GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 2801-2801
    Abstract: Background : In primary myelofibrosis (PMF), ̴ 88% of patients harbor JAK2, CALR or MPL, with respective mutational frequencies of approximately 60%, 22% and 6%; ̴ 12% are wild type for all three driver mutations and are labelled as being "triple negative" (Blood. 2014;124:2507). It is now well-established that CALR-mutated patients with PMF display significantly better survival, compared to all the other mutational categories; in addition, preliminary data have suggested inferior survival for "triple-negative" cases (Leukemia. 2014;28:1472; Blood. 2014;124:1062) and the possibility that the favorable impact of CALR mutations might be restricted to CALR type 1 or type 1-like variants (Blood. 2014;124:2465). In the current study, we sought to clarify the prognostic impact of driver mutations, and CALR variants, on overall (OS) and leukemia-free (LFS) survival, in a two-center study involving 1118 patients. Methods: A total of 1118 patients with PMF from the Mayo Clinic (n =722) and University of Florence, Italy (n =396) were included in the current study. Study patients were selected based on availability of mutation information. PMF diagnosis was according to World Health Organization criteria (Blood. 2009;114:937). Previously published methods were used for CALR, JAK2 and MPL mutation analyses and determination of CALR variants (Blood. 2014;124:2465). Kaplan-Meier survival analysis was considered from the date of first referral for the Mayo cohort and date of diagnosis for the Florence cohort. Leukemic transformation events replaced death as the uncensored variable during LFS analysis and Cox proportional hazard regression model was used for multivariable analysis. Results : Analysis was first conducted on the Mayo cohort of 722 patients (median age 64 years; 64% males). DIPSS-plus risk distributions were 14% low, 17% intermediate-1, 37% intermediate-2 and 33% high. All patients were annotated for JAK2/CALR/MPL mutations as well as CALR variants; 477 harbored JAK2, 139 CALR and 41 MPL mutations; 65 patients were triple-negative. The 139 CALR -mutated patients included type 1/type 1-like (n =115) and type 2/type 2-like (n =24). At a median follow-up time of 3.1 years, 439 (61%) deaths and 63 (8.7%) blast transformations were documented. In univariate analysis, survival in patients with CALR type 1/type 1-like mutations was significantly longer than every other mutational category: HR (95% CI) were for triple-negative 2.7 (1.8-4.0), JAK2 2.7 (2.0-3.7), CALR type 2/type 2-like 2.3 (1.3-4.2) and MPL 1.9 (1.1-3.1) (Figure 1); these differences were sustained during multivariable analysis that included ASXL1 mutations (n =480) and DIPSS-plus, and were also apparent in the Florence cohort of 396 cases that included 251 JAK2, 53 CALR type 1/type 1-like, 21 CALR type 2/type 2-like, 50 triple-negative and 21 MPL mutated cases (Figure 2). There was no difference in survival among the Mayo cohort across mutational categories not including CALR type 1/type 1-like variants (p=0.33). Because specific driver mutations in PMF differentially cluster with age and sex, multivariable analysis including these two parameters was also performed and confirmed the significant survival advantage of CALR type 1/type 1-like over triple-negative, JAK2 and CALR type 2/type 2-like mutated cases. Based on the above information, we divided driver mutational categories into "type 1/type 1-like" (median survival 10.3 years) and "all other mutational categories" (median survival 3.9 years; p 〈 0.01). The difference in survival between these two groups remained significant (HR 2.1, 95% CI 1.5-2.9) during multivariable analysis that included both DIPSS-plus (P 〈 0.01) and ASXL1 mutation (HR 1.8, 95% CI 1.4-2.2). LFS was similar between these two groups (p=0.4) but it was inferior in triple-negative cases compared to CALR type 1/type 1-like variants (HR 2.8, 95% CI 1.3-6.2), MPL (HR 4.6; 95% CI 1.04-20.4) and JAK2 (HR 2.5, 95% CI 1.3-4.8) but not CALR type 2/type 2-like variants (p=0.56). There was no difference in LFS among mutational categories not including triple-negative cases (p=0.33). Conclusions : In PMF, driver mutations might be classified into two prognostically distinct categories: "favorable" (type 1/type 1-like) and "unfavorable" (all other mutational categories). In addition, triple-negative cases might be at a higher risk of leukemic transformation. Disclosures Pardanani: Stemline: Research Funding. Vannucchi:Novartis: Other: Research Funding paid to institution (University of Florence), Research Funding; Shire: Speakers Bureau; Baxalta: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. LBA-2-LBA-2
    Abstract: BCR-ABL negative myeloproliferative neoplasms (MPNs), such as polycythemia vera (PV), essential thrombocythemia (ET) and myelofibrosis (MF) are chronic myeloid malignancies characterized by overproduction of hematopoietic cells. JAK2 mutations are found in most patients with PV, and in only 50-60% of patients with ET and MF. JAK2 mutation testing has greatly simplified MPN diagnosis, but distinguishing JAK2-wildtype ET from reactive thrombocytosis remains a diagnostic challenge. Mutations in signalling pathways (MPL, LNK) and epigenetic regulators (TET2, DNMT3A, IDH1/2, EXH2, ASXL1) have been found in a minority of MPNs. However genome-wide data are lacking and the pathogenesis of MPNs that do not harbor JAK2 or MPLmutations remains obscure. Methods Exome sequencing was performed in 151 MPN patients on matched tumor and constitutional samples. CALR status was assessed in 3412 samples using Sanger sequencing and analysis of exome/genome sequencing data. Presence of CALR mutations in hematopoietic stem and progenitor cells was assessed by flow sorting and sequencing. Phylogenetic trees were established using hematopoietic colonies. Calreticulin cellular localisation was assessed in patient samples and cell lines expressing CALR variants by flow cytometry and immunofluorescence. Results Exome sequencing identified 1498 somatic mutations with a median of 6.5 mutations in PV and ET, and 13 in MF (MF vs ET, P=0.0002; MF vs PV, P=0.008). JAK2V617F was found in all cases of PV (n=48), 56% of ET (35/62), and 69% of MF (27/39), and MPL mutations in 7 ET and MF cases.  Mutations in epigenetic regulators TET2, DNMT3A, ASXL1, EZH2, and IDH1/2 were identified in 22, 12, 12, 4, 3 patients respectively, and components of the splicing machinery (U2AF1, SF3B1 or SRSF2) were mutated in 9 patients.  Mutations in rare genes reported to be mutated in MPNs were found in four patients (1 CBL; 2 NFE2; 1 SH2B3/LNK). We found novel somatic mutations in CHEK2 (1 PV, 1 ET and 1 MF) which have not been previously reported in MPNs.  The mutation spectrum showed a predominance of C 〉 T transitions. Pairwise associations between MPN genes demonstrated that ASXL1 and SRSF2 mutations were positively correlated with mutations in epigenetic modifiers. Novel somatic mutations in calreticulin (CALR) were identified by exome sequencing in the majority (26/31) of JAK2 or MPL unmutated patients. CALR and JAK2/MPL mutations were mutually exclusive, and 97% of patients harbored a mutation in 1 of these 3 genes. In an extended follow up screen of 1345 hematological malignancies, 1517 other cancers and 550 controls we found CALR mutations in 71% of ET (80/112), 56% of idiopathic MF (18/32), 86% of post ET-MF (12/14) and 8% of myelodysplasia (10/115), but not in other myeloid, lymphoid or solid cancers. Compared to JAK2-mutated MPNs, those with CALR mutations presented with higher platelet counts (Wilcoxon rank-sum, P=0.0003), lower hemoglobin levels (Student’s t test, P=0.02) and showed a higher incidence of transformation to MF (Fishers exact, P=0.03). All CALR mutations were insertions or deletions affecting exon 9, with 2 common variants L367fs*46 (52 bp deletion) and K385fs*47 (5 bp insertion). Loss of heterozygosity over CALR was seen in a minority of patients. Of 148 CALR mutations identified, there were 19 distinct variants. Remarkably, all generated a +1 basepair frameshift, which results in loss of most of the C-terminal acidic domain of the protein as well as the KDEL Golgi-to-endoplasmic reticulum (ER) retrieval signal, raising the possibility of compromised ER retention. Mutant proteins were readily detected in transfected cell lines and localised to the ER in the same manner as wildtype CALR, without Golgi or cell surface accumulation. These results are consistent with studies reporting KDEL-independent mechanisms of ER retention. Mutation of CALR was detected in highly purified hematopoietic stem/progenitor cells. Clonal analyses demonstrated CALR mutations in the earliest phylogenetic node in 5/5 patients, consistent with it being an initiating mutation in these individuals. Conclusions We describe the mutational landscape of BCR-ABL negative MPNs and demonstrate that somatic mutations in the endoplasmic reticulum chaperone CALR are found in the majority of patients with JAK2-unmutated MPNs. These results reveal a novel biological pathway as a target for tumorigenic mutations and will simplify diagnosis of MPN patients. Disclosures: Bowen: Celgene: Honoraria. Harrison:S Bio: Honoraria, Membership on an entity’s Board of Directors or advisory committees; Shire: Speakers Bureau; Celgene: Honoraria; YM Bioscience: Honoraria, Membership on an entity’s Board of Directors or advisory committees; Sanofi: Honoraria, Membership on an entity’s Board of Directors or advisory committees, Speakers Bureau; Novartis: Honoraria, Membership on an entity’s Board of Directors or advisory committees, Research Funding, Speakers Bureau; Gilead: Honoraria, Membership on an entity’s Board of Directors or advisory committees. Vannucchi:Novartis: Honoraria, Membership on an entity’s Board of Directors or advisory committees.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 1587-1587
    Abstract: The Dynamic International Prognostic Scoring System-plus (DIPSS-plus) uses eight adverse factors to predict survival in primary myelofibrosis (PMF): age 〉 65 years, hemoglobin 〈 10 g/dL, leukocytes 〉 25 x 109/L, circulating blasts 31%, constitutional symptoms, red cell transfusion need, platelets 〈 100 x 109/L and unfavorable karyotype (J Clin Oncol. 2011;29:392). The latter two also predicted leukemic transformation. Most recently, an international study of 879 PMF patients identified ASXL1, SRSF2, EZH2 and IDH1/2 mutations as inter-independent predictors of shortened overall or leukemia-free survival (Leukemia 2013Apr 26 doi: 101038/leu2013119. 2013). Molecular prognostication is now well established for acute myeloid leukemia with normal karyotype (NK). There is currently little information on clinical or molecular prognostication in NK-PMF. Methods Diagnosis of PMF was according to WHO criteria. The study population included all referrals to the Mayo Clinic, Rochester, MN, USA or the University of Florence, Florence, Italy. Information on clinical and laboratory parameters and karyotype was available in all study patients, at time of referral, which coincided with time of sample collection for mutation screening. The current study considered only those patients with NK-PMF. ASXL1, EZH2, SRSF2, IDH1/2 and JAK2 mutations were performed on variable number of patients depending on bone marrow or granulocyte DNA availability. Clinical parameters examined for prognostic relevance included those listed for DIPSS-plus, less unfavorable karyotype. Results A total of 690 patients with NK-PMF patients were studied. Median age was 65 years (14-89). The percentage of patients with age 〉 65 years was 48%, red cell transfusion dependent 35%, hemoglobin 〈 10 g/dL 49%, platelets 〈 100 x 10(9)/L 19%, leukocyte count 〉 25 x 10(9)/L 15%, circulating blasts ≥1% 49% and constitutional symptoms 33%. At a median follow-up of 29 months, 351 (51%) deaths and 39 (6%) leukemic transformations were recorded. Mutational frequencies and clinical correlates The respective frequencies (mutated/number of patients studied) of JAK2V617F, ASXL1, EZH2, SRSF2 and IDH1/2 mutations were 60% (284/473), 34% (72/214), 8% (14/179), 15% (37/249) and 5% (14/262). There was no significant association between JAK2V617F and any one of the aforementioned mutations. Inter-mutation association was evident only between SRSF2 and IDH1/2 (p=0.0005). Each one of the DIPSS-plus risk parameters was examined for correlation with a specific mutation: ASXL1 was associated with leukocyte count 〉 25 x 10(9)/L (p 〈 0.0001) and circulating blasts ≥1% (p=0.0005); EZH2 with leukocyte count 〉 25 x 10(9)/L (p=0.008); and SRSF2 with age 〉 65 years (p=0.0007), transfusion need (p=0.04) and hemoglobin 〈 10 g/dL (p=0.03). JAK2V617F was associated with age 〉 65 years (p=0.003) and leukocyte count 〉 25 x 10(9)/L (p=0.02). Predictors of overall and leukemia-free survival In univariate analysis, all 7 DIPSS-plus parameters, as well as ASXL1, EZH2, SRSF2 and IDH1/2 mutations showed significant association with shortened survival: When each of these analyses was adjusted for age, all except constitutional symptoms and SRSF2 mutations retained their significance. Multivariable analysis of the six age-independent DIPSS-plus variables identified all but transfusion-need as independent predictors of inferior survival. A similar analysis restricted to mutations identified ASXL1 and EZH2 as being independently adverse. When both mutations and DIPSS-plus variables were included in the multivariable model, only four parameters remained significant: age 〉 65 years (HR 4.2; p 〈 0.0001), platelets 〈 100 x 10(9)/L (HR 3.4; p 〈 0.0001), ASXL1 mutations (HR 2.2; p=0.0001) and EZH2 mutations (HR 2.7; p=0.001). A similar analysis identified SRSF2 mutations (HR 5.9; p=0.0003) and platelets 〈 100 x 10(9)/L (HR 4.3; p=0.005) as the only predictors of leukemia-free survival. Conclusions In NK-PMF, molecular markers might be prognostically more useful than conventional models that rely on clinical features. In the current study, thrombocytopenia was the only clinical variable, other than age, with additional value to molecular prediction of survival and leukemic transformation. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 2812-2812
    Abstract: Abstract 2812 Since the discovery of the JAK2-V617F mutation, the relative level of the mutant allele has been widely studied to gain further insights into the biology of myeloproliferative neoplasms and to establish utility in predicting clinical outcome. Mutant allele burden has been correlated with risk of thrombotic events in essential thrombocythemia, severity of the disease phenotype in polycythemia vera (PV) and survival in primary myelofibrosis (PMF). JAK2-V617F has also been quantified to assess disease response, with significant reductions in allele burden reported in PV patients following interferon (IFN) α-2b and pegylated IFN α-2a treatment. Moreover, serial DNA-based quantitative polymerase chain reaction (Q-PCR) assays have been used after allogeneic transplantation for myelofibrosis to predict outcome and guide donor lymphocyte infusion. With quantification of JAK2-V617F being considered an endpoint in a number of trials with novel agents including JAK2 inhibitors, it is important that assays are robust, mutant-specific and afford a suitable level of sensitivity. However, numerous JAK2-V617F Q-PCR assays have now been published, which based on experience of molecular monitoring in other hematological malignancies, are likely to vary markedly in their performance. Indeed, this concern was confirmed by the results of a quality control (QC) exercise involving 5 European LeukemiaNet (ELN) laboratories, each with their own established JAK2-V617F Q-PCR assay, who reported markedly differing levels of mutant allele percentage (ranging from 23–80%) in a QC sample distributed by UK NEQAS that comprised a 1 in 30 dilution of HEL cells (which harbor multiple copies of JAK2-V617F and lack the wild type [WT] JAK2 allele) in K562 cells (WT JAK2). To address this issue, a joint initiative was established within European LeukemiaNet between the Chronic Myeloproliferative Diseases and Minimal Residual Disease working groups (WP9 & WP12) to systematically evaluate a range of published and “in-house” JAK2-V617F assays, with the aim of identifying sensitive mutant-specific assays that perform similarly on different Q-PCR platforms – factors of fundamental importance for reliable tracking of disease response. The study involved 12 expert centers - in 6 European countries and included 1 US laboratory, which between them used Q-PCR platforms from 3 different manufacturers. The study involved central distribution of DNA from serial dilutions of JAK2-V617F mutant cell lines (HEL & /or UKE-1) in K562 and plasmid standards for JAK2 WT, JAK2-V617F and independent control genes (albumin, cyclophilin). Reagents for each assay were coded and also centrally distributed; reaction conditions for each coded assay were provided. Laboratories with established published or in-house Q-PCR assays were encouraged to perform these in parallel with the coded test assays on the distributed QC materials, to control for local variations in test conditions. Data from each QC round were submitted by participating laboratories for independent central analysis (Dept. of Genetics, King's College London); conclusions were drawn concerning the performance of the various assays before breaking the code to reveal their identities. Overall, 7 successive QC rounds were conducted, evaluating 9 assays (6 published, 3 unpublished “in-house”). Six assays were eliminated, due to insufficient/inferior sensitivity (n=5) or inconsistent performance (n=1). Of the 3 assays with better performance profile, the assay published by Lippert et al (Blood 2006;108 :1865) reliably quantified JAK2-V617F across the diagnostic range, with consistent results obtained between laboratories irrespective of Q-PCR platform. The 2 remaining assays (Larsen et al, Br J Haematol. 2007;136 :745 & in-house assay from the Oppliger Leibundgut lab [Bern]), afforded better sensitivity, capable of detecting a 0.08–0.008 % level of JAK2-V617F in DNA preparations from HEL and K562 cells. The latter assays were tested on serial post-transplant samples from 2 patients allografted for JAK2-V617F mutant PMF, successfully identifying the presence of residual disease, complementing chimerism analysis to predict clinical outcome. In conclusion, this ELN study has identified robust assays suitable for quantifying JAK2-V617F in clinical trials and which merit further investigation as a tool to guide management of patients undergoing allogeneic transplant. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 124, No. 16 ( 2014-10-16), p. 2507-2513
    Abstract: Survival in ET is superior to that of PV, regardless of mutational status, but remains inferior to the sex- and age-matched US population. JAK2/CALR/MPL mutational status is prognostically informative in PMF, regarding overall and leukemia-free survival.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 3040-3040
    Abstract: Background : Thrombosis is a characteristic feature in essential thrombocythemia (ET) and polycythemia vera (PV). Previous studies had focused on thrombotic events occurring after diagnosis and did not always distinguish between arterial and venous thrombosis. Arterial thrombosis in ET has been associated with age 〉 60 years, JAK2 mutations, leukocyte count 〉 11 x 109/l, history of thrombosis and cardiovascular risk factors, and venous thrombosis with male sex (Blood 2011;117:5857). In the current two-enter study, we looked for associations between both driver and other mutations, with first incidences of arterial and venous thrombosis in ET and PV, analyzed together and separately. Methods : Study patients were recruited from the Mayo Clinic, Rochester, MN, USA and the University of Florence, Florence, Italy, based on availability of next-generation sequencing-derived mutation information. Diagnoses were according to the 2016 World Health Organization criteria (Blood. 2016;127:2391). Conventional statistics was employed to examine significance of associations, with separate analysis of arterial vs venous thrombosis, occurring at any time before or after formal diagnosis of ET or PV. For the purposes of the current study, only first events were considered. Results: 906 molecularly-annotated patients (416 from Mayo and 490 from Florence), including 502 ET and 404 PV cases, were included in the current study. The Mayo/Florence cohorts included 270/232 ET (median age 57/54 years, 60%/59% females) and 146/258 PV (median age 63/58 years, 52%/44% females) patients; median follow-up was 9.9/12.9 years for ET and 10.7/12.4 years for PV. Driver mutation distribution in ET was 55% JAK2, 27% CALR, 5% MPL and 13% triple-negative. Most frequent mutations, other than JAK2/CALR/MPL, were TET2 (14%; 11% in ET and 18% in PV) and ASXL1 (13%; 13% in ET and 13% in PV). 301 (33%) patients experienced first time thrombosis, before or after diagnosis, including 152 (30%) in ET and 149 (37%) in PV (p=0.03); arterial events occurred in 174 (19%) patients, including 96 (19%) in ET and 78 (19%) in PV (p=0.9); venous events occurred in 177 (20%) patients, including 82 (16%) in ET and 95 (24%) in PV (p=0.007). The number of vascular events after diagnosis was 174 (19%) for arterial and 154 (17%) for venous thrombosis, with no significant difference between ET and PV. Associations with arterial thrombosis: In multivariable logistic regression analysis that included both ET and PV (n=906), age 〉 60 years (23% vs 16%; OR, 1.6, 95% CI 1.1-2.2) and absence of ASXL1 and RUNX1 mutations (21% vs 9%; OR 2.6, 95% CI 1.4-4.8) were associated with arterial thrombosis; these associations remained significant (or near significant) when ET and PV were analyzed separately; in addition, JAK2 mutations in ET displayed borderline significance (p=0.05). Overall incidence of arterial events in ET was estimated at 27% in the presence of all three risk factors (i.e. older age, presence of JAK2 mutations and absence of ASXL1/RUNX1 mutations), 21% with two risk factors, 13% with one risk factor and 9% in the absence of all three risk factors (p=0.01). Associations with venous thrombosis: In multivariable analysis, including both ET and PV (n=906), JAK2 mutations (p 〈 0.001), higher leukocyte count (p=0.03) and younger age (p=0.04) were identified as being significant. Analysis of ET patients only (n=502) identified JAK2 mutations (p 〈 0.001; OR 2.4, 95% CI 1.4-4.1), absence of SRSF2 mutations (p=0.03) and younger age (p=0.04) as independent risk factors and in PV patients (n=404), higher leukocyte count (p=0.06). Prognostic interaction between JAK2 mutations and age: In ET, the overall frequency of venous events was 27% for young patients with JAK2 mutations vs 13% for older JAK2 mutated cases vs 10% for JAK2 unmutated young patients vs 13% for JAK2 unmutated older patients (p 〈 0.001). The corresponding percentages for arterial events were 20%, 24%, 14% and 19%, respectively (p=0.1). Conclusions: JAK2 mutations contribute to the risk of both venous (younger patients) and arterial (young and older patients) thrombosis, in an age-dependent manner; the association with venous thrombosis might explain the higher incidence of venous thrombosis in PV, compared to ET. Additional studies are required to confirm the observed lower risk of thrombosis in patients with ASXL1, RUNX1 and SRSF2 mutations and provide insight into the underlying biological explanation. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 3513-3513
    Abstract: Mice with megakaryocytic-targeted ablation of GATA-1 expression (GATA-1low mice) develop while age a syndrome similar to idiopathic myelofibrosis. These mice present with life-long thrombocytopenia and extensive proliferation of differentiation-impaired megakaryocytes. Although GATA-1 affects erythroid cell production, GATA-1low mice are not anemic because of increased erythropoiesis in the spleen. At 8–10-month, GATA-1low mice develop anemia, osteosclerosis, marrow and spleen fibrosis and neoangiogenesis. From 15-month, extensive extramedullary hematopoiesis in the liver occurs. Myelofibrosis is developed by hemizygous male and heterozygous females alike. In order to evaluate the role of the spleen in the development of extramedullary hematopoiesis, young (6-month) wild-type littermates (WT) and GATA-1low mice (both hemizygous males and heterozygous females) were splenectomized. Three- and 9–12 months later, the myelofibrotic trait expressed by the treated animals was compared with that of un-manipulated controls. Although all the mice survived surgery, a dramatic difference in mortality was observed between hemizygous males, which died of severe anemia within 2-weeks, and heterozygous females (no death recorded). Heterozygous GATA-1low females developed a modest anemia 9–12-months after surgery (Htc, 40.1±2.7% vs. 48.2±5.5 in splenectomized GATA-1low and WT littermates, respectively, p & lt;0.01), comparable to that presented by the age-matched untreated mutants. Platelet counts increased 2-to-3-fold in both WT and GATA-1low mice, but remained significantly lower than normal in the latter. There was an increase in the number of CD16+ NK cells in the blood of splenectomized GATA-1low mice (287±61 vs. 61±19 CD16+ cells/mL in splenectomized GATA-1low and WT littermates, respectively), but no other abnormality in blood cell counts was observed. At 3 months post-splenectomy, the number of bone marrow cells in GATA-1low mice increased up to normal [from 4.0(±1.2)x106 to 8.0(±1.3)x106 cells/femur in untreated and splenectomized mutants, respectively]. Histologic evaluation of bone sections showed no overt change in the extent of marrow fibrosis compared to age-matched untouched mutants, while the extent of trabecular bone was reduced. No fibrosis developed in WT mice at any time point after splenectomy, nor were changes in total marrow cellularity observed. There was no sign of extramedullary hematopoiesis at 3 month post-splenectomy in either WT or GATA-1low mice, while at the later time point, mutant mice showed a prominent involvement of the liver with high numbers of megakaryocytes, isolated or in clusters, and maturing cells of all hematopoietic lineages, that in extreme cases diffusely infiltrated the parenchyma. Extramedullary hematopoiesis was not found in either the lung or the kidney. In conclusions, splenectomy affected the extent but not the timing of the development of extramedullary hematopoiesis in GATA-1low mice suggesting that the number of stem cells present in the marrow of old splenectomized animals was sufficient to colonize the liver. The observation that splenectomy per se does not result in extramedullary hematopoiesis support the hypothesis that it is not fibrosis of the spleen, but a second hit within the stem cell compartment, that is possibly responsible for the development of extramedullary hematopoiesis in this mouse model of myelofibrosis.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2005
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 104-104
    Abstract: The median survival (OS) of patients with primary myelofibrosis (PMF; 6.5y) is significantly shortened compared to reference population (Cervantes et al, JCO 2012; 30:2981). OS is predicted by the four risk categories of IPSS, Dynamic-IPSS and DIPSS-plus score, nevertheless pts heterogeneity persists within these categories, necessitating improved risk stratification. We recently reported that pts harboring mutations in any one of the prognostically relevant ASXL1, EZH2, IDH1/2 and SRSF2 genes constitute a IPSS-and DIPPS-plus independent Molecular High Risk category (MHR+) characterized by significant reduction of OS and leukemia free survival (LFS) (Vannucchi et al, Leukemia 2013). The aim of this work was to analyze the impact of the number of prognostically relevant mutated genes on OS and LFS in PMF. Patients and methods Two independent cohorts are included, a “test cohort” from Europe, analyzed at diagnosis, and a “validation cohort” from Mayo Clinic, analyzed at any time after diagnosis. Mutation analysis was performed in DNA from whole blood or granulocytes using RTQ-PCR, HRM and direct sequencing; all mutations were confirmed at least twice. The prognostic value of the molecular variables with regard to OS and LFS was analyzed by Cox regression. Test cohort It included 490 pts (median age 61y; males = 301) risk stratified by IPSS into high (n=74, 15%), intermediate-2 (n=93, 19% ), intermediate-1 (n=147, 30%) and low (n=176, 36%). The median follow-up was 3.63y (95% CI, 0.06-28.33); 161 pts died (33.0%), of whom 76 (15.6%) had progressed to acute leukemia after a median of 3.4y (0.04-28.3) from diagnosis. One hundred forty-six pts (29.8%) presented at least one of the four aforementioned mutated genes and were classified as MHR+. The OS of MHR+ pts was significantly reduced compared to patients with no mutations (n=344): 80.7 vs 148.9 mo (HR 2.2, CI95% 1.6-3.03). One hundred twelve (22.8%) pts had 1 mutation and 34 (6.9%) had 2 or more mutations. In univariate analysis presence of 2 or more mutated genes was significantly more detrimental for OS (29.5mo; HR 4.12, IC95% 2.6-6.4) than having 1 mutated gene (84.2mo; HR 1.8, IC95% 1.2-2.5) compared with having no mutations (148.9mo). Multivariate analysis results adjusted for IPSS indicated that having two or more mutations was an independent prognostic factor for OS (HR 2.9, 95% CI 1.8-4.5). Notably, the prognostic relevance of harboring two or more mutations involved both lower (low and intermediate-1; HR 1.87, 95% CI 1.3-2.6) and higher (intermediate-2 and high; HR 1.6, 95% CI 1.2-2.1) categories of IPSS. Also LFS resulted significantly shorter (129mo; HR 3.1, 95%CI 1.9-4.8) in MHR+ pts compared with pts with no adverse mutations (323mo). Having 2 or more mutations correlated with greater reduction of LFS (79.6mo; HR 7.02, CI95% 3.9-12.6) than having one mutation only (133.9mo; HR 2.2, IC95% 1.3-3.7) as compared with no mutations (323.2mo). Multivariate analysis showed that IPSS high risk category (HR 4.5; 95% CI 2.3-8.8) and two or more mutated genes (HR 5.3; 95% CI 2.9-9.8) predicted independently for a significant reduction in LFS. The negative impact on LFS of having 2 or more mutated genes compared to one or no mutations was maintained in the lower (HR 6.4, 95% CI 2.6-15.2) and higher (HR 5.5, 95% CI 2.3-12.8) risk categories. Validation cohort Validation cohort included 262 patients (median age 64 years; males = 167) risk stratified by DIPSS-plus into high (n=89, 34%), intermediate-2 (n=92, 35%), intermediate-1 (n=46, 18%) and low (n=34, 13%). One hundred forty-six pts (56%) displayed none of the four aforementioned mutations, 93 (36%) harbored one mutation whereas 23 (9%) harbored two or more mutations. In univariate analysis, having two or more mutations (HR 3.7; 95% CI 2.2-6.1) was significantly more detrimental for OS than having no mutations, which is more favorable than having one mutation (HR 1.9; 95% CI 1.4-2.7). When adjusted for DIPSS-plus, the presence of two or more mutations retained its significance (HR 2.1; 95% CI 1.3-3.6) and outperformed ASXL1 mutation alone in its prognostic relevance Conclusions Overall, these results show that the number of prognostically relevant mutated genes correlate with OS and LFS in pts with PMF, suggesting that screening for these mutations might help to improve risk stratification. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 118, No. 19 ( 2011-11-10), p. 5227-5234
    Abstract: We genotyped 370 subjects with primary myelofibrosis (PMF) and 148 with postpolycythemia vera/postessential thrombocythemia (PPV/PET) MF for mutations of EZH2. Mutational status at diagnosis was correlated with hematologic parameters, clinical manifestations, and outcome. A total of 25 different EZH2 mutations were detected in 5.9% of PMF, 1.2% of PPV-MF, and 9.4% of PET-MF patients; most were exonic heterozygous missense changes. EZH2 mutation coexisted with JAK2V617F or ASXL1 mutation in 12 of 29 (41.4%) and 6 of 27 (22.2%) evaluated patients; TET2 and CBL mutations were found in 2 and 1 patients, respectively. EZH2-mutated PMF patients had significantly higher leukocyte counts, blast-cell counts, and larger spleens at diagnosis, and most of them (52.6%) were in the high-risk International Prognostic Score System (IPSS) category. After a median follow-up of 39 months, 128 patients (25.9%) died, 81 (63.3%) because of leukemia. Leukemia-free survival (LFS) and overall survival (OS) were significantly reduced in EZH2-mutated PMF patients (P = .028 and P 〈 .001, respectively); no such impact was seen for PPV/PET-MF patients, possibly due to the low number of mutated cases. In multivariate analysis, survival of PMF patients was predicted by IPSS high-risk category, a 〈 25% JAK2V617F allele burden, and EZH2 mutation status. We conclude that EZH2 mutations are independently associated with shorter survival in patients with PMF.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood Advances, American Society of Hematology, Vol. 1, No. 1 ( 2016-11-29), p. 21-30
    Abstract: More than half of patients with PV or ET harbor DNA mutations/variants other than JAK2/CALR/MPL. The presence of some of these mutations adversely affects overall, leukemia-free, or myelofibrosis-free survival.
    Type of Medium: Online Resource
    ISSN: 2473-9529 , 2473-9537
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 2876449-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...