervantes F, Hernandez-Boluda JC, Bellosillo B. JAK2 exon 12 mutations in polycythemia vera or idiopathic erythrocytosis. Haematologica. 2007;92(12):1717-8.

7. Pardanani AD, Levine RL, Lasho T, Pikman Y, Mesa RA, Wadleigh M, et al. MPL515 mutations in myeloproliferative and other myeloid disorders: a study of 1182 patients. Blood. 2006;108(10):3472-6.

8. Ward AC, Touw I, Yoshimura A. The Jak-Stat pathway in normal and perturbed hematopoiesis. Blood. 2000;95(1):19-29.

9. Rawlings JS, Rosler KM, Harrison DA. The JAK/STAT signaling pathway. J Cell Sci. 2004;117(Pt 8):1281-3.

10. Bunting KD. Another Lnk to STAT activation. Blood. 2010;116(6):862-4. Comment on: Blood. 2010;116(6):988-92.

11. Kisseleva T, Bhattacharya S, Braunstein J, Schindler CW. Signaling through the JAK/STAT pathway, recent advances and future challenges. Gene. 2002;285(1-2):1-24.

12. Skoda R. The genetic basis of myeloproliferative disorders. Hematology Am Soc Hematol Educ Program. 2007:1-10.

13. Akada H, Yan D, Zou H, Fiering S, Hutchison RE, Mohi MG. Conditional expression of heterozygous or homozygous Jak2V617F from its endogenous promoter induces a polycythemia vera-like disease. Blood. 2010;115(17):3589-97.

14. 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(7):2435-7.

15. Harrison C. Rethinking disease definitions and therapeutic strategies in essential thrombocythemia and polycythemia vera. Hematology Am Soc Hematol Educ Program. 2010;2010:129-34.

16. Scott LM, Tong W, Levine RL, Scott MA, Beer PA, Stratton MR, et al. JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. N Engl J Med. 2007;356(5):459-68. Comment in: N Engl J Med. 2007;356(5):444-5.

17. Passamonti F, Elena C, Schnittger S, Skoda RC, Green AR, Girodon F, et al. Molecular and clinical features of the myeloproliferative neoplasm associated with JAK2 exon 12 mutations. Blood. 2011;117(10):2813-6.

18. Wolanskyj AP, Lasho TL, Schwager SM, McClure RF, Wadleigh M, Lee SJ, et al. JAK2 mutation in essential thrombocythaemia: clinical associations and long-term prognostic relevance. Br J Haematol. 2005;131(2):208-13.

19. Passamonti F, Rumi E, Pietra D, Elena C, Boveri E, Arcaini L, et al. A prospective study of 338 patients with polycythemia vera: the impact of JAK2 (V617F) allele burden and leukocytosis on fibrotic or leukemic disease transformation and vascular complications. Leukemia. 2010;24(9):1574-9.

20. Guglielmelli P, Biamonte F, Score J, Hidalgo-Curtis C, Cervantes F, Maffioli M, et al. EZH2 mutational status predicts poor survival in myelofibrosis. Blood. 2011;118(19):5227-34.

21. Tefferi A, Lasho TL, Huang J, Finke C, Mesa RA, Li CY, et al. Low JAK2V617F allele burden in primary myelofibrosis, compared to either a higher allele burden or unmutated status, is associated with inferior overall and leukemia-free survival. Leukemia. 2008;22(4):756-61.

22. Vannucchi AM, Antonioli E, Guglielmelli P, Pancrazzi A, Guerini V, Barosi G, et al. Characteristics and clinical correlates of MPL 515W>L/K mutation in essential thrombocythemia. Blood. 2008;112(3):844-7.

23. Pikman Y, Lee BH, Mercher T, McDowell E, Ebert BL, Gozo M, et al. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med. 2006;3(7):e270.

24. Swaminathan G, Tsygankov AY. The Cbl family proteins: ring leaders in regulation of cell signaling. J Cell Physiol. 2006;209(1):21-43.

25. Schmidt MH, Dikic I. The Cbl interactome and its functions. Nat Rev Mol Cell Biol. 2005;6(12):907-18.

26. Grand FH, Hidalgo-Curtis CE, Ernst T, Zoi K, Zoi C, McGuire C, et al. Frequent CBL mutations associated with 11q acquired uniparental disomy in myeloproliferative neoplasms. Blood. 2009; 113(24):6182-92.

27. Reindl C, Quentmeier H, Petropoulos K, Greif PA, Benthaus T, Argiropoulos B, et al. CBL exon 8/9 mutants activate the FLT3 pathway and cluster in core binding factor/11q deletion acute myeloid leukemia/myelodysplastic syndrome subtypes. Clin Cancer Res. 2009;15(7):2238-47.

28. Grossmann V, Kohlmann A, Eder C, Haferlach C, Kern W, Cross NC, et al. Molecular profiling of chronic myelomonocytic leukemia reveals diverse mutations in > 80% of patients with TET2 and EZH2 being of high prognostic relevance. Leukemia. 2011;25(5):877-9.

29. Sanada M, Suzuki T, Shih LY, Otsu M, Kato M, Yamazaki S, et al. Gain-of-function of mutated C-CBL tumour suppressor in myeloid neoplasms. Nature. 2009;460(7257):904-8. Comment in: Nature. 2009;460(7257):804-7.

30. Gery S, Cao Q, Gueller S, Xing H, Tefferi A, Koeffler HP. Lnk inhibits myeloproliferative disorder-associated JAK2 mutant, JAK2V617F. J Leukoc Biol. 2009;85(6):957-65.

31. Tong W, Zhang J, Lodish HF. Lnk inhibits erythropoiesis and Epo-dependent JAK2 activation and downstream signaling pathways. Blood. 2005;105(12):4604-12.

32. Gery S, Gueller S, Chumakova K, Kawamata N, Liu L, Koeffler HP. Adaptor protein Lnk negatively regulates the mutant MPL, MPLW515L associated with myeloproliferative disorders. Blood. 2007;110(9):3360-4.

33. Pardanani A, Lasho T, Finke C, Oh ST, Gotlib J, Tefferi A. LNK mutation studies in blast-phase myeloproliferative neoplasms, and in chronic-phase disease with TET2, IDH, JAK2 or MPL mutations. Leukemia. 2010;24(10):1713-8.

34. Oh ST, Simonds EF, Jones C, Hale MB, Goltsev Y, Gibbs KD, Jr., et al. Novel mutations in the inhibitory adaptor protein LNK drive JAK-STAT signaling in patients with myeloproliferative neoplasms. Blood. 2010;116(6):988-92. Comment in: Blood. 2010;116(6): 862-4.

35. Lasho TL, Pardanani A, Tefferi A. LNK mutations in JAK2 mutation-negative erythrocytosis. N Engl J Med. 2010;363(12):1189-90.

36. Velazquez L, Cheng AM, Fleming HE, Furlonger C, Vesely S, Bernstein A, et al. Cytokine signaling and hematopoietic homeostasis are disrupted in Lnk-deficient mice. J Exp Med. 2002;195(12):1599-611.

37. Roeder RG. Transcriptional regulation and the role of diverse coactivators in animal cells. FEBS Lett. 2005;579(4):909-15.

38. Delhommeau F, Dupont S, Della Valle V, James C, Trannoy S, Masse A, et al. Mutation in TET2 in myeloid cancers. N Engl J Med. 2009;360(22):2289-301. Comment in: N Engl J Med. 2009;361(11):1117; author reply 1117-8. N Engl J Med. 2009;360(22):2355-7.

39. Gelsi-Boyer V, Trouplin V, Adelaide J, Bonansea J, Cervera N, Carbuccia N, et al. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br J Haematol. 2009;145(6):788-800.

40. Ernst T, Chase AJ, Score J, Hidalgo-Curtis CE, Bryant C, Jones AV, et al. Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nature genetics. 2010;42(8):722-6.

41. Lee SW, Cho YS, Na JM, Park UH, Kang M, Kim EJ, et al. ASXL1 represses retinoic acid receptor-mediated transcription through associating with HP1 and LSD1. J Biol Chem. 2010;285(1):18-29.

42. Stein BL, Williams DM, O'Keefe C, Rogers O, Ingersoll RG, Spivak JL, et al. Disruption of the ASXL1 gene is frequent in primary, post-essential thrombocytosis and post-polycythemia vera myelofibrosis, but not essential thrombocytosis or polycythemia vera: analysis of molecular genetics and clinical phenotypes. Haematologica. 2011; 96(10):1462-9. Comment in: Haematologica. 2011;96(10):1398-402.

43. Abdel-Wahab O, Pardanani A, Patel J, Wadleigh M, Lasho T, Heguy A, et al. Concomitant analysis of EZH2 and ASXL1 mutations in myelofibrosis, chronic myelomonocytic leukemia and blast-phase myeloproliferative neoplasms. Leukemia. 2011;25(7):1200-2.

44. Carbuccia N, Murati A, Trouplin V, Brecqueville M, Adelaide J, Rey J, et al. Mutations of ASXL1 gene in myeloproliferative neoplasms. Leukemia. 2009;23(11):2183-6.

45. Fisher CL, Pineault N, Brookes C, Helgason CD, Ohta H, Bodner C, et al. Loss-of-function Additional sex combs like 1 mutations disrupt hematopoiesis but do not cause severe myelodysplasia or leukemia. Blood. 2010;115(1):38-46.

46. Simon JA, Lange CA. Roles of the EZH2 histone methyltransferase in cancer epigenetics. Mutat Res. 2008;647(1-2):21-9.

47. Ko M, Huang Y, Jankowska AM, Pape UJ, Tahiliani M, Bandukwala HS, et al. Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2. Nature. 2010;468(7325):839-43.

48. Globisch D, Munzel M, Muller M, Michalakis S, Wagner M, Koch S, et al. Tissue distribution of 5-hydroxymethylcytosine and search for active demethylation intermediates. PloS One. 2010;5(12):e15367.

49. Guo JU, Su Y, Zhong C, Ming GL, Song H. Hydroxylation of 5-methylcytosine by TET1 promotes active DNA demethylation in the adult brain. Cell. 2011;145(3):423-34.

50. Wu SC, Zhang Y. Active DNA demethylation: many roads lead to Rome. Nat Rev Mol Cell Biol. 2010 Sep;11(9):607-20. Erratum in: Nat Rev Mol Cell Biol. 2010;11(10):750.

51. Bacher U, Haferlach C, Schnittger S, Kohlmann A, Kern W, Haferlach T. Mutations of the TET2 and CBL genes: novel molecular markers in myeloid malignancies. Ann Hematol. 2010;89(7):643-52.

52. Tefferi A, Pardanani A, Lim KH, Abdel-Wahab O, Lasho TL, Patel J, et al. TET2 mutations and their clinical correlates in polycythemia vera, essential thrombocythemia and myelofibrosis. Leukemia. 2009;23(5):905-11.

53. Li Z, Cai X, Cai CL, Wang J, Zhang W, Petersen BE, et al. Deletion of Tet2 in mice leads to dysregulated hematopoietic stem cells and subsequent development of myeloid malignancies. Blood. 2011; 118(17):4509-18. Comment in: Blood. 2011;118(17):4501-3.

54. Tefferi A. Novel mutations and their functional and clinical relevance in myeloproliferative neoplasms: JAK2, MPL, TET2, ASXL1, CBL, IDH and IKZF1. Leukemia. 2010;24(6):1128-38.

55. Tefferi A, Lasho TL, Abdel-Wahab O, Guglielmelli P, Patel J, Caramazza D, et al. IDH1 and IDH2 mutation studies in 1473 patients with chronic-, fibrotic- or blast-phase essential thrombocythemia, polycythemia vera or myelofibrosis. Leukemia. 2010;24(7):1302-9.

56. Pardanani A, Lasho TL, Finke CM, Mai M, McClure RF, Tefferi A. IDH1 and IDH2 mutation analysis in chronic- and blast-phase myeloproliferative neoplasms. Leukemia. 2010;24(6):1146-51.

57. Tefferi A, Jimma T, Sulai NH, Lasho TL, Finke CM, Knudson RA, et al. IDH mutations in primary myelofibrosis predict leukemic transformation and shortened survival: clinical evidence for leukemogenic collaboration with JAK2V617F. Leukemia. 2011 Sep 13. doi: 10.1038/leu.2011.253. [Epub ahead of print]

58. Dang L, White DW, Gross S, Bennett BD, Bittinger MA, Driggers EM, et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature. 2009;462(7274):739-44.

59. Ward PS, Patel J, Wise DR, Abdel-Wahab O, Bennett BD, Coller HA, 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(3):225-34. Comment in: Cancer Cell. 2010;17(3):215-6.

60. Gross S, Cairns RA, Minden MD, Driggers EM, Bittinger MA, Jang HG, et al. Cancer-associated metabolite 2-hydroxyglutarate accumulates in acute myelogenous leukemia with isocitrate dehydrogenase 1 and 2 mutations. J Exp Med. 2010;207(2):339-44. Comment in: J Exp Med. 2010; 207(2):265. J Exp Med. 2010;207(4):677-80.

61. Xu W, Yang H, Liu Y, Yang Y, Wang P, Kim SH, et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases. Cancer Cell. 2011;19(1):17-30.

62. Abdel-Wahab O, Manshouri T, Patel J, Harris K, Yao J, Hedvat C, et al. Genetic analysis of transforming events that convert chronic myeloproliferative neoplasms to leukemias. Cancer Res. 2010;70(2):447-52.

63. Steensma DP, Gibbons RJ, Mesa RA, Tefferi A, Higgs DR. Somatic point mutations in RUNX1/CBFA2/AML1 are common in high-risk myelodysplastic syndrome, but not in myelofibrosis with myeloid metaplasia. Eur J Haematol. 2005;74(1):47-53.

64. Satoh Y, Matsumura I, Tanaka H, Harada H, Harada Y, Matsui K, et al. C-terminal mutation of RUNX1 attenuates the DNA-damage repair response in hematopoietic stem cells. Leukemia. 2012;26(2):303-11.

65. Ding Y, Harada Y, Imagawa J, Kimura A, Harada H. AML1/RUNX1 point mutation possibly promotes leukemic transformation in myeloproliferative neoplasms. Blood. 2009;114(25):5201-5.

66. Beer PA, Delhommeau F, LeCouedic JP, Dawson MA, Chen E, Bareford D, et al. Two routes to leukemic transformation after a JAK2 mutation-positive myeloproliferative neoplasm. Blood. 2010;115(14):2891-900. Comment in: Blood. 2010;115(14):2727-8.

67. Harutyunyan A, Klampfl T, Cazzola M, Kralovics R. p53 lesions in leukemic transformation. N Engl J Med. 2011;364(5):488-90.

68. Laurie NA, Donovan SL, Shih CS, Zhang J, Mills N, Fuller C, et al. Inactivation of the p53 pathway in retinoblastoma. Nature. 2006;444(7115):61-6. Comment in: Nature. 2006;444(7115):45-6.

69. Riemenschneider MJ, Buschges R, Wolter M, Reifenberger J, Bostrom J, Kraus JA, et al. Amplification and overexpression of the MDM4 (MDMX) gene from 1q32 in a subset of malignant gliomas without TP53 mutation or MDM2 amplification. Cancer Res. 1999;59(24):6091-6.

70. Mullighan CG, Miller CB, Radtke I, Phillips LA, Dalton J, Ma J, et al. BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros. Nature. 2008;453(7191):110-4.

71. Jager R, Gisslinger H, Passamonti F, Rumi E, Berg T, Gisslinger B, et al. Deletions of the transcription factor Ikaros in myeloproliferative neoplasms. Leukemia. 2010;24(7):1290-8.

72. Green A, Beer P. Somatic mutations of IDH1 and IDH2 in the leukemic transformation of myeloproliferative neoplasms. N Engl J Med. 2010;362(4):369-70. Comment on: N Engl J Med. 2009;361(11):1058-66.

Submitted: 10/18/2011

Accepted: 12/21/2011

Conflict-of-interest disclosure: The authors declare no competing financial interest

www.rbhh.org or www.scielo.br/rbhh

  • 1. Swerdlow SH, Campo, E, Harris NL, Jaffe ES, Pileri SA, Stein H, et al. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 5th ed. Geneva, WHO; 2008. (IARC WHO Classification of Tumours n. 2).
  • 2. James C, Ugo V, Le Couedic JP, Staerk J, Delhommeau F, Lacout C, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature. 2005;434(7037):1144-8.
  • 3. Levine RL, Wadleigh M, Cools J, Ebert BL, Wernig G, Huntly BJ, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 2005;7(4):387-97.
  • 4. Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, Swanton S, Vassiliou GS, Bench AJ, Boyd EM, Curtin EM, Scott MA, Erber WN, Green AR; Cancer Genome Project. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet. 2005;365(9464):1054-61. Erratum in: Lancet. 2005;366(9480):122.
  • 5. Kralovics R, Passamonti F, Buser AS, Teo SS, Tiedt R, Passweg JR, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med. 2005;352(17):1779-90. Comment in: N Engl J Med.
  • 2005;353(13):1416-7;
  • author reply 1416-7.
  • N Engl J Med. 2005;352(17):1744-6.
  • 6. Martinez-Aviles L, Besses C, Alvarez-Larran A, Cervantes F, Hernandez-Boluda JC, Bellosillo B. JAK2 exon 12 mutations in polycythemia vera or idiopathic erythrocytosis. Haematologica. 2007;92(12):1717-8.
  • 7. Pardanani AD, Levine RL, Lasho T, Pikman Y, Mesa RA, Wadleigh M, et al. MPL515 mutations in myeloproliferative and other myeloid disorders: a study of 1182 patients. Blood. 2006;108(10):3472-6.
  • 8. Ward AC, Touw I, Yoshimura A. The Jak-Stat pathway in normal and perturbed hematopoiesis. Blood. 2000;95(1):19-29.
  • 9. Rawlings JS, Rosler KM, Harrison DA. The JAK/STAT signaling pathway. J Cell Sci. 2004;117(Pt 8):1281-3.
  • 10. Bunting KD. Another Lnk to STAT activation. Blood. 2010;116(6):862-4.
  • Comment on: Blood. 2010;116(6):988-92.
  • 11. Kisseleva T, Bhattacharya S, Braunstein J, Schindler CW. Signaling through the JAK/STAT pathway, recent advances and future challenges. Gene. 2002;285(1-2):1-24.
  • 12. Skoda R. The genetic basis of myeloproliferative disorders. Hematology Am Soc Hematol Educ Program. 2007:1-10.
  • 13. Akada H, Yan D, Zou H, Fiering S, Hutchison RE, Mohi MG. Conditional expression of heterozygous or homozygous Jak2V617F from its endogenous promoter induces a polycythemia vera-like disease. Blood. 2010;115(17):3589-97.
  • 14. 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(7):2435-7.
  • 15. Harrison C. Rethinking disease definitions and therapeutic strategies in essential thrombocythemia and polycythemia vera. Hematology Am Soc Hematol Educ Program. 2010;2010:129-34.
  • 16. Scott LM, Tong W, Levine RL, Scott MA, Beer PA, Stratton MR, et al. JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. N Engl J Med. 2007;356(5):459-68.
  • Comment in: N Engl J Med. 2007;356(5):444-5.
  • 17. Passamonti F, Elena C, Schnittger S, Skoda RC, Green AR, Girodon F, et al. Molecular and clinical features of the myeloproliferative neoplasm associated with JAK2 exon 12 mutations. Blood. 2011;117(10):2813-6.
  • 18. Wolanskyj AP, Lasho TL, Schwager SM, McClure RF, Wadleigh M, Lee SJ, et al. JAK2 mutation in essential thrombocythaemia: clinical associations and long-term prognostic relevance. Br J Haematol. 2005;131(2):208-13.
  • 19. Passamonti F, Rumi E, Pietra D, Elena C, Boveri E, Arcaini L, et al. A prospective study of 338 patients with polycythemia vera: the impact of JAK2 (V617F) allele burden and leukocytosis on fibrotic or leukemic disease transformation and vascular complications. Leukemia. 2010;24(9):1574-9.
  • 20. Guglielmelli P, Biamonte F, Score J, Hidalgo-Curtis C, Cervantes F, Maffioli M, et al. EZH2 mutational status predicts poor survival in myelofibrosis. Blood. 2011;118(19):5227-34.
  • 21. Tefferi A, Lasho TL, Huang J, Finke C, Mesa RA, Li CY, et al. Low JAK2V617F allele burden in primary myelofibrosis, compared to either a higher allele burden or unmutated status, is associated with inferior overall and leukemia-free survival. Leukemia. 2008;22(4):756-61.
  • 22. Vannucchi AM, Antonioli E, Guglielmelli P, Pancrazzi A, Guerini V, Barosi G, et al. Characteristics and clinical correlates of MPL 515W>L/K mutation in essential thrombocythemia. Blood. 2008;112(3):844-7.
  • 23. Pikman Y, Lee BH, Mercher T, McDowell E, Ebert BL, Gozo M, et al. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med. 2006;3(7):e270.
  • 24. Swaminathan G, Tsygankov AY. The Cbl family proteins: ring leaders in regulation of cell signaling. J Cell Physiol. 2006;209(1):21-43.
  • 25. Schmidt MH, Dikic I. The Cbl interactome and its functions. Nat Rev Mol Cell Biol. 2005;6(12):907-18.
  • 26. Grand FH, Hidalgo-Curtis CE, Ernst T, Zoi K, Zoi C, McGuire C, et al. Frequent CBL mutations associated with 11q acquired uniparental disomy in myeloproliferative neoplasms. Blood. 2009; 113(24):6182-92.
  • 27. Reindl C, Quentmeier H, Petropoulos K, Greif PA, Benthaus T, Argiropoulos B, et al. CBL exon 8/9 mutants activate the FLT3 pathway and cluster in core binding factor/11q deletion acute myeloid leukemia/myelodysplastic syndrome subtypes. Clin Cancer Res. 2009;15(7):2238-47.
  • 28. Grossmann V, Kohlmann A, Eder C, Haferlach C, Kern W, Cross NC, et al. Molecular profiling of chronic myelomonocytic leukemia reveals diverse mutations in > 80% of patients with TET2 and EZH2 being of high prognostic relevance. Leukemia. 2011;25(5):877-9.
  • 29. Sanada M, Suzuki T, Shih LY, Otsu M, Kato M, Yamazaki S, et al. Gain-of-function of mutated C-CBL tumour suppressor in myeloid neoplasms. Nature. 2009;460(7257):904-8.
  • Comment in: Nature. 2009;460(7257):804-7.
  • 30. Gery S, Cao Q, Gueller S, Xing H, Tefferi A, Koeffler HP. Lnk inhibits myeloproliferative disorder-associated JAK2 mutant, JAK2V617F. J Leukoc Biol. 2009;85(6):957-65.
  • 31. Tong W, Zhang J, Lodish HF. Lnk inhibits erythropoiesis and Epo-dependent JAK2 activation and downstream signaling pathways. Blood. 2005;105(12):4604-12.
  • 32. Gery S, Gueller S, Chumakova K, Kawamata N, Liu L, Koeffler HP. Adaptor protein Lnk negatively regulates the mutant MPL, MPLW515L associated with myeloproliferative disorders. Blood. 2007;110(9):3360-4.
  • 33. Pardanani A, Lasho T, Finke C, Oh ST, Gotlib J, Tefferi A. LNK mutation studies in blast-phase myeloproliferative neoplasms, and in chronic-phase disease with TET2, IDH, JAK2 or MPL mutations. Leukemia. 2010;24(10):1713-8.
  • 34. Oh ST, Simonds EF, Jones C, Hale MB, Goltsev Y, Gibbs KD, Jr., et al. Novel mutations in the inhibitory adaptor protein LNK drive JAK-STAT signaling in patients with myeloproliferative neoplasms. Blood. 2010;116(6):988-92.
  • Comment in: Blood. 2010;116(6): 862-4.
  • 35. Lasho TL, Pardanani A, Tefferi A. LNK mutations in JAK2 mutation-negative erythrocytosis. N Engl J Med. 2010;363(12):1189-90.
  • 36. Velazquez L, Cheng AM, Fleming HE, Furlonger C, Vesely S, Bernstein A, et al. Cytokine signaling and hematopoietic homeostasis are disrupted in Lnk-deficient mice. J Exp Med. 2002;195(12):1599-611.
  • 37. Roeder RG. Transcriptional regulation and the role of diverse coactivators in animal cells. FEBS Lett. 2005;579(4):909-15.
  • 38. Delhommeau F, Dupont S, Della Valle V, James C, Trannoy S, Masse A, et al. Mutation in TET2 in myeloid cancers. N Engl J Med. 2009;360(22):2289-301.
  • Comment in: N Engl J Med. 2009;361(11):1117; author reply 1117-8.
  • N Engl J Med. 2009;360(22):2355-7.
  • 39. Gelsi-Boyer V, Trouplin V, Adelaide J, Bonansea J, Cervera N, Carbuccia N, et al. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br J Haematol. 2009;145(6):788-800.
  • 40. Ernst T, Chase AJ, Score J, Hidalgo-Curtis CE, Bryant C, Jones AV, et al. Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nature genetics. 2010;42(8):722-6.
  • 41. Lee SW, Cho YS, Na JM, Park UH, Kang M, Kim EJ, et al. ASXL1 represses retinoic acid receptor-mediated transcription through associating with HP1 and LSD1. J Biol Chem. 2010;285(1):18-29.
  • 42. Stein BL, Williams DM, O'Keefe C, Rogers O, Ingersoll RG, Spivak JL, et al. Disruption of the ASXL1 gene is frequent in primary, post-essential thrombocytosis and post-polycythemia vera myelofibrosis, but not essential thrombocytosis or polycythemia vera: analysis of molecular genetics and clinical phenotypes. Haematologica. 2011; 96(10):1462-9.
  • Comment in: Haematologica. 2011;96(10):1398-402.
  • 43. Abdel-Wahab O, Pardanani A, Patel J, Wadleigh M, Lasho T, Heguy A, et al. Concomitant analysis of EZH2 and ASXL1 mutations in myelofibrosis, chronic myelomonocytic leukemia and blast-phase myeloproliferative neoplasms. Leukemia. 2011;25(7):1200-2.
  • 44. Carbuccia N, Murati A, Trouplin V, Brecqueville M, Adelaide J, Rey J, et al. Mutations of ASXL1 gene in myeloproliferative neoplasms. Leukemia. 2009;23(11):2183-6.
  • 45. Fisher CL, Pineault N, Brookes C, Helgason CD, Ohta H, Bodner C, et al. Loss-of-function Additional sex combs like 1 mutations disrupt hematopoiesis but do not cause severe myelodysplasia or leukemia. Blood. 2010;115(1):38-46.
  • 46. Simon JA, Lange CA. Roles of the EZH2 histone methyltransferase in cancer epigenetics. Mutat Res. 2008;647(1-2):21-9.
  • 47. Ko M, Huang Y, Jankowska AM, Pape UJ, Tahiliani M, Bandukwala HS, et al. Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2. Nature. 2010;468(7325):839-43.
  • 48. Globisch D, Munzel M, Muller M, Michalakis S, Wagner M, Koch S, et al. Tissue distribution of 5-hydroxymethylcytosine and search for active demethylation intermediates. PloS One. 2010;5(12):e15367.
  • 49. Guo JU, Su Y, Zhong C, Ming GL, Song H. Hydroxylation of 5-methylcytosine by TET1 promotes active DNA demethylation in the adult brain. Cell. 2011;145(3):423-34.
  • 50. Wu SC, Zhang Y. Active DNA demethylation: many roads lead to Rome. Nat Rev Mol Cell Biol. 2010 Sep;11(9):607-20.
  • Erratum in: Nat Rev Mol Cell Biol. 2010;11(10):750.
  • 51. Bacher U, Haferlach C, Schnittger S, Kohlmann A, Kern W, Haferlach T. Mutations of the TET2 and CBL genes: novel molecular markers in myeloid malignancies. Ann Hematol. 2010;89(7):643-52.
  • 52. Tefferi A, Pardanani A, Lim KH, Abdel-Wahab O, Lasho TL, Patel J, et al. TET2 mutations and their clinical correlates in polycythemia vera, essential thrombocythemia and myelofibrosis. Leukemia. 2009;23(5):905-11.
  • 53. Li Z, Cai X, Cai CL, Wang J, Zhang W, Petersen BE, et al. Deletion of Tet2 in mice leads to dysregulated hematopoietic stem cells and subsequent development of myeloid malignancies. Blood. 2011; 118(17):4509-18.
  • Comment in: Blood. 2011;118(17):4501-3.
  • 54. Tefferi A. Novel mutations and their functional and clinical relevance in myeloproliferative neoplasms: JAK2, MPL, TET2, ASXL1, CBL, IDH and IKZF1. Leukemia. 2010;24(6):1128-38.
  • 55. Tefferi A, Lasho TL, Abdel-Wahab O, Guglielmelli P, Patel J, Caramazza D, et al. IDH1 and IDH2 mutation studies in 1473 patients with chronic-, fibrotic- or blast-phase essential thrombocythemia, polycythemia vera or myelofibrosis. Leukemia. 2010;24(7):1302-9.
  • 56. Pardanani A, Lasho TL, Finke CM, Mai M, McClure RF, Tefferi A. IDH1 and IDH2 mutation analysis in chronic- and blast-phase myeloproliferative neoplasms. Leukemia. 2010;24(6):1146-51.
  • 57. Tefferi A, Jimma T, Sulai NH, Lasho TL, Finke CM, Knudson RA, et al. IDH mutations in primary myelofibrosis predict leukemic transformation and shortened survival: clinical evidence for leukemogenic collaboration with JAK2V617F. Leukemia. 2011 Sep 13. doi: 10.1038/leu.2011.253. [Epub ahead of print]
  • 58. Dang L, White DW, Gross S, Bennett BD, Bittinger MA, Driggers EM, et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature. 2009;462(7274):739-44.
  • 59. Ward PS, Patel J, Wise DR, Abdel-Wahab O, Bennett BD, Coller HA, 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(3):225-34.
  • Comment in: Cancer Cell. 2010;17(3):215-6.
  • 60. Gross S, Cairns RA, Minden MD, Driggers EM, Bittinger MA, Jang HG, et al. Cancer-associated metabolite 2-hydroxyglutarate accumulates in acute myelogenous leukemia with isocitrate dehydrogenase 1 and 2 mutations. J Exp Med. 2010;207(2):339-44.
  • Comment in: J Exp Med. 2010; 207(2):265.
  • J Exp Med. 2010;207(4):677-80.
  • 61. Xu W, Yang H, Liu Y, Yang Y, Wang P, Kim SH, et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases. Cancer Cell. 2011;19(1):17-30.
  • 62. Abdel-Wahab O, Manshouri T, Patel J, Harris K, Yao J, Hedvat C, et al. Genetic analysis of transforming events that convert chronic myeloproliferative neoplasms to leukemias. Cancer Res. 2010;70(2):447-52.
  • 63. Steensma DP, Gibbons RJ, Mesa RA, Tefferi A, Higgs DR. Somatic point mutations in RUNX1/CBFA2/AML1 are common in high-risk myelodysplastic syndrome, but not in myelofibrosis with myeloid metaplasia. Eur J Haematol. 2005;74(1):47-53.
  • 64. Satoh Y, Matsumura I, Tanaka H, Harada H, Harada Y, Matsui K, et al. C-terminal mutation of RUNX1 attenuates the DNA-damage repair response in hematopoietic stem cells. Leukemia. 2012;26(2):303-11.
  • 65. Ding Y, Harada Y, Imagawa J, Kimura A, Harada H. AML1/RUNX1 point mutation possibly promotes leukemic transformation in myeloproliferative neoplasms. Blood. 2009;114(25):5201-5.
  • 66. Beer PA, Delhommeau F, LeCouedic JP, Dawson MA, Chen E, Bareford D, et al. Two routes to leukemic transformation after a JAK2 mutation-positive myeloproliferative neoplasm. Blood. 2010;115(14):2891-900.
  • Comment in: Blood. 2010;115(14):2727-8.
  • 67. Harutyunyan A, Klampfl T, Cazzola M, Kralovics R. p53 lesions in leukemic transformation. N Engl J Med. 2011;364(5):488-90.
  • 68. Laurie NA, Donovan SL, Shih CS, Zhang J, Mills N, Fuller C, et al. Inactivation of the p53 pathway in retinoblastoma. Nature. 2006;444(7115):61-6.
  • Comment in: Nature. 2006;444(7115):45-6.
  • 69. Riemenschneider MJ, Buschges R, Wolter M, Reifenberger J, Bostrom J, Kraus JA, et al. Amplification and overexpression of the MDM4 (MDMX) gene from 1q32 in a subset of malignant gliomas without TP53 mutation or MDM2 amplification. Cancer Res. 1999;59(24):6091-6.
  • 70. Mullighan CG, Miller CB, Radtke I, Phillips LA, Dalton J, Ma J, et al. BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros. Nature. 2008;453(7191):110-4.
  • 71. Jager R, Gisslinger H, Passamonti F, Rumi E, Berg T, Gisslinger B, et al. Deletions of the transcription factor Ikaros in myeloproliferative neoplasms. Leukemia. 2010;24(7):1290-8.
  • 72. Green A, Beer P. Somatic mutations of IDH1 and IDH2 in the leukemic transformation of myeloproliferative neoplasms. N Engl J Med. 2010;362(4):369-70.
  • Comment on: N Engl J Med. 2009;361(11):1058-66.
  • Corresponding author:

    Paulo Vidal Campregher
    Hospital Israelita Albert Einstein. Laboratório de Técnicas Especializadas
    Avenida Albert Einstein, 627/701Bloco D, 1º andar
    05652-900 - São Paulo, SP, Brazil
    Phone: 55 11 5475-2000
  • Publication Dates

    • Publication in this collection
      11 May 2012
    • Date of issue
      2012

    History

    • Received
      18 Oct 2011
    • Accepted
      21 Dec 2011
    Associação Brasileira de Hematologia e Hemoterapia e Terapia Celular R. Dr. Diogo de Faria, 775 cj 114, 04037-002 São Paulo/SP/Brasil, Tel. (55 11) 2369-7767/2338-6764 - São Paulo - SP - Brazil
    E-mail: secretaria@rbhh.org