Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Blood Adv ; 8(12): 3330-3343, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38386979

ABSTRACT

ABSTRACT: Cerebral venous sinus thrombosis (CVST) is an uncommon venous thromboembolic event accounting for <1% of strokes resulting in brain parenchymal injuries. JAK2V617F mutation, the most frequent driving mutation of myeloproliferative neoplasms, has been reported to be associated with worse clinical outcomes in patients with CVST. We investigated whether hematopoietic JAK2V617F expression predisposes to specific pathophysiological processes and/or worse prognosis after CVST. Using an in vivo mouse model of CVST, we analyzed clinical, biological, and imaging outcomes in mice with hematopoietic-restricted Jak2V617F expression, compared with wild-type Jak2 mice. In parallel, we studied a human cohort of JAK2V617F-positive or -negative CVST. Early after CVST, mice with hematopoietic Jak2V617F expression had increased adhesion of platelets and neutrophils in cerebral veins located in the vicinity of CVST. On day 1, Jak2V617F mice had a worse outcome characterized by significantly more frequent and severe intracranial hemorrhages (ICHs) and higher mortality rates. Peripheral neutrophil activation was enhanced, as indicated by higher circulating platelet-neutrophil aggregates, upregulated CD11b expression, and higher myeloperoxydase plasma level. Concurrently, immunohistological and brain homogenate analysis showed higher neutrophil infiltration and increased blood-brain barrier disruption. Similarly, patients with JAK2V617F-positive CVST tended to present higher thrombotic burden and had significantly higher systemic immune-inflammation index, a systemic thromboinflammatory marker, than patients who were JAK2V617F-negative. In mice with CVST, our study corroborates that Jak2V617F mutation leads to a specific pattern including increased thrombotic burden, ICH, and mortality. The exacerbated thromboinflammatory response, observed both in mice and patients positive for JAK2V617F, could contribute to hemorrhagic complications.


Subject(s)
Inflammation , Janus Kinase 2 , Mutation , Sinus Thrombosis, Intracranial , Animals , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mice , Sinus Thrombosis, Intracranial/genetics , Humans , Prognosis , Inflammation/genetics , Disease Models, Animal , Male , Female , Neutrophils/metabolism
3.
Leukemia ; 38(2): 326-339, 2024 02.
Article in English | MEDLINE | ID: mdl-38148396

ABSTRACT

Current recommended risk scores to predict thrombotic events associated with myeloproliferative neoplasms (MPN) do not discriminate between arterial and venous thrombosis despite their different physiopathology. To define novel stratification systems, we delineated a comprehensive landscape of MPN associated thrombosis across a large long-term follow-up MPN cohort. Prior arterial thrombosis, age >60 years, cardiovascular risk factors and presence of TET2 or DNMT3A mutations were independently associated with arterial thrombosis in multivariable analysis. ARTS, an ARterial Thrombosis Score, based on these four factors, defined low- (0.37% patients-year) and high-risk (1.19% patients-year) patients. ARTS performance was superior to the two-tiered conventional risk stratification in our training cohort, across all MPN subtypes, as well as in two external validation cohorts. Prior venous thrombosis and presence of a JAK2V617F mutation with a variant allelic frequency ≥50% were independently associated with venous thrombosis. The discrimination potential of VETS, a VEnous Thrombosis Score based on these two factors, was poor, similar to the two-tiered conventional risk stratification. Our study pinpoints arterial and venous thrombosis clinico-molecular differences and proposes an arterial risk score for more accurate patients' stratification. Further improvement of venous risk scores, accounting for additional factors and considering venous thrombosis as a heterogeneous entity is warranted.


Subject(s)
Myeloproliferative Disorders , Neoplasms , Thrombosis , Venous Thrombosis , Humans , Middle Aged , Neoplasms/complications , Venous Thrombosis/genetics , Thrombosis/genetics , Thrombosis/complications , Mutation , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/genetics , Risk Factors , Janus Kinase 2/genetics , Risk Assessment
5.
Blood Adv ; 6(9): 2813-2823, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35030630

ABSTRACT

The mechanisms of transformation of chronic myeloproliferative neoplasms (MPN) to leukemia are largely unknown, but TP53 mutations acquisition is considered a key event in this process. p53 is a main tumor suppressor, but mutations in this protein per se do not confer a proliferative advantage to the cells, and a selection process is needed for the expansion of mutant clones. MDM2 inhibitors may rescue normal p53 from degradation and have been evaluated in a variety of cancers with promising results. However, the impact of these drugs on TP53-mutated cells is underexplored. We report herein evidence of a direct effect of MDM2 inhibition on the selection of MPN patients' cells harboring TP53 mutations. To decipher whether these mutations can arise in a specific molecular context, we used a DNA single-cell approach to determine the clonal architecture of TP53-mutated cells. We observed that TP53 mutations are late events in MPN, mainly occurring in the driver clone, whereas clonal evolution frequently consists of sequential branching instead of linear consecutive acquisition of mutations in the same clone. At the single-cell level, the presence of additional mutations does not influence the selection of TP53 mutant cells by MDM2 inhibitor treatment. Also, we describe an in vitro test allowing to predict the emergence of TP53 mutated clones. Altogether, this is the first demonstration that a drug treatment can directly favor the emergence of TP53-mutated subclones in MPN.


Subject(s)
Antineoplastic Agents , Myeloproliferative Disorders , Clone Cells/metabolism , Humans , Mutation , Myeloproliferative Disorders/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Single-Cell Analysis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Br J Haematol ; 196(3): 676-680, 2022 02.
Article in English | MEDLINE | ID: mdl-34562020

ABSTRACT

We assessed the diagnostic performances of erythropoietin and JAK2 mutations in 1,090 patients with suspected polycythemia who were referred for red cell mass (RCM) measurement. In patients with a high haematocrit and/or haemoglobin level, a low erythropoietin level (<=3·3 mUI/ml) and JAK2 mutation showed comparable positive predictive value (PPV) for true polycythemia (RCM>=125%), 92·1% and 90% respectively. A very-low erythropoietin level (<=1·99 mUI/ml) had a PPV of 100% for polycythemia vera (PV) diagnosis. We confirmed the correlations between RCM, erythropoietin and JAK2 variant allelic frequency in PV patients. This study prompts the need to revisit the role of EPO in PV diagnostic criteria.


Subject(s)
Erythropoietin/blood , Janus Kinase 2/genetics , Mutation , Polycythemia Vera/blood , Polycythemia Vera/genetics , Alleles , Amino Acid Substitution , Clinical Decision-Making , Disease Management , Erythrocyte Indices , Erythrocyte Volume , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Plasma Volume , Polycythemia Vera/diagnosis , Polycythemia Vera/epidemiology , Sensitivity and Specificity
8.
Blood Adv ; 5(5): 1442-1451, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33666653

ABSTRACT

We aimed to study the prognostic impact of the mutational landscape in primary and secondary myelofibrosis. The study included 479 patients with myelofibrosis recruited from 24 French Intergroup of Myeloproliferative Neoplasms (FIM) centers. The molecular landscape was studied by high-throughput sequencing of 77 genes. A Bayesian network allowed the identification of genomic groups whose prognostic impact was studied in a multistate model considering transitions from the 3 conditions: myelofibrosis, acute leukemia, and death. Results were validated using an independent, previously published cohort (n = 276). Four genomic groups were identified: patients with TP53 mutation; patients with ≥1 mutation in EZH2, CBL, U2AF1, SRSF2, IDH1, IDH2, NRAS, or KRAS (high-risk group); patients with ASXL1-only mutation (ie, no associated mutation in TP53 or high-risk genes); and other patients. A multistate model found that both TP53 and high-risk groups were associated with leukemic transformation (hazard ratios [HRs] [95% confidence interval], 8.68 [3.32-22.73] and 3.24 [1.58-6.64], respectively) and death from myelofibrosis (HRs, 3.03 [1.66-5.56] and 1.77 [1.18-2.67], respectively). ASXL1-only mutations had no prognostic value that was confirmed in the validation cohort. However, ASXL1 mutations conferred a worse prognosis when associated with a mutation in TP53 or high-risk genes. This study provides a new definition of adverse mutations in myelofibrosis with the addition of TP53, CBL, NRAS, KRAS, and U2AF1 to previously described genes. Furthermore, our results argue that ASXL1 mutations alone cannot be considered detrimental.


Subject(s)
Primary Myelofibrosis , Bayes Theorem , Genomics , Humans , Mutation , Primary Myelofibrosis/diagnosis , Primary Myelofibrosis/genetics , Prognosis , Repressor Proteins/genetics
9.
J Exp Med ; 218(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33075130

ABSTRACT

Interferon α (IFNα) is used to treat JAK2V617F-driven myeloproliferative neoplasms (MPNs) but rarely clears the disease. We investigated the IFNα mechanism of action focusing on PML, an interferon target and key senescence gene whose targeting by arsenic trioxide (ATO) drives eradication of acute promyelocytic leukemia. ATO sharply potentiated IFNα-induced growth suppression of JAK2V617F patient or mouse hematopoietic progenitors, which required PML and was associated with features of senescence. In a mouse MPN model, combining ATO with IFNα enhanced and accelerated responses, eradicating MPN in most mice by targeting disease-initiating cells. These results predict potent clinical efficacy of the IFNα+ATO combination in patients and identify PML as a major effector of therapy, even in malignancies with an intact PML gene.


Subject(s)
Arsenic Trioxide/pharmacology , Interferon-alpha/pharmacology , Janus Kinase 2/metabolism , Myeloproliferative Disorders/drug therapy , Promyelocytic Leukemia Protein/metabolism , Animals , Cell Line , Cell Line, Tumor , Disease Models, Animal , Humans , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloproliferative Disorders/metabolism
11.
Blood Adv ; 4(15): 3708-3715, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32777065

ABSTRACT

Myeloproliferative neoplasms (MPNs) are the most frequent underlying causes of splanchnic vein thromboses (SVTs). MPN patients with SVTs (MPN-SVT) often have a unique presentation including younger age, female predominance, and low Janus kinase 2 (JAK2) mutation allele burden. This study aimed at identifying risk factors for adverse hematologic outcomes in MPN-SVT patients. We performed a retrospective study of a fully characterized cohort of MPN-SVT patients. The primary outcome was the incidence of evolution to myelofibrosis, acute leukemia, or death. Eighty patients were included in the testing cohort. Median follow-up was 11 years. Most of the patients were women with a mean age of 42 years and a diagnosis of polycythemia vera. The primary outcome was met in 13% of the patients and was associated with a JAK2V617F allele burden ≥50% (odds ratio [OR], 14.7) and presence of additional mutations in genes affecting chromatin/spliceosome (OR, 9). We identified high-risk patients (29% of the cohort) as those harboring at least 1 molecular risk factor: JAK2-mutant allele burden ≥50%, presence of chromatin/spliceosome/TP53 mutation. High-risk patients had worse event-free survival (81% vs 100%; P = .001) and overall survival at 10 years (89% vs 100%; P = .01) than low-risk patients. These results were confirmed in an independent validation cohort of 30 MPN-SVT patients. In conclusion, molecular profiling identified MPN-SVT patients with dismal outcome. In this high-risk population, a disease-modifying therapy should be taken into consideration to minimize the probability of transformation.


Subject(s)
Myeloproliferative Disorders , Polycythemia Vera , Primary Myelofibrosis , Venous Thrombosis , Adult , Female , Humans , Male , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/genetics , Retrospective Studies
12.
Int J Lab Hematol ; 42(6): 827-832, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32614523

ABSTRACT

INTRODUCTION: The pathogenesis of myeloproliferative neoplasms (MPNs) is closely related to the acquisition of specific molecular alterations in JAK2, MPL, or CALR genes, the presence of which represent major diagnostic criteria in the WHO classification. The CALR exon 9 insertions and deletions are very heterogeneous, and their detection mainly relies on polymerase chain reaction (PCR) fragment length analysis. METHODS: We report on the rare nonclassical profiles that we observed among the 1382 patients analyzed by PCR fragment length analysis. In difficult cases, we tested germline DNA and performed NGS analysis. RESULTS: We faced some troubling results because of the presence of several unexpected peaks. Our investigations showed that they resulted from a mix of isolated or double somatic mutations combined with germline alterations which could be misleading for a correct diagnosis. The precise interpretation of such difficult cases is mandatory as a misinterpretation could lead to the prescription of cytoreductive drugs to nondiseased persons or to an absence of treatment in true MPN patients. CONCLUSION: Our observations showed that every mutation should be verified by direct Sanger sequencing, and we show that sometimes it may be necessary to study germline DNA and to complement with NGS analysis to precisely interpret the molecular alterations.


Subject(s)
Calreticulin/genetics , Exons , Hematologic Neoplasms/genetics , Mutation , Myeloproliferative Disorders/genetics , Neoplasm Proteins/genetics , DNA Mutational Analysis , Female , Humans , Male , Middle Aged , Polymerase Chain Reaction
13.
Haematologica ; 105(6): 1539-1551, 2020 06.
Article in English | MEDLINE | ID: mdl-31488557

ABSTRACT

Myelodysplastic syndromes and acute myeloid leukemia with TP53 mutations are characterized by frequent relapses, poor or short responses, and poor survival with the currently available therapies including chemotherapy and 5-azacitidine (AZA). PRIMA-1Met(APR-246,APR) is a methylated derivative of PRIMA-1, which induces apoptosis in human tumor cells through restoration of the transcriptional transactivation function of mutant p53. Here we show that low doses of APR on its own or in combination with AZA reactivate the p53 pathway and induce an apoptosis program. Functionally, we demonstrate that APR exerts these activities on its own and that it synergizes with AZA in TP53-mutated myelodysplastic syndromes (MDS)/acute myeloid leukemia (AML) cell lines and in TP53-mutated primary cells from MDS/AML patients. Low doses of APR on its own or in combination with AZA also show significant efficacy in vivo Lastly, using transcriptomic analysis, we found that the APR + AZA synergy was mediated by downregulation of the FLT3 pathway in drug-treated cells. Activation of the FLT3 pathway by FLT3 ligand reversed the inhibition of cell proliferation by APR + AZA. These data suggest that TP53-mutated MDS/AML may be better targeted by the addition of APR-246 to conventional treatments.


Subject(s)
Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Aza Compounds , Azacitidine/pharmacology , Bridged Bicyclo Compounds, Heterocyclic , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Quinuclidines , Tumor Suppressor Protein p53/genetics
14.
Ann Hematol ; 98(1): 111-118, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30259120

ABSTRACT

The JAK2V617F mutation is part of the major criteria for diagnosis of myeloproliferative neoplasms (MPN). Allele-specific quantitative PCR (qPCR) is the most prevalent method used in laboratories but with the advent of next-generation sequencing (NGS) techniques, we felt necessary to evaluate this approach for JAK2 mutations testing. Among DNA samples from 427 patients analyzed by qPCR and NGS, we found an excellent concordance between both methods when allelic burden was superior to 2% (the detection limit of our NGS assay). Only one sample among 298 was found negative by NGS while allelic burden by qPCR was 3%. Because NGS detection limit is higher, sensitivity was lower as exemplified by 21 samples found negative whereas qPCR measured allelic burdens between 0.1 and 1%. Importantly, quantitative data of samples found positive by both techniques were highly correlated (R2 = 0.9477). We also evaluated 40 samples tested for JAK2 exon 12 mutations by HRM. The concordance with NGS was of 100%. Using NGS, the full coding region of JAK2 was analyzed leading to identification of several variants outside of exon 12 and 14 which were previously described or not. Interestingly, we found one somatic mutation (c.1034A>T p.H345L) which induced constitutive activation of the JAK/STAT pathway leading to an increased proliferation of BaF/3 cells with low-dose EPO. This study showed that NGS is a robust method highly correlated to qPCR, although less sensitive, but providing the opportunity to identify other JAK2 variants with potential impact on disease initiation or evolution.


Subject(s)
Exons , Hematologic Neoplasms/genetics , High-Throughput Nucleotide Sequencing/methods , Janus Kinase 2/genetics , Mutation, Missense , Myeloproliferative Disorders/genetics , Amino Acid Substitution , Cell Line, Tumor , DNA Mutational Analysis/methods , Female , Humans , Janus Kinase 2/metabolism , Male , Real-Time Polymerase Chain Reaction/methods
15.
Blood Cancer J ; 8(10): 94, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30287855

ABSTRACT

Polycythemia vera is characterized by the acquisition of the JAK2V617F mutation. Recommended treatments include hydroxyurea and interferon-alpha. Several groups have reported a reduction in the JAK2 mutant allele burden in interferon-treated patients, but significance of this observation is questioned. We characterized the activity of ropeginterferon alpha-2b, a novel form of interferon-alpha recently shown to be safe and efficacious in polycythemia vera. Ropeginterferon was able to inhibit the proliferation of the HEL, UKE-1, and UT-7 JAK2-mutant cell lines while sparing JAK2-wild-type UT-7 and normal CD34+ cells growth. In vitro treatment of erythroid progenitors derived from PV patients showed that ropeginterferon could considerably inhibit the growth of endogenous erythroid colonies, a hallmark of polycythemia vera. Finally, we could study in sequential samples the clonal architecture of erythroid progenitors derived from patients included in a randomized study comparing hydroxyurea to ropeginterferon. After 1 year of treatment with ropeginterferon, the ratio of JAK2-mutated to wild-type colonies grown from bone marrow progenitors was reduced by 64%, compared to 25% in patients receiving hydroxyurea. This study shows that ropeginterferon has a potent targeted activity against JAK2-mutant cells and is able to drastically reduce the proportion of malignant progenitors in patients treated with this drug.


Subject(s)
Alleles , Amino Acid Substitution , Interferon alpha-2/pharmacology , Interferon-alpha/pharmacology , Janus Kinase 2 , Mutation , Polycythemia Vera/genetics , Polyethylene Glycols/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Gene Frequency , Genotype , Humans , Interferon alpha-2/therapeutic use , Interferon-alpha/therapeutic use , Polycythemia Vera/diagnosis , Polycythemia Vera/drug therapy , Polyethylene Glycols/therapeutic use , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use
16.
Thromb Haemost ; 118(9): 1586-1599, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30103245

ABSTRACT

Thromboembolic events are the main cause of mortality in BCR-ABL1-negative myeloproliferative neoplasms (MPNs) but their underlying mechanisms are largely unrecognized. The Janus kinase 2 (JAK2)V617F mutation is the most frequent genetic alteration leading to MPN. Usually found in haematopoietic progenitors and stem cells, this mutation has also been described in endothelial cells (ECs) of MPN patients. In this study, we have questioned the impact of the JAK2V617F mutation on EC phenotype and functions. We developed an induced pluripotent stem cells strategy to compare JAK2 mutant and wild-type ECs. Transcriptomic assays showed that several genes and pathways involved in inflammation, cell adhesion and thrombotic events were over-represented in JAK2V617F ECs and expression levels of von Willebrand factor and P-selectin (CD62P) proteins were increased. Finally, we found that leucocytes from MPN patients adhere more tightly to JAK2V617F ECs. Our results show that JAK2V617F ECs have a pro-inflammatory and pro-thrombotic phenotype and were functionally pro-adherent.


Subject(s)
Blood Platelets/physiology , Endothelial Cells/physiology , Induced Pluripotent Stem Cells/physiology , Janus Kinase 2/genetics , Myeloid Progenitor Cells/physiology , Myeloproliferative Disorders/genetics , Thrombosis/genetics , Cell Adhesion/genetics , Cell Differentiation , Cells, Cultured , Fusion Proteins, bcr-abl/metabolism , Gene Expression Profiling , Humans , Mutation/genetics , Transgenes/genetics
19.
J Mol Diagn ; 18(1): 68-74, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26596525

ABSTRACT

With the recent discovery of CALR mutations, >80% of patients with myeloproliferative neoplasms carry a phenotype-driving mutation. For JAK2 V617F, the most frequent mutation in myeloproliferative neoplasms, accurate determination of mutational loads is of interest at diagnosis, for phenotypic and prognostic purposes, and during follow-up for minimal residual disease assessment. We developed a digital PCR technique that allowed the accurate determination of CALR allelic burdens for the main mutations (types 1 and 2). Compared with the commonly used fluorescent PCR product analysis, digital PCR is more precise, reproducible, and accurate. Furthermore, this method reached a very high sensitivity. We detected at least 0.025% CALR mutants. It can thus be used for patient characterization at diagnosis and for minimal residual disease monitoring. When applied to patients with primary myelofibrosis who underwent hematopoietic stem cell transplant, the digital PCR detected low levels of minimal residual disease. After negativation of the mutational load in all patients, the disease reappeared at a low level in one patient, preceding hematologic relapse. In conclusion, digital PCR adapted to type 1 and 2 CALR mutations is an inexpensive, highly precise, and sensitive technique suitable for evaluation of myeloproliferative neoplasm patients during follow-up.


Subject(s)
Bone Marrow Transplantation , Calreticulin/genetics , Hematopoietic Stem Cell Transplantation , Neoplasm, Residual/diagnosis , Polymerase Chain Reaction/methods , Primary Myelofibrosis/diagnosis , Alleles , Exons , Humans , Mutation , Neoplasm, Residual/genetics , Primary Myelofibrosis/genetics
20.
Blood ; 126(24): 2585-91, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26486786

ABSTRACT

Myeloproliferative neoplasms are clonal disorders characterized by the presence of several gene mutations associated with particular hematologic parameters, clinical evolution, and prognosis. Few therapeutic options are available, among which interferon α (IFNα) presents interesting properties like the ability to induce hematologic responses (HRs) and molecular responses (MRs) in patients with JAK2 mutation. We report on the response to IFNα therapy in a cohort of 31 essential thrombocythemia (ET) patients with CALR mutations (mean follow-up of 11.8 years). HR was achieved in all patients. Median CALR mutant allelic burden (%CALR) significantly decreased from 41% at baseline to 26% after treatment, and 2 patients even achieved complete MR. In contrast, %CALR was not significantly modified in ET patients treated with hydroxyurea or aspirin only. Next-generation sequencing identified additional mutations in 6 patients (affecting TET2, ASXL1, IDH2, and TP53 genes). The presence of additional mutations was associated with poorer MR on CALR mutant clones, with only minor or no MRs in this subset of patients. Analysis of the evolution of the different variant allele frequencies showed that the mutated clones had a differential sensitivity to IFNα in a given patient, but no new mutation emerged during treatment. In all, this study shows that IFNα induces high rates of HRs and MRs in CALR-mutated ET, and that the presence of additional nondriver mutations may influence the MR to therapy.


Subject(s)
Calreticulin/genetics , Interferon-alpha/therapeutic use , Mutation , Polyethylene Glycols/therapeutic use , Thrombocythemia, Essential/drug therapy , Adolescent , Adult , Alleles , Aspirin/therapeutic use , Clonal Evolution/drug effects , Clone Cells/drug effects , DNA Mutational Analysis , DNA-Binding Proteins/genetics , Dioxygenases , Female , Follow-Up Studies , Genes, p53 , Humans , Hydroxyurea/therapeutic use , Interferon-alpha/adverse effects , Isocitrate Dehydrogenase/genetics , Male , Middle Aged , Off-Label Use , Polyethylene Glycols/adverse effects , Proto-Oncogene Proteins/genetics , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use , Remission Induction , Repressor Proteins/genetics , Thrombocythemia, Essential/blood , Thrombocythemia, Essential/genetics , Thrombocythemia, Essential/pathology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL