ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Overview of the myeloproliferative neoplasms

Overview of the myeloproliferative neoplasms
Author:
Ayalew Tefferi, MD
Section Editor:
Richard A Larson, MD
Deputy Editor:
Alan G Rosmarin, MD
Literature review current through: Jan 2024.
This topic last updated: Sep 13, 2022.

INTRODUCTION — An overview of the four classic myeloproliferative neoplasms (MPN): polycythemia vera, essential thrombocythemia, primary myelofibrosis, and chronic myeloid leukemia will be presented here. Detailed information on each of these disorders is presented separately.

(See "Clinical manifestations and diagnosis of polycythemia vera".)

(See "Essential thrombocythemia: Treatment and prognosis".)

(See "Clinical manifestations and diagnosis of primary myelofibrosis".)

(See "Clinical manifestations and diagnosis of chronic myeloid leukemia".)

CLASSIFICATION OF HEMATOLOGIC MALIGNANCIES — The hematopoietic pluripotent stem cell is capable of both self-renewal and stepwise differentiation into either the lymphoid or myeloid lineage (which appears to be determined stochastically) [1]. Thus, during normal hematopoiesis, there exists a cellular hierarchy headed by a stable population of pluripotent stem cells that generate lineage-specific progenitors, which differentiate into the various types of mature blood cells [2]. Effective hematopoiesis is facilitated by interactions of hematopoietic growth factors, various receptors, and the bone marrow microenvironment.

An operational classification of hematologic malignancies distinguishes lymphoid from myeloid neoplasms (table 1 and table 2); in turn, each of these categories is classified as either acute or chronic, depending on the proportion of morphologically and immunophenotypically immature precursors (blasts) in the bone marrow or peripheral blood. Thus, the myeloid hematologic malignancies are divided into acute myeloid leukemia (AML; bone marrow or peripheral blood blasts of 20 percent or more) and the chronic myeloid disorders (bone marrow or peripheral blood blasts less than 20 percent). However, in the presence of certain recurrent cytogenetic abnormalities (eg, t(8;21), inv(16)), a diagnosis of AML is made regardless of the blast count.

The chronic myeloid disorders — The chronic myeloid disorders encompass several clinicopathologic entities. Conceptually, they can be organized into those that display significant morphologic dysplasia in the erythroid and/or granulocyte lineages (the myelodysplastic syndromes and the myelodysplastic/myeloproliferative overlap neoplasms) or those that do not display dysplastic changes (myeloproliferative neoplasms) [3,4]:

Myelodysplastic syndromes (MDS) are characterized by cellular dysplasia, variable degrees of peripheral blood cytopenias, and bone marrow hyperplasia (or less often, hypoplasia) [5]. The paradox of proliferative bone marrow together with peripheral blood cytopenias in MDS may be explained by increased intramedullary myeloid precursor cell apoptosis [6].

Myeloproliferative neoplasms (MPN), unlike MDS, usually exhibit terminal myeloid cell expansion in the peripheral blood [7]. MPNs include polycythemia vera, essential thrombocythemia, chronic myeloid leukemia (CML), primary myelofibrosis, chronic neutrophilic leukemia, and other less well defined entities such as chronic eosinophilic leukemia, not otherwise categorized.

Myelodysplastic/myeloproliferative neoplasms (MDS/MPN) include disorders that manifest both dysplastic and proliferative features. These include chronic myelomonocytic leukemia, juvenile myelomonocytic leukemia, atypical CML (aCML, BCR::ABL1 negative), MDS/MPN with ring sideroblasts and thrombocytosis, and unclassifiable MDS/MPN.

Clonal studies in the chronic myeloid disorders — Genetic and enzyme studies based upon X-chromosome inactivation patterns have revealed a multipotent progenitor cell origin for the neoplastic clone in both MDS [8] and MPN [9]. The clonal process may develop at different progenitor cell levels in individual patients, and the clonal progenitor cell may or may not involve T and B lymphocytes [10]. (See 'Potential significance' below.)

Studies using refined methods of X-linked DNA analysis have demonstrated polyclonal hematopoiesis in some patients with essential thrombocythemia and monoclonal hematopoiesis in healthy older adult females [11]. However, JAK2 V617F mutation analysis has revealed that patients with "polyclonal" essential thrombocythemia might also display the mutation [12]. Therefore, X-chromosome-based clonal assays might not be adequate in detecting a minor clonal population against a polyclonal background.

These observations underline the heterogeneity between individuals with MPNs, and raise the possibility that monoclonal hematopoiesis may antedate rather than follow the development of mutations (described below) that are associated with MPNs or MDS. (See 'Potential significance' below and "Clonal hematopoiesis of indeterminate potential (CHIP) and related disorders of clonal hematopoiesis".)

Familial disease — Familial clustering of polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF) has been well described [13-16]. Abnormalities that have been described in such families include mutations in the JAK2, MPL, CBL, TET2, CALR, or RBBP6 genes, most often somatic but occasionally germline [16-18].

In a study of 458 adult patients with apparently sporadic disease, familial involvement was noted in 8.7, 6.0, and 8.2 percent of those with PV, ET, or PMF, respectively [13].

In a population-based study from Sweden involving 11,039 patients with MPN, their 24,577 first degree relatives had significantly increased risks of PV (relative risk [RR] 5.7), ET (RR 7.4), and unclassifiable MPN (RR 7.5), and a borderline increased risk of CML (RR 1.9; 95% CI 0.9-3.8) [14].

Patients with familial disease developed similar complications and disease evolution as those with sporadic disease. Age distribution between parent and offspring and telomere length shortening provided evidence for disease anticipation in one study [13]. However, this age difference between parent and offspring was not seen in the large population-based study from Sweden [14].

Presence of the JAK2 mutation does not appear to represent the genetic predisposing factor [13,19-23], although it might indicate which family members are at higher risk of developing one of the MPNs at a future date [24]. (See "Molecular pathogenesis of congenital erythrocytoses and polycythemia vera", section on 'Familial PV'.)

Diagnostic criteria for PV and ET may not pertain to children, owing to known mutations in several other genes (eg, hereditary mutations of the erythropoietin receptor, thrombopoietin, MPL genes) in families with polycythemia or ET presenting in childhood [25]. (See "Clinical manifestations, pathogenesis, and diagnosis of essential thrombocythemia", section on 'Familial essential thrombocythemia' and "Molecular pathogenesis of congenital erythrocytoses and polycythemia vera", section on 'Other disorders'.)

THE CLASSIC MYELOPROLIFERATIVE DISEASES

Diagnosis — Among the four classic MPNs, only chronic myeloid leukemia (CML) is genetically characterized by reciprocal translocation between chromosomes 9 and 22; this t(9;22) translocation is associated with a shortened chromosome 22 (the Philadelphia chromosome) in 95 percent of the cases [26]. In the remaining cases, t(9;22) can be demonstrated by either fluorescence in situ hybridization (FISH) or reverse transcriptase polymerase chain reaction techniques for detection of BCR::ABL1. While cytogenetic abnormalities are common in primary myelofibrosis (approximately 30 percent at time of diagnosis) and uncommon in essential thrombocythemia (approximately 5 percent at time of diagnosis), no specific cytogenetic abnormality has been associated with MPNs other than CML [27]. (See "Molecular genetics of chronic myeloid leukemia".)

The discovery that JAK2 mutations are found in virtually all patients with polycythemia vera (PV) and approximately 50 percent of those with either essential thrombocythemia (ET) or primary myelofibrosis (PMF) has refined diagnostic criteria in the classic MPNs [4,28-31]:

PV is considered to be present when an otherwise unexplained increased hematocrit/red blood cell mass is accompanied by the presence of a JAK2 mutation (table 3). (See "Clinical manifestations and diagnosis of polycythemia vera".)

PMF (agnogenic myeloid metaplasia, chronic idiopathic myelofibrosis) is characterized by the presence of bone marrow fibrosis that cannot be attributed to another myeloid disorder such as CML, PV, ET, or MDS (table 4 and table 5). However, it should be noted that the morphological phenotype of PMF can exist without the presence of overt bone marrow fibrosis (ie, prefibrotic PMF). (See "Pathogenetic mechanisms in primary myelofibrosis" and "Clinical manifestations and diagnosis of primary myelofibrosis".)

ET is a diagnosis of exclusion, representing clonal or autonomous thrombocytosis not classifiable as PV, PMF, CML, or MDS (table 6). (See "Clinical manifestations, pathogenesis, and diagnosis of essential thrombocythemia".)

Within the context of the MPNs, an elevated red cell mass is specific for PV. Occasionally both CML and MDS may present with either isolated thrombocytosis suggesting ET [32], or with bone marrow fibrosis suggesting myelofibrosis [33,34]. As a result, the diagnostic evaluation of patients with suspected MPN should always include cytogenetic studies and careful morphologic examination to exclude the presence of t(9;22) (CML) and dysmyelopoiesis (myelodysplastic syndrome [MDS]), respectively.

Although most of the chronic myeloid disorders are classifiable as MDS, CML, PV, ET, or PMF, some are difficult to categorize and may be referred to as MPN-unclassifiable or MDS/MPN-unclassifiable [35,36]. In general, BCR::ABL1 is specific to CML, but rare patients expressing both BCR::ABL1 and a JAK2 mutation have been described [37,38].

Complications — The major complications of the MPNs include thrombosis, bleeding, or evolution to acute myeloid leukemia (AML) or a fibrotic phase of the disease.

Malignancies and disease transformation — As a group, the MPNs are predisposed to clonal evolution and disease transformation to AML, MDS, or fibrotic phase of the disease [39,40]. The propensity to transform into AML differs among the subgroups, being highest for CML (greater than 90 percent in the absence of effective therapy) and least for ET (less than 5 percent). Patients with PV have an approximately 10 and 25 percent chance of transforming into a myelofibrotic stage at 10 and 25 years of follow-up, respectively [41]. (See "Clinical manifestations and diagnosis of primary myelofibrosis", section on 'Transformation to acute leukemia' and "Polycythemia vera and secondary polycythemia: Treatment and prognosis".)

The risk for disease transformation in the MPNs is influenced by both disease-related and treatment-related factors. Disease-related factors associated with a higher risk of progression to myelofibrosis or AML include mutations of CALR, epigenetic regulators, splicing factors, and RAS signaling molecules [42-46].

The effect of treatment on disease transformation was assessed in a nested case-control study of a nationwide cohort of 11,039 subjects with PV, ET, and PMF that included 153 who developed AML, 9 who developed MDS, and 242 matched controls. Results included [47]:

Forty-one of the 162 patients with MPN who developed AML or MDS (25 percent) had never been exposed to alkylating agents, radioactive phosphorus (32P), or hydroxyurea, indicating a substantial propensity of the MPNs to undergo disease transformation in the absence of cytoreductive treatment.

Increased risk of developing AML/MDS was associated with high exposures to 32P (odds ratio [OR] 4.6; 95% CI 2.1-9.8) and alkylating agents (OR 3.4; 95% CI 1.1-10.6), but not with exposure to hydroxyurea (OR for the highest exposure 1.3; 95% CI 0.5-3.3).

Risk of AML/MDS was increased in patients receiving two or more cytoreductive treatments (OR 2.9; 95% CI 1.4-5.9).

Results of a Danish population-based cohort study indicated an increased risk of development of both hematologic and non-hematologic malignancies in patients with PV, ET, and CML [48]:

The standardized incidence ratios for hematologic malignancies were 5.0, 3.8, and 5.2 for ET, PV, and CML, respectively.

The standardized incidence ratios for non-hematologic malignancies were 1.2, 1.4, and 1.6 for ET, PV, and CML, respectively.

The frequency of lymphoproliferative neoplasms in two series of patients with MPNs was 1.3 to 1.6 percent, with a cumulative risk of LPN development between 1 and 3 percent at 5 to 10 years of follow-up, corresponding to a standardized incidence ratio >4 [49,50]. This risk was higher in those harboring the JAK2 V617F mutation and was increased as much as 12-fold for the development of chronic lymphocytic leukemia. It has not been definitively shown that these two entities, when coexisting, arise from a common hematopoietic stem cell [51].

Despite these observations, it remains uncertain if disease transformation or development of secondary neoplasms in MPN is affected by treatment of any kind.

Treatment of MPN-associated acute leukemia — Leukemic/blast phase transformation (LT) of an MPN is difficult to treat. Current consensus is to treat with curative intent using an AML-like induction chemotherapy, followed by allogeneic hematopoietic cell transplantation (HCT). However, few reports demonstrate the value of such an approach. Two examples are discussed below:

In a single center report of 75 patients with MPN-associated LT, the two-year overall survival (OS) was 15 percent [52]. Selected patients with a reasonable fitness level achieved long-term control after chemotherapy with curative intent, followed by allogeneic HCT. Two-year OS were as follows for the following subgroups:

Treatment with non-curative regimens – 3 percent

Complete remission following chemotherapy with curative intent – 15 percent

Complete remission following chemotherapy with curative intent, followed by HCT – 47 percent

A retrospective report from the European Group for Blood and Marrow Transplantation included results from 57 patients with an initial diagnosis of PV or ET who underwent myeloablative allogeneic HCT following the development of AML. Results included the following [53]:

At the time of transplant only 22 percent were in complete remission from AML.

At a median follow-up of 13 months, estimated three-year OS and nonrelapse mortality were 28 and 29 percent, respectively. The high rate of relapse of AML following HCT (53 percent) contributed to the low OS noted in this group of patients.

Adverse prognostic features included age >55 years at the time of transplant and the use of an unrelated donor.

Thrombosis and bleeding — Another biologic complication shared among the MPNs is the risk of thrombohemorrhagic complications, which is most pronounced in PV and ET [54].

Thrombotic complications include arterial and venous thromboses, and microcirculatory disorders such as erythromelalgia (picture 1) and visual and neurologic symptoms [55]. Serial in vivo activation of leukocytes, vascular endothelium, platelets, and the coagulation system have been postulated as possible causes of these complications [56-60]. Erythromelalgia is associated with platelet consumption and platelet thrombi in the microvasculature; both the shortened platelet survival and symptoms can be ameliorated by aspirin, but not by heparin or warfarin, suggesting that a prostaglandin synthase product in platelets plays an important role [61].

Rates of arterial and venous thromboses are elevated in all categories of MPN, in all age groups, and equally in males and females, according to a population-based study of 9429 Swedish patients [62]. The incidence of thrombosis is highest shortly after diagnosis. Compared with the control population, the incidence of arterial and venous thromboses in patients with MPNs in the first three months after diagnosis was increased three- and 10-fold, respectively; the hazard ratios declined in subsequent years, but remained elevated throughout the five years of observation.

Major thrombotic events can occur in patients who otherwise have few clinical and laboratory features of PV. Examples include splanchnic vein thrombosis (eg, Budd-Chiari syndrome, portal, splenic, or mesenteric vein thrombosis) [63], in whom the ensuing portal hypertension and hypersplenism may mask the increase in blood cell counts [64-69]. PV should be suspected in patients with these diagnoses, particularly females under the age of 45. (See "Etiology of the Budd-Chiari syndrome", section on 'Myeloproliferative disorders'.)

Bleeding episodes are usually mild, with spontaneous bleeding primarily occurring in patients with high platelet counts (eg, those who develop acquired von Willebrand disease); both quantitative and qualitative changes in platelet function may contribute to bleeding [55,70]. (See "Acquired von Willebrand syndrome", section on 'Myeloproliferative neoplasms including ET'.)

Because of the variable risks of blastic transformation and thrombosis-associated deaths, OS in the MPNs ranges from a "near-normal" life expectancy in patients with ET to a median of less than five years in patients with PMF [71].

Other complications — A Danish population-based cohort study revealed an increased risk of osteoporotic bone fractures in patients with MPNs when compared with the general population, with a step-wise increasing pattern from ET (hazard ratio [HR] 1.19) to PV (HR 1.82) to CML (HR 2.67) [72]. The cause is unknown, although the presence of comorbidities (eg, smoking, alcohol-related diagnoses) did not explain the increased fracture risks.

Symptoms and quality of life — In addition to the major complications noted above (eg, thrombosis, hemorrhage, evolution to AML), patients with MPN also suffer from symptoms that may interfere with their quality of life. These issues were evaluated in an Internet-based symptom survey of 1179 patients with MPN [73]. The frequency of self-reporting for the following complaints were as follows:

Fatigue – 81 percent

Pruritus – 52 percent

Night sweats – 49 percent

Bone pain – 44 percent

Fever – 14 percent

Weight loss – 13 percent

Among each of the MPNs, fatigue was the most common complaint, being present in 85, 72, and 84 percent of those with PV, ET, and PMF, respectively. Approximately one-third of the respondents needed assistance with activities of daily living and 11 percent reported MPN-related medical disability.

A Myeloproliferative Neoplasm Symptom Assessment Form (MPN-SAF) has been devised and prospectively validated as an assessment tool for patients with MPNs embarking on clinical trials [74]. This form has been modified to include an abbreviated version (the MPN-SAF Total symptom score) that focuses on the most representative and clinically relevant MPN symptoms, which include the following 10 items: fatigue, early satiety, abdominal discomfort, inactivity, concentration problems, night sweats, itching, bone pain, fever, and weight loss [75,76]. However, there is heterogeneity of symptoms within each MPN subtype, sometimes independent of disease features or prognosis [77].

Role of hematologic growth factors — The role of hematologic growth factors in the development and/or propagation of the clonal process in MPN remains undefined. Growth factor independence of the clonal process in MPNs is suggested by the following observations:

MPNs are characterized by spontaneous hematopoietic colony formation (ie, without the addition of a growth factor) [78].

Erythropoietin (EPO) levels in patients with PV are appropriately downregulated, and studies have not revealed genetic mutations involving the EPO receptor [79]. Overexpression of Bcl-xL (an inhibitor of apoptosis) in erythroid precursors from patients with PV provides a possible explanation for how these cells can grow autonomously in the absence of EPO [80]. (See "Molecular pathogenesis of congenital erythrocytoses and polycythemia vera".)

The clonal process in ET appears to be independent of thrombopoietin (TPO) signaling, because molecular lesions have not been identified in TPO and mutations of the TPO receptor (MPL) are seen in only approximately 5 percent of patients with ET [81]. Serum TPO concentrations in patients with ET are either normal or above normal, suggesting defective feedback regulation [82]. Consistent with this observation, Mpl expression in platelets and megakaryocytes is reduced in patients with ET [83].

TPO receptor (MPL) expression in platelets and megakaryocytes is absent or markedly reduced in PV, suggesting that proliferation of platelets in PV is independent of TPO (as it is in ET) [84].

While growth factors have not been implicated in the neoplastic process, they are believed to contribute to the pathogenesis of bone marrow fibrosis associated with the MPNs. In an experiment in mice, lethal irradiation was followed by grafting of bone marrow cells infected with a retrovirus carrying murine TPO cDNA [85]. The initial phase was characterized by marked elevations in the platelet and white blood cell counts followed by a syndrome of bone marrow fibrosis, extramedullary hematopoiesis, hepatosplenomegaly, anemia, and leukemic transformation. The PMF-like illness is thought to be mediated by overproduction of transforming growth factor (TGF)-beta by megakaryocytes, which has also been implicated in human myelofibrosis [86].

Gene expression profiling and microRNAs — Gene expression profiling (GEP) and mutational analysis were used to characterize 55 patients with PV, ET, or PMF [87]. The major findings in this study included the following:

A GEP consistent with activated signaling through the JAK-STAT pathway was seen in all MPN patients, regardless of clinical phenotype (ie, PV, ET, or PMF) or mutational status (ie, JAK2 V617F or Calreticulin [CALR] mutation), consistent with a shared mechanism of transformation among the MPNs.

Transcriptional profiling discriminated subsets of MPNs based on JAK2 V617F allele burden and on the presence of CALR, ASXL1, and TET2 mutations, suggesting that these patterns might explain phenotypic differences among the three MPNs.

The role of microRNAs in the MPNs is unclear; reviews of this subject are available [88,89].

Mutations in PV, ET, and PMF

Overview — The most commonly mutated genes in PV, ET, and PMF are JAK2, CALR, and MPL; estimates of their frequency follow [46,90-95]:

PV – 100 percent JAK2 mutations (exon 14 or 12)

ET – 60 to 65 percent JAK2 mutation, 20 to 25 percent CALR mutation, 3 percent MPL mutation

PMF – 60 to 65 percent JAK2 mutation, 20 to 25 percent CALR mutation, 7 percent MPL mutation

Genomic characterization of 2035 patients with these MPNs found that one of these three mutations was the sole abnormality in 45 percent [46]. Approximately 8 to 10 percent of PMF cases and 10 to 15 percent of ET cases lack conventional mutations in JAK2, CALR, and MPL (ie, "triple negative" MPNs). Studies employing whole exome sequencing have revealed that a minority of these cases have non-canonical mutations in JAK2, MPL, and other genes (some of which are discussed below) [46,96,97]. (See 'Other mutations' below.)

JAK2 mutations — Mutations of the tyrosine kinase Janus kinase 2 (JAK2), a gene found on the short arm of chromosome 9 (9p), appear to account for hypersensitivity of hematopoietic progenitor cells in MPNs to growth factors and other cytokines [98-106]. (See "Clinical manifestations and diagnosis of polycythemia vera", section on 'JAK2 mutations' and "Molecular pathogenesis of congenital erythrocytoses and polycythemia vera", section on 'JAK2 mutations'.)

JAK2 mutations arise preferentially on a common constitutional JAK2 haplotype (variously called 46/1 or GGCC), which is found in approximately 50 percent of normal persons [107-109]. JAK2 mutations are most abundant in erythroid and myeloid cells, although its presence in other cell lines (eg, lymphoid cells of B cell, T cell, and NK cell lineages, endothelial cells) is debated [57,110-115]. In one study of 115 patients with MPN, the JAK2 mutation, when present, was found slightly more often in platelets than in granulocytes (72 versus 62 percent) [116].

Mutation in exon 14 of JAK2 substitutes a bulky phenylalanine for a conserved valine at position 617 in the JH2 or pseudokinase domain (Val617Phe, V617F), which negatively regulates the kinase domain, causing cytokine-independent activation of JAK-STAT, other pathways implicated in erythropoietin (EPO) receptor signaling, and the phosphatidylinositide-3 kinase (PI3K) pathway (which regulates apoptosis) [117,118].

Incidence — A single gain-of-function point mutation in JAK2 in either exon 14 or 12 has been identified in virtually all patients with PV, and in 60 to 65 percent of those with ET and PMF [103,119-121]. The presence of JAK2 mutations in EPO-independent erythroid colonies links this mutation to growth factor hypersensitivity [122]. Further study of JAK2 and the STAT pathway might allow for a more reliable classification/diagnostic system for MPNs and a more complete understanding of their pathophysiology [46,117,123-126].

A discussion of refractory anemia with ringed sideroblasts and thrombocytosis (RARS-T), a myelodysplastic variant with myeloproliferative features and a high frequency of the JAK2 V617F mutation, is found elsewhere. (See "Clinical manifestations, diagnosis, and classification of myelodysplastic syndromes (MDS)".)

As an example, the JAK2 V617F mutation has been found only infrequently in other chronic myeloid disorders (eg, MDS, chronic myelomonocytic leukemia, systemic mastocytosis, chronic neutrophilic leukemia, unclassifiable MDS/MPN), but has been seen in some patients with acute myeloid leukemia following a prior MPN, strengthening the specific association between this mutation and the classical BCR::ABL1-negative MPNs (ie, PV, ET, PMF) [127-131]. Of interest, one strain of transgenic mice highly expressing this JAK2 mutation developed phenotypes closely resembling ET and PV, and developed PMF-like symptoms as they aged [132].

The finding of dysregulated tyrosine kinase (TK) activity also suggests a possible therapeutic approach to these disorders, via the use of small molecule JAK2 TK inhibitors [133-139], similar to those successfully employed in CML and other malignancies. (See "Cellular and molecular biology of chronic myeloid leukemia", section on 'BCR-ABL1 signaling'.)

Allele burden — The mechanism(s) through which this single gene mutation is associated with such phenotypic variability is unknown, although gene dosage effects (allele burden), occurrence of other genetic events, and genetic instability have been postulated [140]. As an example, many patients with PV and PMF harbor homozygous JAK2 mutations, whereas most cases of ET appear to be heterozygous for this mutation [46,141]. In one study, larger numbers of homozygous-mutant erythroid colonies were associated with older age and male sex in PV, but only with female sex in ET, potentially explaining the preponderance of males with PV but of females in ET [142].

Documenting the presence of a JAK2 mutation and the allele burden may help to distinguish PV from secondary polycythemia, and ET from reactive thrombocytosis [143,144]. Mutation analysis can be done with equal sensitivity and specificity using either peripheral blood granulocytes or bone marrow [145]. Such testing may serve as a marker to measure measurable residual disease (MRD; also referred to as minimal residual disease) following treatment with interferon or hematopoietic cell transplantation [146,147], and stratify the risk of thrombosis [148] or evolution to post-PV or post-ET myelofibrosis [149]. (See "Essential thrombocythemia: Treatment and prognosis".)

As examples:

A number of studies have found the JAK2 V617F mutation in a high percentage of patients with Budd-Chiari syndrome, many of whom had negative results from standard testing for one of the MPNs. It has been suggested that patients with this syndrome, in whom no underlying disorder can be identified, be tested for this mutation. (See "Etiology of the Budd-Chiari syndrome", section on 'Myeloproliferative disorders'.)

In patients with PMF undergoing hematopoietic cell transplantation, achievement of negativity for JAK2 V617F was associated with a decreased incidence of relapse [147]. (See "Myelofibrosis (MF): Management of primary MF and secondary MF", section on 'Transplant-eligible patients'.)

In a prospective study of 26 patients with PV and 21 with ET, treatment with hydroxyurea was able to achieve a partial molecular response in 57 percent, as indicated by a significant decrease in JAK2 V617F allele load after 36 months of treatment [150]. In contrast, a control group that was not treated with hydroxyurea showed a slight increase in allele load over this same period of time.

Calreticulin (CALR) mutations — Calreticulin (also known as calregulin) is a Ca++ binding protein localized primarily in the endoplasmic reticulum (ER), but it is also found in the nucleus, cell membranes, and extracellular matrix [151]. The encoding gene (CALR) is located on chromosome 19p13.2 and contains nine exons. The C-terminal region of wild-type calreticulin includes both a calcium-binding domain and an endoplasmic reticulum retention motif.

CALR mutations have been observed in approximately 70 percent of patients with ET or PMF who do not carry a mutation in either JAK2 or MPL, and have only rarely been observed in patients with PV; all known CALR mutations involve frameshift mutations within exon 9 that generate a mutant protein with a novel C-terminus [90,91,94,152]. Normal functions of calreticulin include proper folding of newly synthesized glycoproteins within the ER and modulating calcium homeostasis [153,154]; mutations associated with ET and PMF disrupt the calcium-binding and endoplasmic reticulum retention domains. These mutations appear to alter the protein's function, resulting in cytokine-independent growth and activation of JAK/STAT signaling that is sensitive to pharmacologic JAK2 inhibition [90,155]. Mutant CALR interacts directly with the thrombopoietin receptor (MPL) resulting in constitutive activation of MPL and downstream signaling molecules in the JAK/STAT pathway [156-158]. (See "Clinical manifestations, pathogenesis, and diagnosis of essential thrombocythemia", section on 'Genetic features'.)

More than 50 different mutations in CALR have been described, but a 52 base pair deletion (type 1) or a 5 base pair insertion (type 2) account for more than 80 percent of mutations [95]. In a study of 1027 patients with ET, both CALR variants were associated with higher platelet and lower hemoglobin and leukocyte counts than those with mutant JAK2 [159]; the platelet count was significantly higher in those with the type 2 mutation than in those with the type 1 mutation, suggesting differential effects of CALR variants on thrombopoiesis.

MPL mutations — Activating mutations of MPL, which encodes the thrombopoietin receptor (TPO-R), have been found in patients with familial ET. (See "Clinical manifestations, pathogenesis, and diagnosis of essential thrombocythemia", section on 'Familial essential thrombocythemia' and "Biology and physiology of thrombopoietin", section on 'Familial thrombocythemia'.)

However, such mutations have not been commonly observed in patients with MPNs, and their overall clinical significance is unclear [160,161].

In one large study, novel somatic activating mutations MPL (W515L and W515K) were found in approximately 5 percent of patients with PMF, 1 percent of those with ET, and in none of the patients with PV [162].

In a second study, MPL mutations were seen in 16 of 143 patients (11 percent) with ET and 3 of 32 patients (9 percent) with PMF [163].

In a study of 603 patients with PMF, mutant MPL was detected in 8.1 percent and JAK2 V617F in 58 percent. Multivariate analysis disclosed no significant difference in OS or leukemia-free survival between MPL-mutated, JAK2-mutated, and JAK2/MPL-mutated groups [164].

In a study of 508 subjects with MPNs, an MPL exon 10 somatic mutation was found in 7 percent. Unlike the finding in those with the JAK2 V617F mutation, MPL mutations were not significantly associated with the JAK2 GGCC haplotype, suggesting a different genetic background for these two molecular lesions [109].

Other mutations — A large number of other somatic mutations have been found in patients with MPNs, but their contribution to the pathogenesis of MPNs is largely undefined [165-167]. Some of these mutations may be "drivers" of the malignant process, while others may be a consequence of genomic instability or related to clonal hematopoiesis of indeterminate potential (CHIP) [168-170]. (See "Clonal hematopoiesis of indeterminate potential (CHIP) and related disorders of clonal hematopoiesis", section on 'Clonal hematopoiesis of indeterminate potential (CHIP)'.)

Besides JAK2, CALR, and MPL, the genes that are most commonly mutated in PV, ET, and PMF are [46,165-167,170-176]:

TET2 (TET oncogene family member 2)

ASXL1 (Additional sex combs 1)

DNMT3A (a DNA methyltransferase)

CBL (Casitas B-lineage lymphoma)

TP53 (Tumor protein p53)

PPMD1 (p53–inducible protein phosphatase 1)

SRSF2 (Splicing factor, arginine/serine-rich 2)

U2AF1 (Splicing factor U2AF1)

KMT2C/MLL3 (Lysine N-methyltransferase 2C/mixed-lineage leukemia protein 3)

SF3B1 (Splicing factor 3b 1)

NFE2 (Nuclear factor E2)

EZH2 (histone H3 lysine 27 methyltransferase)

LNK (Src homology 2 B3, SH2B3)

IDH 1/2 (Isocitrate dehydrogenase 1/2)

IKZF1 (IKAROS family zinc finger 1)

Mutations of some of these genes have been associated with transformation to AML or other malignancies:

In one study of 422 patients with PV and ET, a polymorphism in the DNA repair gene XPD showed a strong association with both leukemic transformation (OR 4.9; 95% CI 2.0-12) and the development of non-myeloid malignancies (OR 4.2; 95% CI 1.5-12) [177].

A study of 22 genes in 53 patients with leukemic transformation after an MPN identified mutations in some of the above genes (eg, JAK2, TET2, ASXL1, IDH1/2, MPL) as well as in the serine/arginine-rich splicing factor 2 (SRSF2) [178].

Using next generation sequencing in 197 MPN patients, somatic mutations in one or more of 104 specified genes were found in 90 percent, with 37 percent carrying somatic mutations in genes other than JAK2 V617F and CALR [169]. The presence of ≥2 somatic mutations reduced overall survival and increased the risk of transformation to acute myeloid leukemia. A similar observation was made in a separate study of mutations in five prognostically-detrimental genes in 797 patients with PMF; in this study, median survivals were 12.3, 7.0, and 2.6 years, respectively for those with no, one, or two or more mutations in ASXL1, EZH2, SRSF2, IDH1, or IDH2 [179].

Potential significance — The significance of JAK2, MPL, CALR, and other mutations in the genesis of the MPNs as well as their relative roles in determining disease phenotype, leukemic transformation, and the level of involvement of stem cells in these disorders are unclear at present [113,114,164,180-185]. However, data from integrated genomic analyses suggest that patients with PV, ET, or PMF, regardless of diagnosis or JAK2 mutational status, are characterized by a distinct gene expression signature with upregulation of JAK-STAT target genes, demonstrating the central importance of this pathway in the pathogenesis of the MPNs [87]. (See 'Gene expression profiling and microRNAs' above.)

In some patients with MPNs, clonal myelopoiesis may antedate acquisition of JAK2, MPL, or CALR mutations, and the latter mutations may be acquired in a lympho-myeloid progenitor cell [46,113,186-188]. Mutations of TET2, DNMT3A, SF3B1, ASXL1, and other genes are often seen in older patients with MPNs and in individuals with CHIP, but their pathogenetic and prognostic relevance to development of MPNs is uncertain [87,171,189-192]. (See "Clonal hematopoiesis of indeterminate potential (CHIP) and related disorders of clonal hematopoiesis", section on 'Clonal hematopoiesis of indeterminate potential (CHIP)'.)

However, initial data suggest that the order in which these mutations are acquired impacts the clinical phenotype. This was illustrated in a study that determined the genetic evolution of MPNs (PV, ET, or PMF) with mutations in both JAK2 and TET2 [193]. Neoplasms that acquired the JAK2 mutation prior to the TET2 mutation were more likely to present at a younger age, have clinical features of PV, and develop thrombosis. In contrast, the presence of a TET2 mutation prior to the acquisition of a JAK2 mutation may alter the impact of the JAK2 mutation, perhaps through methylation-based silencing of genes that are normally expressed.

Homozygosity or heterozygosity for a JAK2 mutation, the presence of other mutations (MPL, CALR, or TET2 mutations), the order in which the mutations occurred, the balance between the sizes of their respective clones (ie, allele burden), MPL receptor expression, loss of heterozygosity in TP53, and/or other host genetic variations (eg, single nucleotide polymorphisms [194] or specific haplotypes within the JAK2 gene) may determine predisposition to development of one of these mutations, the clinical phenotype (eg, degree of thrombocytosis, leukocytosis, and erythrocytosis, splenic size, pruritus, transformation to myelofibrosis or AML) [141,159,169,187,195-209], as well as the response to treatment [210].

However, at this time one can make the following conclusions concerning these mutations [95]:

The diagnostic approach to the MPNs must now include evaluation of the mutation status of JAK2, MPL, and CALR. Such information will also be of value for determining prognosis as well as individual risk assessment.

Quantitative assays for the JAK2 V617F mutation are preferred because they enable better estimation of allele burden (ie, lower in ET, higher in PV, and highest in PV which has evolved into myelofibrosis) as well as molecular monitoring of treatment response [92,211]. (See 'Allele burden' above.)

We suggest performing mutation analysis at the time of diagnosis, but this decision may be influenced by available resources and by patient choice. This evaluation may be postponed until a time of disease progression when therapeutic decisions include the possibility of hematopoietic cell transplantation.

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topic (see "Patient education: Polycythemia vera (PV) (The Basics)")

SUMMARY

Description – Myeloproliferative neoplasms (MPN) are clonal hematopoietic stem cell disorders characterized by proliferation of one or more myeloid lineages, peripheral blood cytosis with effective maturation, bone marrow hypercellularity/fibrosis, and splenomegaly (table 1).

The four classic MPNs are: chronic myeloid leukemia, polycythemia vera, essential thrombocythemia, and primary myelofibrosis. Other MPNs include chronic eosinophilic leukemia; chronic neutrophilic leukemia; and MPN, unclassifiable.

Complications – MPNs are associated with variable risks for thrombohemorrhagic complications, progression to acute leukemia, and bone marrow failure from myelofibrosis and/or ineffective hematopoiesis. (See 'Complications' above.)

Genetics – Chronic myeloid leukemia is associated with the Philadelphia chromosome (Ph), an acquired reciprocal translocation between chromosomes 9 and 22.

Most other MPNs (ie, Ph-negative MPNs) are associated with acquired mutations of JAK2, CALR, or MPL. Other mutations may be present, but it is uncertain if they are drivers of the malignancy, consequences of genomic instability, and/or related to clonal hematopoiesis of indeterminate potential (CHIP). (See 'Mutations in PV, ET, and PMF' above.)

Chronic myeloid leukemia (CML) – Cells of the granulocytic lineage are the predominant proliferative component. CML is characterized by t(9;22), which creates a BCR::ABL1 rearrangement that encodes the constitutively active tyrosine kinase, BCR::ABL1.

Most cases of CML present in the relatively indolent chronic phase (CP); some patients present with advanced disease (accelerated phase [AP] or blast crisis [BC]), which are characterized by rising blast percentages with systemic symptoms and/or progressive splenomegaly. Most cases of advanced disease arise from progression of CP CML during treatment with a BCR::ABL1 tyrosine kinase inhibitor (TKI). (See "Clinical manifestations and diagnosis of chronic myeloid leukemia".)

Polycythemia vera (PV) – PV is characterized by increased red blood cell proliferation. Virtually all patients carry JAK2 V617F, a somatic gain-of-function mutation (table 3).

Most cases present in a polycythemic phase with elevated hemoglobin/hematocrit. Some patients present in a spent phase, with cytopenias (including anemia) in association with post-polycythemic myelofibrosis, ineffective hematopoiesis, and increasing splenomegaly. (See "Clinical manifestations and diagnosis of polycythemia vera".)

Primary myelofibrosis (PMF) – PMF is characterized by proliferation of abnormal megakaryocytes and granulocytes in bone marrow, a leukoerythroblastic blood picture, splenomegaly, and bone marrow fibrosis. (See "Clinical manifestations and diagnosis of primary myelofibrosis".)

A prefibrotic phase of PMF (table 4) may precede fibrotic stages (table 5), which are associated with marrow fibrosis and extramedullary hematopoiesis. (See "Clinical manifestations and diagnosis of primary myelofibrosis".)

Essential thrombocythemia (ET) – ET is manifest as sustained thrombocytosis with megakaryocytic proliferation.

ET is a diagnosis of exclusion, with clonal autonomous thrombocytosis not classifiable as PV, PMF, CML, or myelodysplastic syndrome (MDS) (table 6). (See "Clinical manifestations, pathogenesis, and diagnosis of essential thrombocythemia".)

ACKNOWLEDGMENT — The editors of UpToDate acknowledge the contributions of Stanley L Schrier, MD as Section Editor on this topic, his tenure as the founding Editor-in-Chief for UpToDate in Hematology, and his dedicated and longstanding involvement with the UpToDate program.

  1. Abkowitz JL, Catlin SN, Guttorp P. Evidence that hematopoiesis may be a stochastic process in vivo. Nat Med 1996; 2:190.
  2. McCulloch EA. Stem cell renewal and determination during clonal expansion in normal and leukaemic haemopoiesis. Cell Prolif 1993; 26:399.
  3. Tefferi A. Chronic myeloid disorders: Classification and treatment overview. Semin Hematol 2001; 38:1.
  4. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 2016; 127:2391.
  5. Kouides PA, Bennett JM. Morphology and classification of the myelodysplastic syndromes and their pathologic variants. Semin Hematol 1996; 33:95.
  6. Raza A, Alvi S, Broady-Robinson L, et al. Cell cycle kinetic studies in 68 patients with myelodysplastic syndromes following intravenous iodo- and/or bromodeoxyuridine. Exp Hematol 1997; 25:530.
  7. Dickstein JI, Vardiman JW. Hematopathologic findings in the myeloproliferative disorders. Semin Oncol 1995; 22:355.
  8. Tefferi A, Thibodeau SN, Solberg LA Jr. Clonal studies in the myelodysplastic syndrome using X-linked restriction fragment length polymorphisms. Blood 1990; 75:1770.
  9. Raskind WH, Fialkow PJ. The use of cell markers in the study of human hematopoietic neoplasia. Adv Cancer Res 1987; 49:127.
  10. Tsukamoto N, Morita K, Maehara T, et al. Clonality in chronic myeloproliferative disorders defined by X-chromosome linked probes: demonstration of heterogeneity in lineage involvement. Br J Haematol 1994; 86:253.
  11. Champion KM, Gilbert JG, Asimakopoulos FA, et al. Clonal haemopoiesis in normal elderly women: implications for the myeloproliferative disorders and myelodysplastic syndromes. Br J Haematol 1997; 97:920.
  12. Levine RL, Belisle C, Wadleigh M, et al. X-inactivation-based clonality analysis and quantitative JAK2V617F assessment reveal a strong association between clonality and JAK2V617F in PV but not ET/MMM, and identifies a subset of JAK2V617F-negative ET and MMM patients with clonal hematopoiesis. Blood 2006; 107:4139.
  13. Rumi E, Passamonti F, Della Porta MG, et al. Familial chronic myeloproliferative disorders: clinical phenotype and evidence of disease anticipation. J Clin Oncol 2007; 25:5630.
  14. 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.
  15. Hemminki K, Sundquist J, Bermejo JL. Associated cancers in parents and offspring of polycythaemia vera and myelofibrosis patients. Br J Haematol 2009; 147:526.
  16. Rumi E, Harutyunyan AS, Pietra D, et al. CALR exon 9 mutations are somatically acquired events in familial cases of essential thrombocythemia or primary myelofibrosis. Blood 2014.
  17. Lundberg P, Nienhold R, Ambrosetti A, et al. Somatic mutations in calreticulin can be found in pedigrees with familial predisposition to myeloproliferative neoplasms. Blood 2014; 123:2744.
  18. Harutyunyan AS, Giambruno R, Krendl C, et al. Germline RBBP6 mutations in familial myeloproliferative neoplasms. Blood 2016; 127:362.
  19. Cario H, Goerttler PS, Steimle C, et al. The JAK2V617F mutation is acquired secondary to the predisposing alteration in familial polycythaemia vera. Br J Haematol 2005; 130:800.
  20. Bellanné-Chantelot C, Chaumarel I, Labopin M, et al. Genetic and clinical implications of the Val617Phe JAK2 mutation in 72 families with myeloproliferative disorders. Blood 2006; 108:346.
  21. Rumi E, Passamonti F, Pietra D, et al. JAK2 (V617F) as an acquired somatic mutation and a secondary genetic event associated with disease progression in familial myeloproliferative disorders. Cancer 2006; 107:2206.
  22. Pardanani A, Lasho T, McClure R, et al. Discordant distribution of JAK2V617F mutation in siblings with familial myeloproliferative disorders. Blood 2006; 107:4572.
  23. Kralovics R, Stockton DW, Prchal JT. Clonal hematopoiesis in familial polycythemia vera suggests the involvement of multiple mutational events in the early pathogenesis of the disease. Blood 2003; 102:3793.
  24. Bellanné-Chantelot C, Jego P, Lionne-Huyghe P, et al. The JAK2(V617F) mutation may be present several years before the occurrence of overt myeloproliferative disorders. Leukemia 2008; 22:450.
  25. Teofili L, Giona F, Martini M, et al. The revised WHO diagnostic criteria for Ph-negative myeloproliferative diseases are not appropriate for the diagnostic screening of childhood polycythemia vera and essential thrombocythemia. Blood 2007; 110:3384.
  26. NOWELL PC, HUNGERFORD DA. Chromosome studies on normal and leukemic human leukocytes. J Natl Cancer Inst 1960; 25:85.
  27. Reilly JT. Pathogenetic insight and prognostic information from standard and molecular cytogenetic studies in the BCR-ABL-negative myeloproliferative neoplasms (MPNs). Leukemia 2008; 22:1818.
  28. Vannucchi AM, Guglielmelli P, Tefferi A. Advances in understanding and management of myeloproliferative neoplasms. CA Cancer J Clin 2009; 59:171.
  29. 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.
  30. Bench AJ, White HE, Foroni L, et al. Molecular diagnosis of the myeloproliferative neoplasms: UK guidelines for the detection of JAK2 V617F and other relevant mutations. Br J Haematol 2013; 160:25.
  31. Nielsen C, Birgens HS, Nordestgaard BG, Bojesen SE. Diagnostic value of JAK2 V617F somatic mutation for myeloproliferative cancer in 49 488 individuals from the general population. Br J Haematol 2013; 160:70.
  32. Cervantes F, Colomer D, Vives-Corrons JL, et al. Chronic myeloid leukemia of thrombocythemic onset: a CML subtype with distinct hematological and molecular features? Leukemia 1996; 10:1241.
  33. Buhr T, Georgii A, Choritz H. Myelofibrosis in chronic myeloproliferative disorders. Incidence among subtypes according to the Hannover Classification. Pathol Res Pract 1993; 189:121.
  34. Singh M, Bofinger A, Taylor K, Ba Pe R. Myelodysplasia with myelofibrosis--a distinct subgroup within the myelodysplastic syndromes. Pathology 1994; 26:69.
  35. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues, revised 4th edition, Swerdlow SH, Campo E, Harris NL, et al. (Eds), International Agency for Research on Cancer (IARC), Lyon 2017.
  36. Neuwirtová R, Mociková K, Musilová J, et al. Mixed myelodysplastic and myeloproliferative syndromes. Leuk Res 1996; 20:717.
  37. Hussein K, Bock O, Theophile K, et al. Chronic myeloproliferative diseases with concurrent BCR-ABL junction and JAK2V617F mutation. Leukemia 2008; 22:1059.
  38. Cambier N, Renneville A, Cazaentre T, et al. JAK2V617F-positive polycythemia vera and Philadelphia chromosome-positive chronic myeloid leukemia: one patient with two distinct myeloproliferative disorders. Leukemia 2008; 22:1454.
  39. Gaidano G, Guerrasio A, Serra A, et al. Molecular mechanisms of tumor progression in chronic myeloproliferative disorders. Leukemia 1994; 8 Suppl 1:S27.
  40. Noor SJ, Tan W, Wilding GE, et al. Myeloid blastic transformation of myeloproliferative neoplasms--a review of 112 cases. Leuk Res 2011; 35:608.
  41. Najean Y, Rain JD. The very long-term evolution of polycythemia vera: an analysis of 318 patients initially treated by phlebotomy or 32P between 1969 and 1981. Semin Hematol 1997; 34:6.
  42. Vannucchi AM, Lasho TL, Guglielmelli P, et al. Mutations and prognosis in primary myelofibrosis. Leukemia 2013; 27:1861.
  43. Tefferi A, Lasho TL, Guglielmelli P, et al. Targeted deep sequencing in polycythemia vera and essential thrombocythemia. Blood Adv 2016; 1:21.
  44. Tefferi A, Vannucchi AM. Genetic Risk Assessment in Myeloproliferative Neoplasms. Mayo Clin Proc 2017; 92:1283.
  45. Al Assaf C, Van Obbergh F, Billiet J, et al. Analysis of phenotype and outcome in essential thrombocythemia with CALR or JAK2 mutations. Haematologica 2015; 100:893.
  46. Grinfeld J, Nangalia J, Baxter EJ, et al. Classification and Personalized Prognosis in Myeloproliferative Neoplasms. N Engl J Med 2018; 379:1416.
  47. Björkholm M, Derolf AR, Hultcrantz M, et al. Treatment-related risk factors for transformation to acute myeloid leukemia and myelodysplastic syndromes in myeloproliferative neoplasms. J Clin Oncol 2011; 29:2410.
  48. Frederiksen H, Farkas DK, Christiansen CF, et al. Chronic myeloproliferative neoplasms and subsequent cancer risk: a Danish population-based cohort study. Blood 2011; 118:6515.
  49. Vannucchi AM, Masala G, Antonioli E, et al. Increased risk of lymphoid neoplasms in patients with Philadelphia chromosome-negative myeloproliferative neoplasms. Cancer Epidemiol Biomarkers Prev 2009; 18:2068.
  50. Chintapatla R, Battini R, Wiernik PH. Chronic lymphocytic leukemia with essential thrombocythemia: asbestos exposure association? Clin Adv Hematol Oncol 2012; 10:752.
  51. Malhotra J, Kremyanskaya M, Schorr E, et al. Coexistence of myeloproliferative neoplasm and plasma-cell dyscrasia. Clin Lymphoma Myeloma Leuk 2014; 14:31.
  52. Kennedy JA, Atenafu EG, Messner HA, et al. Treatment outcomes following leukemic transformation in Philadelphia-negative myeloproliferative neoplasms. Blood 2013; 121:2725.
  53. Lussana F, Rambaldi A, Finazzi MC, et al. Allogeneic hematopoietic stem cell transplantation in patients with polycythemia vera or essential thrombocythemia transformed to myelofibrosis or acute myeloid leukemia: a report from the MPN Subcommittee of the Chronic Malignancies Working Party of the European Group for Blood and Marrow Transplantation. Haematologica 2014; 99:916.
  54. Elliott MA, Tefferi A. Thrombosis and haemorrhage in polycythaemia vera and essential thrombocythaemia. Br J Haematol 2005; 128:275.
  55. Landolfi R, Marchioli R, Patrono C. Mechanisms of bleeding and thrombosis in myeloproliferative disorders. Thromb Haemost 1997; 78:617.
  56. Falanga A, Marchetti M, Evangelista V, et al. Polymorphonuclear leukocyte activation and hemostasis in patients with essential thrombocythemia and polycythemia vera. Blood 2000; 96:4261.
  57. Teofili L, Martini M, Iachininoto MG, et al. Endothelial progenitor cells are clonal and exhibit the JAK2(V617F) mutation in a subset of thrombotic patients with Ph-negative myeloproliferative neoplasms. Blood 2011; 117:2700.
  58. Barbui T, Finazzi G, Falanga A. Myeloproliferative neoplasms and thrombosis. Blood 2013; 122:2176.
  59. Casini A, Fontana P, Lecompte TP. Thrombotic complications of myeloproliferative neoplasms: risk assessment and risk-guided management. J Thromb Haemost 2013; 11:1215.
  60. Hobbs CM, Manning H, Bennett C, et al. JAK2V617F leads to intrinsic changes in platelet formation and reactivity in a knock-in mouse model of essential thrombocythemia. Blood 2013; 122:3787.
  61. van Genderen PJ, Lucas IS, van Strik R, et al. Erythromelalgia in essential thrombocythemia is characterized by platelet activation and endothelial cell damage but not by thrombin generation. Thromb Haemost 1996; 76:333.
  62. Hultcrantz M, Björkholm M, Dickman PW, et al. Risk for Arterial and Venous Thrombosis in Patients With Myeloproliferative Neoplasms: A Population-Based Cohort Study. Ann Intern Med 2018; 168:317.
  63. Sekhar M, McVinnie K, Burroughs AK. Splanchnic vein thrombosis in myeloproliferative neoplasms. Br J Haematol 2013; 162:730.
  64. Acharya J, Westwood NB, Sawyer BM, et al. Identification of latent myeloproliferative disease in patients with Budd-Chiari syndrome using X-chromosome inactivation patterns and in vitro erythroid colony formation. Eur J Haematol 1995; 55:315.
  65. De Stefano V, Teofili L, Leone G, Michiels JJ. Spontaneous erythroid colony formation as the clue to an underlying myeloproliferative disorder in patients with Budd-Chiari syndrome or portal vein thrombosis. Semin Thromb Hemost 1997; 23:411.
  66. Chait Y, Condat B, Cazals-Hatem D, et al. Relevance of the criteria commonly used to diagnose myeloproliferative disorder in patients with splanchnic vein thrombosis. Br J Haematol 2005; 129:553.
  67. Patel RK, Lea NC, Heneghan MA, et al. Prevalence of the activating JAK2 tyrosine kinase mutation V617F in the Budd-Chiari syndrome. Gastroenterology 2006; 130:2031.
  68. Boissinot M, Lippert E, Girodon F, et al. Latent myeloproliferative disorder revealed by the JAK2-V617F mutation and endogenous megakaryocytic colonies in patients with splanchnic vein thrombosis. Blood 2006; 108:3223.
  69. Primignani M, Barosi G, Bergamaschi G, et al. Role of the JAK2 mutation in the diagnosis of chronic myeloproliferative disorders in splanchnic vein thrombosis. Hepatology 2006; 44:1528.
  70. Patrono C, Rocca B, De Stefano V. Platelet activation and inhibition in polycythemia vera and essential thrombocythemia. Blood 2013; 121:1701.
  71. Rozman C, Giralt M, Feliu E, et al. Life expectancy of patients with chronic nonleukemic myeloproliferative disorders. Cancer 1991; 67:2658.
  72. Farmer S, Horváth-Puhó E, Vestergaard H, et al. Chronic myeloproliferative neoplasms and risk of osteoporotic fractures; a nationwide population-based cohort study. Br J Haematol 2013; 163:603.
  73. Mesa RA, Niblack J, Wadleigh M, et al. The burden of fatigue and quality of life in myeloproliferative disorders (MPDs): an international Internet-based survey of 1179 MPD patients. Cancer 2007; 109:68.
  74. Scherber R, Dueck AC, Johansson P, et al. The Myeloproliferative Neoplasm Symptom Assessment Form (MPN-SAF): international prospective validation and reliability trial in 402 patients. Blood 2011; 118:401.
  75. Emanuel RM, Dueck AC, Geyer HL, et al. Myeloproliferative neoplasm (MPN) symptom assessment form total symptom score: prospective international assessment of an abbreviated symptom burden scoring system among patients with MPNs. J Clin Oncol 2012; 30:4098.
  76. Abelsson J, Andréasson B, Samuelsson J, et al. Patients with polycythemia vera have worst impairment of quality of life among patients with newly diagnosed myeloproliferative neoplasms. Leuk Lymphoma 2013; 54:2226.
  77. Geyer HL, Emanuel RM, Dueck AC, et al. Distinct clustering of symptomatic burden amongst myeloproliferative neoplasm patients: retrospective assessment in 1470 patients. Blood 2014.
  78. Siitonen T, Zheng A, Savolainen ER, Koistinen P. Spontaneous granulocyte-macrophage colony growth by peripheral blood mononuclear cells in myeloproliferative disorders. Leuk Res 1996; 20:187.
  79. Gregg XT, Prchal JT. Erythropoietin receptor mutations and human disease. Semin Hematol 1997; 34:70.
  80. Silva M, Richard C, Benito A, et al. Expression of Bcl-x in erythroid precursors from patients with polycythemia vera. N Engl J Med 1998; 338:564.
  81. Kiladjian JJ, Elkassar N, Hetet G, et al. Study of the thrombopoitin receptor in essential thrombocythemia. Leukemia 1997; 11:1821.
  82. Pitcher L, Taylor K, Nichol J, et al. Thrombopoietin measurement in thrombocytosis: dysregulation and lack of feedback inhibition in essential thrombocythaemia. Br J Haematol 1997; 99:929.
  83. Horikawa Y, Matsumura I, Hashimoto K, et al. Markedly reduced expression of platelet c-mpl receptor in essential thrombocythemia. Blood 1997; 90:4031.
  84. Moliterno AR, Hankins WD, Spivak JL. Impaired expression of the thrombopoietin receptor by platelets from patients with polycythemia vera. N Engl J Med 1998; 338:572.
  85. Villeval JL, Cohen-Solal K, Tulliez M, et al. High thrombopoietin production by hematopoietic cells induces a fatal myeloproliferative syndrome in mice. Blood 1997; 90:4369.
  86. Martyré MC. TGF-beta and megakaryocytes in the pathogenesis of myelofibrosis in myeloproliferative disorders. Leuk Lymphoma 1995; 20:39.
  87. Rampal R, Al-Shahrour F, Abdel-Wahab O, et al. Integrated genomic analysis illustrates the central role of JAK-STAT pathway activation in myeloproliferative neoplasm pathogenesis. Blood 2014; 123:e123.
  88. Zhan H, Cardozo C, Raza A. MicroRNAs in myeloproliferative neoplasms. Br J Haematol 2013; 161:471.
  89. Alizadeh S, Azizi SG, Soleimani M, et al. The Role of MicroRNAs in Myeloproliferative Neoplasia. Int J Hematol Oncol Stem Cell Res 2016; 10:172.
  90. Klampfl T, Gisslinger H, Harutyunyan AS, et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med 2013; 369:2379.
  91. Nangalia J, Massie CE, Baxter EJ, et al. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med 2013; 369:2391.
  92. Rumi E, Pietra D, Ferretti V, et al. JAK2 or CALR mutation status defines subtypes of essential thrombocythemia with substantially different clinical course and outcomes. Blood 2013.
  93. Rotunno G, Mannarelli C, Guglielmelli P, et al. Impact of calreticulin mutations on clinical and hematological phenotype and outcome in essential thrombocythemia. Blood 2013.
  94. Tefferi A, Lasho TL, Finke CM, et al. CALR vs JAK2 vs MPL-mutated or triple-negative myelofibrosis: clinical, cytogenetic and molecular comparisons. Leukemia 2014; 28:1472.
  95. Cazzola M, Kralovics R. From Janus kinase 2 to calreticulin: the clinically relevant genomic landscape of myeloproliferative neoplasms. Blood 2014; 123:3714.
  96. Milosevic Feenstra JD, Nivarthi H, Gisslinger H, et al. Whole-exome sequencing identifies novel MPL and JAK2 mutations in triple-negative myeloproliferative neoplasms. Blood 2016; 127:325.
  97. Cabagnols X, Favale F, Pasquier F, et al. Presence of atypical thrombopoietin receptor (MPL) mutations in triple-negative essential thrombocythemia patients. Blood 2016; 127:333.
  98. Dai CH, Krantz SB, Dessypris EN, et al. Polycythemia vera. II. Hypersensitivity of bone marrow erythroid, granulocyte-macrophage, and megakaryocyte progenitor cells to interleukin-3 and granulocyte-macrophage colony-stimulating factor. Blood 1992; 80:891.
  99. Kobayashi S, Teramura M, Hoshino S, et al. Circulating megakaryocyte progenitors in myeloproliferative disorders are hypersensitive to interleukin-3. Br J Haematol 1993; 83:539.
  100. Axelrad AA, Eskinazi D, Correa PN, Amato D. Hypersensitivity of circulating progenitor cells to megakaryocyte growth and development factor (PEG-rHu MGDF) in essential thrombocythemia. Blood 2000; 96:3310.
  101. Fleischman AG, Aichberger KJ, Luty SB, et al. TNFα facilitates clonal expansion of JAK2V617F positive cells in myeloproliferative neoplasms. Blood 2011; 118:6392.
  102. 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.
  103. Baxter EJ, Scott LM, Campbell PJ, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 2005; 365:1054.
  104. Kaushansky K. On the molecular origins of the chronic myeloproliferative disorders: it all makes sense. Blood 2005; 105:4187.
  105. 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.
  106. Levine RL, Pardanani A, Tefferi A, Gilliland DG. Role of JAK2 in the pathogenesis and therapy of myeloproliferative disorders. Nat Rev Cancer 2007; 7:673.
  107. Jones AV, Campbell PJ, Beer PA, et al. The JAK2 46/1 haplotype predisposes to MPL-mutated myeloproliferative neoplasms. Blood 2010; 115:4517.
  108. Smalberg JH, Koehler E, Darwish Murad S, et al. The JAK2 46/1 haplotype in Budd-Chiari syndrome and portal vein thrombosis. Blood 2011; 117:3968.
  109. Pietra D, Casetti I, Da Vià MC, et al. JAK2 GGCC haplotype in MPL mutated myeloproliferative neoplasms. Am J Hematol 2012; 87:746.
  110. Ishii T, Bruno E, Hoffman R, Xu M. Involvement of various hematopoietic-cell lineages by the JAK2V617F mutation in polycythemia vera. Blood 2006; 108:3128.
  111. Delhommeau F, Dupont S, Tonetti C, et al. Evidence that the JAK2 G1849T (V617F) mutation occurs in a lymphomyeloid progenitor in polycythemia vera and idiopathic myelofibrosis. Blood 2007; 109:71.
  112. Larsen TS, Christensen JH, Hasselbalch HC, Pallisgaard N. The JAK2 V617F mutation involves B- and T-lymphocyte lineages in a subgroup of patients with Philadelphia-chromosome negative chronic myeloproliferative disorders. Br J Haematol 2007; 136:745.
  113. Pardanani A, Lasho TL, Finke C, et al. Extending Jak2V617F and MplW515 mutation analysis to single hematopoietic colonies and B and T lymphocytes. Stem Cells 2007; 25:2358.
  114. Pardanani A, Lasho TL, Finke C, et al. Demonstration of MPLW515K, but not JAK2V617F, in in vitro expanded CD4+ T lymphocytes. Leukemia 2007; 21:2206.
  115. Rosti V, Villani L, Riboni R, et al. Spleen endothelial cells from patients with myelofibrosis harbor the JAK2V617F mutation. Blood 2013; 121:360.
  116. Toyama K, Karasawa M, Yamane A, et al. JAK2-V617F mutation analysis of granulocytes and platelets from patients with chronic myeloproliferative disorders: advantage of studying platelets. Br J Haematol 2007; 139:64.
  117. Campbell PJ, Green AR. The myeloproliferative disorders. N Engl J Med 2006; 355:2452.
  118. Grimwade LF, Happerfield L, Tristram C, et al. Phospho-STAT5 and phospho-Akt expression in chronic myeloproliferative neoplasms. Br J Haematol 2009; 147:495.
  119. Kralovics R, Passamonti F, Buser AS, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med 2005; 352:1779.
  120. Jones AV, Kreil S, Zoi K, et al. Widespread occurrence of the JAK2 V617F mutation in chronic myeloproliferative disorders. Blood 2005; 106:2162.
  121. Lippert E, Boissinot M, Kralovics R, et al. The JAK2-V617F mutation is frequently present at diagnosis in patients with essential thrombocythemia and polycythemia vera. Blood 2006; 108:1865.
  122. Goerttler PS, Steimle C, März E, et al. The Jak2V617F mutation, PRV-1 overexpression, and EEC formation define a similar cohort of MPD patients. Blood 2005; 106:2862.
  123. Tefferi A, Gilliland DG. The JAK2V617F tyrosine kinase mutation in myeloproliferative disorders: status report and immediate implications for disease classification and diagnosis. Mayo Clin Proc 2005; 80:947.
  124. Teofili L, Martini M, Cenci T, et al. Different STAT-3 and STAT-5 phosphorylation discriminates among Ph-negative chronic myeloproliferative diseases and is independent of the V617F JAK-2 mutation. Blood 2007; 110:354.
  125. Yan D, Hutchison RE, Mohi G. Critical requirement for Stat5 in a mouse model of polycythemia vera. Blood 2012; 119:3539.
  126. Walz C, Ahmed W, Lazarides K, et al. Essential role for Stat5a/b in myeloproliferative neoplasms induced by BCR-ABL1 and JAK2(V617F) in mice. Blood 2012; 119:3550.
  127. Steensma DP, Dewald GW, Lasho TL, 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.
  128. Jelinek J, Oki Y, Gharibyan V, et al. JAK2 mutation 1849G>T is rare in acute leukemias but can be found in CMML, Philadelphia chromosome-negative CML, and megakaryocytic leukemia. Blood 2005; 106:3370.
  129. Levine RL, Loriaux M, Huntly BJ, et al. The JAK2V617F activating mutation occurs in chronic myelomonocytic leukemia and acute myeloid leukemia, but not in acute lymphoblastic leukemia or chronic lymphocytic leukemia. Blood 2005; 106:3377.
  130. Fröhling S, Lipka DB, Kayser S, et al. Rare occurrence of the JAK2 V617F mutation in AML subtypes M5, M6, and M7. Blood 2006; 107:1242.
  131. Wang SA, Hasserjian RP, Fox PS, et al. Atypical chronic myeloid leukemia is clinically distinct from unclassifiable myelodysplastic/myeloproliferative neoplasms. Blood 2014; 123:2645.
  132. Xing S, Wanting TH, Zhao W, et al. Transgenic expression of JAK2V617F causes myeloproliferative disorders in mice. Blood 2008; 111:5109.
  133. Pardanani A. JAK2 inhibitor therapy in myeloproliferative disorders: rationale, preclinical studies and ongoing clinical trials. Leukemia 2008; 22:23.
  134. Liu PC, Caulder E, Li J, et al. Combined inhibition of Janus kinase 1/2 for the treatment of JAK2V617F-driven neoplasms: selective effects on mutant cells and improvements in measures of disease severity. Clin Cancer Res 2009; 15:6891.
  135. Chen AT, Prchal JT. JAK2 kinase inhibitors and myeloproliferative disorders. Curr Opin Hematol 2010; 17:110.
  136. Apostolidou E, Kantarjian HM, Verstovsek S. JAK2 inhibitors: A reality? A hope? Clin Lymphoma Myeloma 2009; 9 Suppl 3:S340.
  137. Wadleigh M, Tefferi A. Preclinical and clinical activity of ATP mimetic JAK2 inhibitors. Clin Adv Hematol Oncol 2010; 8:557.
  138. Agrawal M, Garg RJ, Cortes J, et al. Experimental therapeutics for patients with myeloproliferative neoplasias. Cancer 2011; 117:662.
  139. Stein BL, Crispino JD, Moliterno AR. Janus kinase inhibitors: an update on the progress and promise of targeted therapy in the myeloproliferative neoplasms. Curr Opin Oncol 2011; 23:609.
  140. Levine RL, Gilliland DG. Myeloproliferative disorders. Blood 2008; 112:2190.
  141. 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.
  142. Godfrey AL, Chen E, Pagano F, et al. Clonal analyses reveal associations of JAK2V617F homozygosity with hematologic features, age and gender in polycythemia vera and essential thrombocythemia. Haematologica 2013; 98:718.
  143. Tefferi A, Sirhan S, Lasho TL, et al. Concomitant neutrophil JAK2 mutation screening and PRV-1 expression analysis in myeloproliferative disorders and secondary polycythaemia. Br J Haematol 2005; 131:166.
  144. Spivak JL. Narrative review: Thrombocytosis, polycythemia vera, and JAK2 mutations: The phenotypic mimicry of chronic myeloproliferation. Ann Intern Med 2010; 152:300.
  145. Takahashi K, Patel KP, Kantarjian H, et al. JAK2 p.V617F detection and allele burden measurement in peripheral blood and bone marrow aspirates in patients with myeloproliferative neoplasms. Blood 2013; 122:3784.
  146. Kiladjian JJ, Cassinat B, Turlure P, et al. High molecular response rate of polycythemia vera patients treated with pegylated interferon alpha-2a. Blood 2006; 108:2037.
  147. Lange T, Edelmann A, Siebolts U, et al. JAK2 p.V617F allele burden in myeloproliferative neoplasms one month after allogeneic stem cell transplantation significantly predicts outcome and risk of relapse. Haematologica 2013; 98:722.
  148. Austin SK, Lambert JR. The JAK2 V617F mutation and thrombosis. Br J Haematol 2008; 143:307.
  149. Alvarez-Larrán A, Bellosillo B, Pereira A, et al. JAK2V617F monitoring in polycythemia vera and essential thrombocythemia: clinical usefulness for predicting myelofibrotic transformation and thrombotic events. Am J Hematol 2014; 89:517.
  150. Besses C, Alvarez-Larrán A, Martínez-Avilés L, et al. Modulation of JAK2 V617F allele burden dynamics by hydroxycarbamide in polycythaemia vera and essential thrombocythaemia patients. Br J Haematol 2011; 152:413.
  151. Eggleton P, Michalak M. Calreticulin for better or for worse, in sickness and in health, until death do us part. Cell Calcium 2013; 54:126.
  152. Broséus J, Park JH, Carillo S, et al. Presence of calreticulin mutations in JAK2-negative polycythemia vera. Blood 2014; 124:3964.
  153. Michalak M, Groenendyk J, Szabo E, et al. Calreticulin, a multi-process calcium-buffering chaperone of the endoplasmic reticulum. Biochem J 2009; 417:651.
  154. Wang WA, Groenendyk J, Michalak M. Calreticulin signaling in health and disease. Int J Biochem Cell Biol 2012; 44:842.
  155. Levine RL. Another piece of the myeloproliferative neoplasms puzzle. N Engl J Med 2013; 369:2451.
  156. Marty C, Pecquet C, Nivarthi H, et al. Calreticulin mutants in mice induce an MPL-dependent thrombocytosis with frequent progression to myelofibrosis. Blood 2016; 127:1317.
  157. Chachoua I, Pecquet C, El-Khoury M, et al. Thrombopoietin receptor activation by myeloproliferative neoplasm associated calreticulin mutants. Blood 2016; 127:1325.
  158. Araki M, Yang Y, Masubuchi N, et al. Activation of the thrombopoietin receptor by mutant calreticulin in CALR-mutant myeloproliferative neoplasms. Blood 2016; 127:1307.
  159. Tefferi A, Wassie EA, Guglielmelli P, et al. Type 1 versus Type 2 calreticulin mutations in essential thrombocythemia: a collaborative study of 1027 patients. Am J Hematol 2014; 89:E121.
  160. Xu W, Li JY, Xia J, et al. MPL W515L mutation in Chinese patients with myeloproliferative diseases. Leuk Lymphoma 2008; 49:955.
  161. Rumi E, Pietra D, Guglielmelli P, et al. Acquired copy-neutral loss of heterozygosity of chromosome 1p as a molecular event associated with marrow fibrosis in MPL-mutated myeloproliferative neoplasms. Blood 2013; 121:4388.
  162. 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.
  163. Boyd EM, Bench AJ, Goday-Fernández A, et al. Clinical utility of routine MPL exon 10 analysis in the diagnosis of essential thrombocythaemia and primary myelofibrosis. Br J Haematol 2010; 149:250.
  164. Pardanani A, Guglielmelli P, Lasho TL, et al. Primary myelofibrosis with or without mutant MPL: comparison of survival and clinical features involving 603 patients. Leukemia 2011; 25:1834.
  165. Tefferi A. Mutations galore in myeloproliferative neoplasms: would the real Spartacus please stand up? Leukemia 2011; 25:1059.
  166. Vainchenker W, Delhommeau F, Constantinescu SN, Bernard OA. New mutations and pathogenesis of myeloproliferative neoplasms. Blood 2011; 118:1723.
  167. Tefferi A, Jimma T, Sulai NH, et al. IDH mutations in primary myelofibrosis predict leukemic transformation and shortened survival: clinical evidence for leukemogenic collaboration with JAK2V617F. Leukemia 2012; 26:475.
  168. Scott LM, Rebel VI. JAK2 and genomic instability in the myeloproliferative neoplasms: a case of the chicken or the egg? Am J Hematol 2012; 87:1028.
  169. Lundberg P, Karow A, Nienhold R, et al. Clonal evolution and clinical correlates of somatic mutations in myeloproliferative neoplasms. Blood 2014; 123:2220.
  170. Tefferi A, Lasho TL, Finke CM, et al. Targeted deep sequencing in primary myelofibrosis. Blood Adv 2016; 1:105.
  171. Tefferi A, Vainchenker W. Myeloproliferative neoplasms: molecular pathophysiology, essential clinical understanding, and treatment strategies. J Clin Oncol 2011; 29:573.
  172. Abdel-Wahab O. Genetics of the myeloproliferative neoplasms. Curr Opin Hematol 2011; 18:117.
  173. Abdel-Wahab O, Pardanani A, Rampal R, et al. DNMT3A mutational analysis in primary myelofibrosis, chronic myelomonocytic leukemia and advanced phases of myeloproliferative neoplasms. Leukemia 2011; 25:1219.
  174. Rao N, Butcher CM, Lewis ID, et al. Clonal and lineage analysis of somatic DNMT3A and JAK2 mutations in a chronic phase polycythemia vera patient. Br J Haematol 2012; 156:268.
  175. Ricci C, Spinelli O, Salmoiraghi S, et al. ASXL1 mutations in primary and secondary myelofibrosis. Br J Haematol 2012; 156:404.
  176. Verger E, Andreoli A, Chomienne C, et al. TET2 gene sequencing may be helpful for myeloproliferative neoplasm diagnosis. Br J Haematol 2014; 165:416.
  177. Hernández-Boluda JC, Pereira A, Cervantes F, et al. A polymorphism in the XPD gene predisposes to leukemic transformation and new nonmyeloid malignancies in essential thrombocythemia and polycythemia vera. Blood 2012; 119:5221.
  178. Zhang SJ, Rampal R, Manshouri T, et al. Genetic analysis of patients with leukemic transformation of myeloproliferative neoplasms shows recurrent SRSF2 mutations that are associated with adverse outcome. Blood 2012; 119:4480.
  179. Guglielmelli P, Lasho TL, Rotunno G, et al. The number of prognostically detrimental mutations and prognosis in primary myelofibrosis: an international study of 797 patients. Leukemia 2014; 28:1804.
  180. 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.
  181. Kilpivaara O, Levine RL. JAK2 and MPL mutations in myeloproliferative neoplasms: discovery and science. Leukemia 2008; 22:1813.
  182. Plo I, Chaligne R, James C, Vainchenker W. New insights into the molecular pathogenessis of Bcr-Abl-negative myeloproliferative disorders. Clin Leuk 2009; 3:33.
  183. Beer PA, Delhommeau F, LeCouédic JP, et al. Two routes to leukemic transformation after a JAK2 mutation-positive myeloproliferative neoplasm. Blood 2010; 115:2891.
  184. Thoennissen NH, Krug UO, Lee DH, et al. Prevalence and prognostic impact of allelic imbalances associated with leukemic transformation of Philadelphia chromosome-negative myeloproliferative neoplasms. Blood 2010; 115:2882.
  185. Plo I, Vainchenker W. Molecular and genetic bases of myeloproliferative disorders: questions and perspectives. Clin Lymphoma Myeloma 2009; 9 Suppl 3:S329.
  186. Kralovics R. Genetic complexity of myeloproliferative neoplasms. Leukemia 2008; 22:1841.
  187. Cross NC, Daley GQ, Green AR, et al. BCR-ABL1-positive CML and BCR-ABL1-negative chronic myeloproliferative disorders: some common and contrasting features. Leukemia 2008; 22:1975.
  188. Beer PA, Jones AV, Bench AJ, et al. Clonal diversity in the myeloproliferative neoplasms: independent origins of genetically distinct clones. Br J Haematol 2009; 144:904.
  189. Delhommeau F, Dupont S, Della Valle V, et al. Mutation in TET2 in myeloid cancers. N Engl J Med 2009; 360:2289.
  190. Tefferi A, Pardanani A, Lim KH, et al. TET2 mutations and their clinical correlates in polycythemia vera, essential thrombocythemia and myelofibrosis. Leukemia 2009; 23:905.
  191. Saint-Martin C, Leroy G, Delhommeau F, et al. Analysis of the ten-eleven translocation 2 (TET2) gene in familial myeloproliferative neoplasms. Blood 2009; 114:1628.
  192. Schaub FX, Looser R, Li S, et al. Clonal analysis of TET2 and JAK2 mutations suggests that TET2 can be a late event in the progression of myeloproliferative neoplasms. Blood 2010; 115:2003.
  193. Ortmann CA, Kent DG, Nangalia J, et al. Effect of mutation order on myeloproliferative neoplasms. N Engl J Med 2015; 372:601.
  194. Klampfl T, Harutyunyan A, Berg T, et al. Genome integrity of myeloproliferative neoplasms in chronic phase and during disease progression. Blood 2011; 118:167.
  195. Lasho TL, Pardanani A, McClure RF, et al. Concurrent MPL515 and JAK2V617F mutations in myelofibrosis: chronology of clonal emergence and changes in mutant allele burden over time. Br J Haematol 2006; 135:683.
  196. Moliterno AR, Williams DM, Rogers O, Spivak JL. Molecular mimicry in the chronic myeloproliferative disorders: reciprocity between quantitative JAK2 V617F and Mpl expression. Blood 2006; 108:3913.
  197. Guglielmelli P, Pancrazzi A, Bergamaschi G, et al. Anaemia characterises patients with myelofibrosis harbouring Mpl mutation. Br J Haematol 2007; 137:244.
  198. Vannucchi AM, Antonioli E, Guglielmelli P, et al. Clinical profile of homozygous JAK2 617V>F mutation in patients with polycythemia vera or essential thrombocythemia. Blood 2007; 110:840.
  199. Dupont S, Massé A, James C, et al. The JAK2 617V>F mutation triggers erythropoietin hypersensitivity and terminal erythroid amplification in primary cells from patients with polycythemia vera. Blood 2007; 110:1013.
  200. Williams DM, Kim AH, Rogers O, et al. Phenotypic variations and new mutations in JAK2 V617F-negative polycythemia vera, erythrocytosis, and idiopathic myelofibrosis. Exp Hematol 2007; 35:1641.
  201. Spivak JL, Moliterno AR. The roles of genetics and epigenetics in the diagnosis of chronic myeloproliferative disorders. Clinical Leukemia 2007; 1:275.
  202. Pardanani A, Fridley BL, Lasho TL, et al. Host genetic variation contributes to phenotypic diversity in myeloproliferative disorders. Blood 2008; 111:2785.
  203. Tiedt R, Hao-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.
  204. Kota J, Caceres N, Constantinescu SN. Aberrant signal transduction pathways in myeloproliferative neoplasms. Leukemia 2008; 22:1828.
  205. Hussein K, Bock O, Theophile K, et al. Biclonal expansion and heterogeneous lineage involvement in a case of chronic myeloproliferative disease with concurrent MPLW515L/JAK2V617F mutation. Blood 2009; 113:1391.
  206. Jones AV, Chase A, Silver RT, et al. JAK2 haplotype is a major risk factor for the development of myeloproliferative neoplasms. Nat Genet 2009; 41:446.
  207. Olcaydu D, Harutyunyan A, Jäger R, et al. A common JAK2 haplotype confers susceptibility to myeloproliferative neoplasms. Nat Genet 2009; 41:450.
  208. 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. Nat Genet 2009; 41:455.
  209. Hsiao HH, Liu YC, Tsai HJ, et al. JAK2V617F mutation is associated with special alleles in essential thrombocythemia. Leuk Lymphoma 2011; 52:478.
  210. Sirhan S, Lasho TL, Hanson CA, et al. The presence of JAK2V617F in primary myelofibrosis or its allele burden in polycythemia vera predicts chemosensitivity to hydroxyurea. Am J Hematol 2008; 83:363.
  211. Vannucchi AM, Antonioli E, Guglielmelli P, et al. Clinical correlates of JAK2V617F presence or allele burden in myeloproliferative neoplasms: a critical reappraisal. Leukemia 2008; 22:1299.
Topic 4511 Version 82.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟