Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Nov 20.
Published in final edited form as: Leukemia. 2009 Mar 5;23(5):900–904. doi: 10.1038/leu.2009.37

Frequent TET2 mutations in systemic mastocytosis: clinical, KITD816V and FIP1L1-PDGFRA correlates

A Tefferi 1, RL Levine 2,3, K-H Lim 1,4, O Abdel-Wahab 2,3, TL Lasho 1, J Patel 2, CM Finke 1, A Mullally 5, C-Y Li 1, A Pardanani 1, DG Gilliland 5,6
PMCID: PMC4654631  NIHMSID: NIHMS735618  PMID: 19262599

Abstract

TET2 (TET oncogene family member 2) is a candidate tumor suppressor gene located at chromosome 4q24, and was recently reported to be mutated in ~14% of patients with JAK2 V617F-positive myeloproliferative neoplasms. We used high-throughput DNA sequence analysis to screen for TET2 mutations in bone marrow-derived DNA from 48 patients with systemic mastocytosis (SM), including 42 who met the 2008 WHO (World Health Organization) diagnostic criteria for SM and 6 with FIP1L1-PDGFRA. Twelve (29%) SM, but no FIP1L1-PDGFRA patients, had TET2 mutations. A total of 17 mutations (13 frameshift, 2 nonsense and 2 missense) were documented in 2 (15%) of 13 indolent SM patients, 2 (40%) of 5 aggressive SM, and 8 (35%) of 23 SM associated with a clonal non-mast cell-lineage hematopoietic disease (P = 0.52). KITD816V was detected by PCR sequencing in 50 or 20% of patients with or without TET2 mutation (P = 0.05), respectively. Multivariable analysis showed a significant association between the presence of TET2 mutation and monocytosis (P = 0.0003) or female sex (P = 0.05). The association with monocytosis was also observed in non-indolent SM (n = 29), in which the presence of mutant TET2 did not affect survival (P = 0.98). We conclude that TET2 mutations are frequent in SM, segregate with KIT D816V and influence phenotype without necessarily altering prognosis.

Keywords: KIT, PDGFRA, JAK2, MPL, myeloproliferative, eosinophilia

Introduction

Delhommeau et al.1 recently reported an acquired loss of heterozygosity and somatic deletions at chromosome 4q24 in patients with myeloproliferative neoplasm (MPN). These findings are in consonance with a report by Viguie et al.2 describing four female patients with acute myeloid leukemia or myelodysplastic syndrome and a 4q24 rearrangement; t(3;4)(q26;q24), t(4;5)(q24;p16), t(4;7)(q24;q21) and del(4)(q23;q24).2 The specific cytogenetic abnormality was present in both myeloid and lymphoid lineage cells, as well as in lymphoma cells from one of the four patients who developed concomitant malignant lymphoma. Fluorescence in situ hybridization with bacterial artificial chromosome clones identified a 0.5-Mb commonly deleted region that accompanied the 4q24 breakpoint, and this microdeletion was subsequently observed in another patient with normal karyotype.2 These observations were interpreted as indicating a stem cell-derived molecular event involving a putative tumor suppressor gene located at 4q24.2

Delhommeau et al.1 showed that TET2 (TET oncogene family member 2), contained in the minimal loss of heterozygosity region of 4q24, harbored loss-of-function mutations in patients with myeloid malignancy, including a stop codon mutation in exon 3 and a 9 nucleotide deletion in exon 6. Subsequently, the authors sequenced TET2 in 181 JAK2 V617F-positive patients with polycythemia vera, essential thrombocythemia and primary myelofibrosis, and identified a spectrum of putative loss-of-function mutations, with an overall mutational frequency of 14%.1 Additional studies showed that these mutations often involved both copies of the TET2 gene, consistent with a tumor suppressor function for the wild-type alleles, occurred in both multipotent and committed progenitors, and antedated acquisition of the JAK2 V617F allele in a small cohort of informative patients.1 Here we report that loss-of-function mutations in TET2 occur at a high frequency in systemic mastocytosis (SM), and are associated with KITD816V mutations. Collectively, these observations indicate that acquired TET2 mutations contribute to pathogenesis of a spectrum of myeloid malignancies that are associated with activating mutations in tyrosine kinases.3

Material and methods

After approval by the institutional review board, the Mayo Clinic database of adult SM patients (age ≥18 years) was utilized to select consecutive patients in whom stored bone marrow (BM) cells were available for DNA extraction and mutation analysis. All study patients met the 2001 WHO (World Health Organization) diagnostic criteria for SM4 but data analysis was adjusted by considering FIP1L1-PDGFRA -positive patients separately, in line with the 2008 WHO criteria.3 Patients were seen at the Mayo Clinic between January 1976 and October 2007 and were required to have BM pathology reviewed on site. Special stains, including tryptase and KIT, were used to identify mast cells in biopsy material and BM mast cell burden and other aspects of BM histology were recorded for each case. Cytogenetic analysis and mutation screening for KIT D816V (PCR sequencing) and JAK2 V617F (reverse transcriptase-PCR) were performed using BM-derived cells, according to the earlier published methods.57

M13-appended gene-specific primers were designed to amplify and sequence all coding exons of all isoforms of TET2. In all, 1 µg of genomic DNA was used to amplify all exons of TET2, followed by bidirectional sequencing (Agencourt Bioscience, Beverly, MA, USA). Mutations were detected in bidirectional sequence traces using Mutation Surveyor (Soft-genetics, Inc., State College, PA, USA), and all traces were manually reviewed for the presence of frameshift mutations. Frameshift and nonsense mutations were annotated according to the predicted effects on the TET2 coding sequence (Table 2). Non-synonymous mutations were annotated as somatic mutations on the basis of sequence analysis of paired MPN/myelodysplastic syndrome samples that showed that these mutations were present in tumor and not in matched normal DNA.

Table 2.

Clinical and laboratory details of 12 systemic mastocytosis patients with TET2 mutation

SM type Exon Nucleotide change Consequence Age Sex Hgb g/
100 ml
WBC
×109/L
Plt.
×109/L
AMC
×109/L
JAK2
V617F
KIT
D816V
Cyto.
ISM 4 1777_1778insG Frameshift 78 F 9.9 3.2 238 0.4 Neg. Pos. Normal
ISM 12 723_724delAG Frameshift 64 F 12.7 5.0 202 0.6 Neg. Neg. Normal
ASM 4 3248_delT Frameshift 69 M 12.6 6.5 149 1.6 Neg. Neg. -Y
8 278_279insA Frameshift
ASM 4 1554_delG Frameshift 80 F 10.6 21.5 64 2.8 Neg. Pos. Normal
SM-CMML-1 12 1305A>G H1881R 83 M 15.0 26.3 202 1.3 Pos. Neg. 13q-
SM-CMML-1 4 2628C>T Nonsense 70 F 12.4 5.1 153 1.4 Neg. Pos. Normal
SM- 10 231 C>T R1358C 69 F 8.9 21.5 72 8.3 Neg. Neg. Normal
CMML-1 11 287_delA Frameshift
SM-CMML-1 12 695_696insT Frameshift 75 M 10.8 51.1 12 11.2 Neg. Pos. Normal
SM- 4 3070_3071insA Frameshift 73 F 7.9 11.0 115 1.3 Neg. Neg. Normal
RAEB-1 12 955_956insT Frameshift
SM- 4 2812_2813insT Frameshift 50 M 8.7 8.2 97 NA Neg. Pos. −7
RCMD 12 252_262delCTCTACAGAAG Frameshift
SM-MPN 8 236_delC Frameshift 53 M 8.8 73.4 54 2.9 Neg. Neg. Normal
SM-MPN 4 2468_2469insA Frameshift 61 F 14.5 16.4 33 0.7 Neg. Pos. Normal
6 208 C>T Nonsense

Abbreviations: AMC, absolute monocyte count; ASM, aggressive SM; CMML, chronic myelomonocytic leukemia; Cyto., bone marrow cytogenetic results; Hgb, hemoglobin level; ISM, indolent SM; MPN, myeloproliferative neoplasm; NA, not assessed; neg., negative; Plt., platelet count; pos., positive; RAEB, refractory anemia with excess blasts; RCMD, refractory cytopenia with multilineage dysplasia; SM, systemic mastocytosis; WBC, white blood cell count.

Statistical procedures utilized were conventional and all data were analyzed by using StatView (SAS Institute, Cary, NC, USA). All P-values were two-tailed and statistical significance was set at the level of P<0.05. Clinical and laboratory correlative studies considered values obtained at the time of diagnosis or referral to the Mayo Clinic. Continuous variables were summarized as medians and ranges. Categorical variables were described as count and relative frequency (%). Comparison between categorical variables was performed by the χ2 statistics. Comparison between categorical and continuous variables was performed by either the Mann–Whitney U test or Kruskal–Wallis test. The association of variables selected from univariate analysis was explored using logistic regression models. Survival curves for the patients with and without TET2 mutation were constructed by Kaplan–Meier method, taking the interval from the date of diagnosis to death or last contact, and compared using the log-rank test.

Results

Forty-eight patients were included in the current study, including 42 who fulfilled the 2008 WHO diagnostic criteria for SM (Table 1) and the six with FIP1L1-PDGFRA fusion gene that contributes to eosinophilia associated with mastocytosis. SM disease subclassification included 13 patients with indolent SM (ISM), 5 with aggressive SM (ASM), 23 with SM associated with a clonal non-mast cell-lineage hematopoietic disease (SM-AHNMD) and one with mast cell leukemia (MCL). Presenting clinical and laboratory features for each of these SM subcategories are outlined in Table 1. Overall, 12 (29%) of the 42 patients with WHO-defined SM, but none of the 6 with FIP1L1-PDGFRA, displayed a TET2 mutation. Furthermore, 5 of the 12 TET2 -mutated patients had additional TET2 mutations (all involving different exons) bringing the total to 17 mutations, including 13 frameshift, 2 nonsense and 2 missense; the location of these mutations (i.e. exon number), the specific nucleotide changes and their consequences are detailed in Table 2. Exons 4 and 12 were the most frequently affected regions.

Table 1.

Clinical and laboratory features of 42 patients with World Health Organization-defined systemic mastocytosis

All patients TET2
mutated
TET2
unmutated
ISM ASM SM-AHNMD
N (%) 42 (100) 12 (29) 30 (71) 13 (31) 5 (12) 23 (55)
Males, n (%) 27 (64) 5 (42) 22 (73) 8 (62) 3 (60) 15 (65)
Age in years, median (range) 64.5 (28–87) 69.5 (50–83) 62 (28–87) 57 (28–78) 69 (43–80) 67 (34–87)
UP, n (%) 12 (29) 2 (17) 10 (33) 8 (62) 0 4 (17)
MCMRSa, n (%) 19 (45) 2 (17) 17 (57) 9 (69) 2 (40) 7 (30)
Splenomegaly, n (%) 14 (33) 7 (58) 7 (23) 0 1 (20) 12 (52)
AML transformation, n (%) 4 (10) 1 (8) 3 (10) 0 0 4 (17)
Hemoglobin (g/dl), median (range) 11.9 (7.3–16) 10.7 (7.9–15) 12.3 (7.3–16) 14.3 (9.9–15.2) 10.8 (10.5–16) 9.7 (7.3–15.9)
Leukocyte count (× 109/l), median (range) 8.3 (1.2–73.4) 13.7 (3.2–73.4) 8.3 (1.2–21.3) 6.6 (1.6–9.9) 11.1(6.5–21.5) 9.1 (1.2–73.4)
Monocyte count (× 109/l), median (range) 0.6 (0–11.2) 1.4 (0.4–11.2) 0.4 (0–4) 0.4 (0–0.7) 0.6 (0.4–2.8) 0.7 (0–11.2)
Eosinophil count (× 109/l), median (range) 0.3 (0–9.2) 0.1 (0–8.8) 0.3 (0–9.2) 0.2 (0–0.5) 0.4 (0–9.2) 0.3 (0–8.8)
Platelet count (× 109/l), median (range) 179 (12–431) 106 (12–238) 204.5 (14–431) 233 (39–431) 149 (20–345) 115 (12–392)
Serum tryptase (ng/ml), median (range) n evaluated = 26 55.8 (13.1–1450) 179 (13.1–674) 47.2 (13.2–1450) 38 (13.1–440) 66.5 (29–1450) 73.7 (13.2–674)
BM mast cell %, median (range) 10 (5–70) 20 (5–30) 10 (5–70) 10 (5–25) 17.5 (10–30) 10 (5–30)
BM blast % ≥5, n (%) 6 (14) 2 (17) 4 (13) 0 0 6 (26)
Abnormal karyotype, n (%), n evaluated = 39 11 (28) 3 (25) 8 (30) 1 (9) 1 (20) 9 (41)
KITD816V, n (%) 12 (29) 6 (50) 6 (20) 2 (15) 2 (40) 8 (35)
JAK2V617F, n (%) 2 (5) 1 (8) 1 (3) 0 0 2 (9)

Abbreviations: AHNMD, associated hematologic non-mast cell lineage disease; AML, acute myeloid leukemia; ASM, aggressive SM; BM, bone marrow; ISM, indolent SM; MCMRS, mast cell mediator release symptoms; SM, systemic mastocytosis; UP, urticaria pigmentosa.

a

Included headache, dizziness/light-headedness, syncope/pre-syncope, hypotension, anaphylaxis, palpitation/tachycardia, bronchoconstriction/wheezing, and peptic ulcer disease. One of the 42 study patients had mast cell leukemia.

TET2 mutational frequencies for ISM, ASM and SM-AHNMD were 15% (2 of 13), 40% (2 of 5) and 35% (8 of 23) (P = 0.52). Among the 23 patients with SM-AHNMD, the non-mast cell-lineage hematopoietic disease was chronic myelomonocytic leukemia in 8, myelodysplastic syndrome in 6, MPN in 6 and lymphoid neoplasm in 3; the corresponding TET2 mutational frequencies were 50, 33, 33 and 0% (P = 0.5). Six (50%) of the 12 TET2-mutated patients also displayed KIT D816V and one JAK2 V617F (low allele burden). Only one other patient (also with SM-AHNMD) expressed JAK2 V617F. The single patient with MCL was negative for all the three mutations; TET2, KIT and JAK2. KIT D816V was not detected in the 6 patients with FIP1L1-PDGFRA.

Correlations of TET2 mutation presence with several clinical and laboratory parameters were performed in the 42 patients with WHO-defined SM, as well as in the subgroup of 29 patients with non-indolent SM (Table 3). Multivariable analysis of all the 42 patients showed that monocytosis and female sex remained significant in their association with the presence of TET2 mutation. A similar analysis restricted to the 29 patients with non-indolent SM (i.e. SM-AHNMD, ASM and MCL) validated the significant association between monocytosis and the presence of TET2 mutation. The presence of mutant TET2 did not affect the survival of patients with non-indolent SM (Figure 1). Median follow-up in this latter group of patients was 14 months (range 0.1–69). ISM patients were excluded from the survival analysis because of their well-known longevity; regardless, the two TET2 mutated ISM patients in the current study were both female and remain alive and well after a follow-up period of 15–22 months.

Table 3.

P-values during univariate analysis correlating the presence of TET2 mutations with clinical and laboratory features at presentation

Variables (continuous) Variables (categorical) Patients with WHO-defined
SM (N = 42)
Patients with non-indolent SM
(SM-AHNMD/ASM/MCL) (N = 29)
Age 0.02 0.19
Female gender 0.05 0.20
SM subgroup 0.52 0.74
Palpable spleen 0.03 0.09
Hemoglobin level 0.23 0.99
Hemoglobin <10 g/100 ml 0.47 0.70
Leukocyte count 0.09 0.03
Leukocyte >10 × 109/l 0.02 0.05
Platelet count 0.05 0.15
Platelet <100 × 109/l 0.09 0.14
Monocyte count 0.002 0.005
Monocyte >1 × 109/l 0.0003 0.001
Eosinophil count 0.68 0.64
Serum tryptase level (n = 27) 0.18 0.10
BM mast cell content 0.07 0.06
BM blasts ≥5% 0.78 0.95
KITD816V 0.05 0.20
Abnormal karyotype 0.50 0.68

Abbreviations: AHNMD, associated hematologic non-mast cell lineage disease; ASM, aggressive SM; BM, bone marrow; MCL, mast cell leukemia; SM, systemic mastocytosis; WHO, World Health Organization.

Bold, italic and underline entries are statistically significant.

Figure 1.

Figure 1

Survival curves of TET2 mutated and unmutated patients with non-indolent systemic mastocytosis (n = 29).

Discussion

Primarily on the basis of its stem cell-derived clonal heritage,8 SM is currently classified with polycythemia vera, essential thrombocythemia and primary myelofibrosis, under the MPN category of myeloid neoplasms.3 Phenotypic diversity and similarity among chronic myeloid neoplasms is currently attributed to differences in their underlying molecular pathogenesis. Some molecular markers, such as JAK2 V617F, are widely shared among MPN and, albeit infrequently, are also present in SM.9,10 Others, such as BCR-ABL1 in CML, are more specific. The current study suggests that TET2 mutations transcend clinicopathological class and are not unique to JAK2 V617F-positive MPN.1 The high prevalence of TET2 mutations in SM is of particular interest because, although present in the majority of affected patients,11 KIT D816V might require the presence of a collaborating mutation that contributes to known phenotypic heterogeneity.12 The striking association between the presence of a TET2 mutation and monocytosis in the current study lends further support to this contention. Furthermore, the existence of KIT -independent recurrent mutations in SM might explain the limited clinical activity of KIT-targeted therapy in this disease.13,14

World Health Organization-defined SM is further subclassified into ISM, ASM, SM-AHNMD and MCL.15 The presence of organ dysfunction (e.g. ascites, cytopenia and lytic bone lesions) distinguishes indolent from ASM. Both myeloid and lymphoid neoplasms qualify as the ‘second’ non-mast cell-lineage neoplasm in SM-AHNMD whereas a diagnosis of MCL requires the presence of ≥10% circulating mast cells. In the current study, there was only one patient with MCL and he was negative for both TET2 mutation and KIT D816V. Obviously, additional comments regarding TET2 mutations in MCL, based on this single case, are not warranted but KIT D816V-negative MCL has been reported earlier by others as well.11 The demonstration of TET2 mutations in ISM, with a mutational frequency that was not significantly different than that seen with the more aggressive disease variants, undermines but does not exclude their role as markers of tumor progression. Similarly, the statistically non-significant but apparently lower associated mutational frequency might be a function of tumor burden rather than tumor aggression. This is also consistent with the lack of significant association between TET2 mutation and survival in patients with non-indolent SM. However, a larger study with longer follow-up is necessary before refuting the prognostic relevance of TET2 mutations in SM.

It has been suggested that all patients with SM carry a KIT mutation, but this may be mitigated in part by cell source and assay sensitivity.11,16 For example, among patients carrying a KIT mutation in purified mast cell preparations, the mutation is detected in less than half of the patients when other nucleated BM cells were studied.11 It may therefore not be surprising that KIT D816V was detected in less than a third of the patients in this study, as we used a direct sequencing technique of whole BM specimens that were not enriched for mast cells. As such, KIT D816V detection in the current study will reflect the degree of BM infiltration by clonal mast cells, but will lack sensitivity for mutation in the mast cell lineage. Additional studies will be needed to explore clonal distribution in terms of both TET2 and KIT mutations in early progenitor cells and specific cell compartments, including mast cells, monocytes, neutrophils and eosinophils.

The association between TET2 mutations and the female gender is of interest. Of note, the four patients with 4q24-rearranged acute myeloid leukemia/myelodysplastic syndrome, described in the original report by Viguie et al.,2 were also all females. Such gender predilection has also been observed in PDGFR-rearranged MPN, in which virtually all affected patients are male. There are no mechanistic insights into this gender predilection.17 In contrast and unlike the case with JAK2 V617F in essential thrombocythemia18 and primary myelofibrosis,19 the association between age and the detection of TET2 mutations in SM has not been verified by multivariable analysis. Whether or not these observations hold true in the setting of other myeloid neoplasms is expected to be clarified soon.

Because clonal BM mastocytosis also occurs in association with another WHO category of myeloid malignancies, FIP1L1-PDGFRA -positive myeloid neoplasm,20,21 we included six such patients in the current study, none of whom displayed a TET2 mutation. Although the number of patients studied is too small to allow one to make definitive conclusions, the particular observation reinforces the distinction between WHO-defined and FIP1L1-PDGFRA-associated SM, and is consistent with the contention that FIP1L1-PDGFRA is oncogenically more robust than KITD816V.12,22 Of note, patients with FIP1L1-PDGFRA-associated MPN are also not known to co-express JAK2 V617F.23 Taken together, and as has been pointed out earlier in the context of acute myeloid leukemia,24 the observations from the current study underscore the complexity of molecular pathogenesis in myeloid neoplasms and, in this regard, suggest a cooperative but not necessarily a primary role for TET2 mutations.

Acknowledgements

Adriana Heguay and Igor Dolgalev from the Memorial Sloan-Kettering Cancer Center, New York, NY, USA are acknowledged for their assistance with sequence analysis. The current study was supported in part by grants from the Myeloproliferative Disorders Foundation, Chicago, IL, USA.

Footnotes

Conflict of interest

The authors declare no conflict of interest.

References

  • 1.Delhommeau F, Dupont S, James C, Masse A, le Couedic JP, Valle VD, et al. TET2 is a novel tumor suppressor gene inactivated in myeloproliferative neoplasms: identification of a Pre-JAK2 V617F event. ASH Annual Meeting Abstracts. 2008;112 lba-3: Late-Breaking Abstract. [Google Scholar]
  • 2.Viguie F, Aboura A, Bouscary D, Ramond S, Delmer A, Tachdjian G, et al. Common 4q24 deletion in four cases of hematopoietic malignancy: early stem cell involvement? Leukemia. 2005;19:1411–1415. doi: 10.1038/sj.leu.2403818. [DOI] [PubMed] [Google Scholar]
  • 3.Tefferi A, Vardiman JW. Classification and diagnosis of myeloproliferative neoplasms: the 2008 World Health Organization criteria and point-of-care diagnostic algorithms. Leukemia. 2008;22:14–22. doi: 10.1038/sj.leu.2404955. [DOI] [PubMed] [Google Scholar]
  • 4.Jaffe ES, Harris NL, Stein H, Vardiman JW. World Health Organization Classification of Tumours of Hematopoietic and Lymphoid Tissues. Lyon, France: IARC Press; 2001. pp. 1–351. [Google Scholar]
  • 5.Dewald GW, Broderick DJ, Tom WW, Hagstrom JE, Pierre RV. The efficacy of direct, 24-h culture, and mitotic synchronization methods for cytogenetic analysis of bone marrow in neoplastic hematologic disorders. Cancer Genet Cytogenet. 1985;18:1–10. doi: 10.1016/0165-4608(85)90032-9. [DOI] [PubMed] [Google Scholar]
  • 6.Pardanani A, Reeder TL, Kimlinger TK, Baek JY, Li CY, Butterfield JH, et al. Flt-3 and c-kit mutation studies in a spectrum of chronic myeloid disorders including systemic mast cell disease. Leuk Res. 2003;27:739–742. doi: 10.1016/s0145-2126(02)00303-x. [DOI] [PubMed] [Google Scholar]
  • 7.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;22:756–761. doi: 10.1038/sj.leu.2405097. [DOI] [PubMed] [Google Scholar]
  • 8.Afonja O, Amorosi E, Ashman L, Takeshita K. Multilineage involvement and erythropoietin-‘independent’ erythroid progenitor cells in a patient with systemic mastocytosis. Ann Hematol. 1998;77:183–186. doi: 10.1007/s002770050439. [DOI] [PubMed] [Google Scholar]
  • 9.Steensma DP, Dewald GW, Lasho TL, Powell HL, McClure RF, Levine RL, et al. The JAK2 V617F activating tyrosine kinase mutation is an infrequent event in both ‘atypical’ myeloproliferative disorders and myelodysplastic syndromes. Blood. 2005;106:1207–1209. doi: 10.1182/blood-2005-03-1183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Sotlar K, Bache A, Stellmacher F, Bultmann B, Valent P, Horny HP. Systemic mastocytosis associated with chronic idiopathic myelofibrosis: a distinct subtype of systemic mastocytosis associated with a (corrected) clonal hematological non-mast (corrected) cell lineage disorder carrying the activating point mutations KITD816V and JAK2V617F. J Mol Diagn. 2008;10:58–66. doi: 10.2353/jmoldx.2008.070061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Garcia-Montero AC, Jara-Acevedo M, Teodosio C, Sanchez ML, Nunez R, Prados A, 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. doi: 10.1182/blood-2006-04-015545. [DOI] [PubMed] [Google Scholar]
  • 12.Mayerhofer M, Gleixner KV, Hoelbl A, Florian S, Hoermann G, Aichberger KJ, et al. Unique effects of KIT D816V in BaF3 cells: induction of cluster formation, histamine synthesis, and early mast cell differentiation antigens. J Immunol. 2008;180:5466–5476. doi: 10.4049/jimmunol.180.8.5466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Verstovsek S, Tefferi A, Cortes J, O’Brien S, Garcia-Manero G, Pardanani A, et al. Phase II study of dasatinib in Philadelphia chromosome-negative acute and chronic myeloid diseases, including systemic mastocytosis. Clin Cancer Res. 2008;14:3906–3915. doi: 10.1158/1078-0432.CCR-08-0366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gotlib J, Berube C, Growney JD, Chen CC, George TI, Williams C, et al. Activity of the tyrosine kinase inhibitor PKC412 in a patient with mast cell leukemia with the D816V KIT mutation. Blood. 2005;106:2865–2870. doi: 10.1182/blood-2005-04-1568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Valent P, Horny HP, Escribano L, Longley BJ, Li CY, Schwartz LB, et al. Diagnostic criteria and classification of mastocytosis: a consensus proposal. Leuk Res. 2001;25:603–625. doi: 10.1016/s0145-2126(01)00038-8. [DOI] [PubMed] [Google Scholar]
  • 16.Kahler C, Didlaukat S, Feller AC, Merz H. Sensitive and reliable detection of Kit point mutation Asp 816 to Val in pathological material. Diagn Pathol. 2007;2:37. doi: 10.1186/1746-1596-2-37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Mertens F, Johansson B, Mitelman F. Age- and gender-related heterogeneity of cancer chromosome aberrations. Cancer Genet Cytogenet. 1993;70:6–11. doi: 10.1016/0165-4608(93)90123-4. [DOI] [PubMed] [Google Scholar]
  • 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;131:208–213. doi: 10.1111/j.1365-2141.2005.05764.x. [DOI] [PubMed] [Google Scholar]
  • 19.Tefferi A, Lasho TL, Schwager SM, Steensma DP, Mesa RA, Li CY, et al. The JAK2 tyrosine kinase mutation in myelofibrosis with myeloid metaplasia: lineage specificity and clinical correlates. Br J Haematol. 2005;131:320–328. doi: 10.1111/j.1365-2141.2005.05776.x. [DOI] [PubMed] [Google Scholar]
  • 20.Cools J, DeAngelo DJ, Gotlib J, Stover EH, Legare RD, Cortes J, et al. A tyrosine kinase created by fusion of the PDGFRA and FIP1L1 genes as a therapeutic target of imatinib in idiopathic hypereosinophilic syndrome. N Engl J Med. 2003;348:1201–1214. doi: 10.1056/NEJMoa025217. [DOI] [PubMed] [Google Scholar]
  • 21.Pardanani A, Brockman SR, Paternoster SF, Flynn HC, Ketterling RP, Lasho TL, et al. FIP1L1-PDGFRA fusion: prevalence and clinicopathologic correlates in 89 consecutive patients with moderate to severe eosinophilia. Blood. 2004;104:3038–3045. doi: 10.1182/blood-2004-03-0787. [DOI] [PubMed] [Google Scholar]
  • 22.Gotlib J, Cools J. Five years since the discovery of FIP1L1-PDGFRA: what we have learned about the fusion and other molecularly defined eosinophilias. Leukemia. 2008;22:1999–2010. doi: 10.1038/leu.2008.287. [DOI] [PubMed] [Google Scholar]
  • 23.Jones AV, Kreil S, Zoi K, Waghorn K, Curtis C, Zhang L, et al. Widespread occurrence of the JAK2 V617F mutation in chronic myeloproliferative disorders. Blood. 2005;106:2162–2168. doi: 10.1182/blood-2005-03-1320. [DOI] [PubMed] [Google Scholar]
  • 24.Deguchi K, Gilliland DG. Cooperativity between mutations in tyrosine kinases and in hematopoietic transcription factors in AML. Leukemia. 2002;16:740–744. doi: 10.1038/sj.leu.2402500. [DOI] [PubMed] [Google Scholar]

RESOURCES