Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Genes, Chromosomes and Cancer, Wiley, Vol. 58, No. 11 ( 2019-11), p. 747-755
    Abstract: Sequential genotyping for phenotype‐driver mutations in JAK2 (exon 14), CALR (exon 9), and MPL (exon 10) is recommended in patients with myeloproliferative neoplasms. Yet, atypical JAK2‐ and MPL‐ mutations were described in some triple‐negative patients. Whether noncanonical and/or concomitant JAK2‐ and MPL‐ mutations exist in myelofibrosis (MF) regardless of phenotype‐driver mutations is not yet elucidated. For this, next‐generation sequencing (NGS) was performed using blood genomic DNA from 128 MF patients (primary MF, n = 93; post‐ET–MF, n = 18; post‐PV–MF, n = 17). While no atypical JAK2‐ or MPL‐ mutations were seen in 24 CALR‐ positive samples, two JAK2‐ mutations [c.3323A  〉  G, p.N1108S; c.3188G  〉  A, p.R1063H] were detected in two of the 21 (9.5%) triple‐negative patients. Twelve of the 82 (14.6%) JAK2 V617F‐positive cases had coexisting germline JAK2‐ mutations [ JAK2 R1063H, n = 6; JAK2 R893T, n = 1; JAK2 T525A, n = 1] or at least one somatic MPL‐ mutation [ MPL Y591D, n = 3; MPL W515 L, n = 2; MPL E335K, n = 1]. Overall, MPL‐ mutations always coexisted with JAK2 V617F and/or other MPL‐ mutations. None of the JAK2 V617F plus a second JAK2‐ mutation carried a TET2‐ mutation but all patients with JAK2 V617F plus an MPL‐ mutation harbored a somatic TET2‐ mutation. Four genomic clusters could be identified in the JAK2 V617F‐positive cohort. Cluster‐I (10%) (noncanonical JAK2 mutated (mut)  +  TET2 wildtype (wt) ) were younger and had less proliferative disease compared with cluster‐IV (5%) ( TET2 mut  +  MPL mut ). In conclusion, recurrent concomitant classical and/or noncanonical JAK2‐ and MPL‐ mutations could be detected by NGS in 15.7% of JAK2 V617F‐ and MPL W515‐positive MF patients with genotype‐phenotype associations. Many of the germline and/or somatic mutations might act as “Significantly Mutated Genes” contributing to the pathogenesis and phenotypic heterogeneity. A cost‐effective NGS‐based approach might be an important step towards patient‐tailored medicine.
    Type of Medium: Online Resource
    ISSN: 1045-2257 , 1098-2264
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2019
    detail.hit.zdb_id: 1018988-9
    detail.hit.zdb_id: 1492641-6
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancers, MDPI AG, Vol. 13, No. 2 ( 2021-01-07), p. 186-
    Abstract: Background: Myelodysplastic syndromes (MDS) are caused by a stem cell failure and often include a dysfunction of the immune system. However, the relationship between spatial immune cell distribution within the bone marrow (BM), in relation to genetic features and the course of disease has not been analyzed in detail. Methods: Histotopography of immune cell subpopulations and their spatial distribution to CD34+ hematopoietic cells was determined by multispectral imaging (MSI) in 147 BM biopsies (BMB) from patients with MDS, secondary acute myeloid leukemia (sAML), and controls. Results: In MDS and sAML samples, a high inter-tumoral immune cell heterogeneity in spatial proximity to CD34+ blasts was found that was independent of genetic alterations, but correlated to blast counts. In controls, no CD8+ and FOXP3+ T cells and only single MUM1p+ B/plasma cells were detected in an area of ≤10 μm to CD34+ HSPC. Conclusions: CD8+ and FOXP3+ T cells are regularly seen in the 10 μm area around CD34+ blasts in MDS/sAML regardless of the course of the disease but lack in the surrounding of CD34+ HSPC in control samples. In addition, the frequencies of immune cell subsets in MDS and sAML BMB differ when compared to control BMB providing novel insights in immune deregulation.
    Type of Medium: Online Resource
    ISSN: 2072-6694
    Language: English
    Publisher: MDPI AG
    Publication Date: 2021
    detail.hit.zdb_id: 2527080-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 1307-1307
    Abstract: Abstract 1307 Iron homeostasis is controlled through a coordinated activity of multiple proteins through different signaling pathways with hepcidin being the central regulatory hormone. Iron overload in recipients of allogeneic hematopoietic cell transplantation (HCT) is associated with an adverse outcome. As causes of disturbed iron homeostatsis are multifactorial with transfusional iron playing a key role, we analysed prospectively the outcome after HCT with pre-transplant patterns of potential interactions between the units of red blood cells (URB) transfused, serum hepcidin (SH), and ferritin (SF). For this purpose, 3 quotients were generated (SF/URB, SF/SH, SH/URB). All parameters were measured 7 days pre-HCT with a median C-reactive protein of 〈 5 mg/l. From February, 2008–January, 2010, 111 consecutive patients (pts) [58 males/53 females, median age 57 (range 19–75) years] with AML in remission (n=85) and MDS (n=26) who underwent HCT at the University of Leipzig were studied. Donors were matched related (MRD) in 23 (21%) and matched unrelated (MUD) in 88 (79%) pts. Preparative regimen was conventional conditioning (CC) with 12 Gy TBI/cyclophosphamide 120 mg/kg in 41 (40%) and RIC with fludarabin 30 mg/m2/day for 3 days and2 Gy TBI in 70 (60%) pts. Median SH measured by C-ELISA was 282 (range 6–1595) ng/ml. Median SF was 1869 (range 36–17995) ng/ml with SF 〉 1000 ng/ml present in 91 (82%) pts. Median URB was 24 (0-95) units. There was a poor linear correlation of SH with SF (r=0.58) and URB (r=0.35). After a median follow up of 16 (5-28) months, OS, EFS, TRM, and RI were 66%, 53%, 17%, and 30% respectively. In multivariate analysis, SH, SF, and URB had no impact on OS, EFS, RI or TRM. The incidences of acute (a) GvHD 〉 grade II was 48%. It was associated with MUD (p=0.004) and the presence of an HLA-mismatch (p=0.02). Chronic (c) GvHD occurred in 46% of pts with 33/104 (32%) pts suffering from extensive cGvHD. RIC (p=0.001), MUD (p=0.06), and a higher SF (p=0.02) correlated with an increased incidence of cGvHD. SH, and URB had no influence on aGvHD or cGvHD. A higher SF (p=0.02) and CC (p=0.001) correlated with the 93 viral, bacterial and fungal infections 〉 grade 3 (according to common toxicity criteria) in the first 100 days post-HCT. SF/SH, and SH/URB had no impact on outcome. On the other hand, SF/URB 〈 75% correlated strongly with an improved OS (p=0.06), and EFS (p 〈 0.0005) as well as a lower TRM (p=0.02), and cGvHD (p=0.06) in both uni- and multivariate analysis. EFS in pts with SF/URB 〈 75% was 60% versus 33% for those with a higher SF/URB (p=0.01) mainly because of a significantly lower TRM of 11% for SF/URB 〈 75% versus 33% for SF/URB 〉 75% (p=0.002). In the absence of relapse, the probability of one-year survival in a patient with SF/URB 〈 75% is 86% compared to 50% for a patient with SF/URB 〉 75% (p=0.001). Median pre-transplant SF of 1844 (range 567–6842) ng/ml was significantly lower (p=0.003) and median URB of 26 (8-95) units higher (p=0.005) in pts with SF/URB 〈 75% compared to pts with higher SF/URB [median SF 2573 (range 463–17995) ng/ml, median URB 19 (range 2–63) units]. SH in both SF/URB groups was similar (p=0.7). SF/URB was not influenced by age, gender, diagnosis, type of conditioning and donor, number of previous therapies, CMV risk status, and female to male HCT. This simple ratio, if validated in larger studies, might provide clinicians prior to HCT with a reliable tool for assessment of TRM after HCT for AML and MDS. Unlike ferritin alone which is a sequel of intracellular iron metabolism, this ratio reflects some of the often simultaneously disturbed iron sensor pathways for hepcidin regulation such as erythropoietic activity, inflammation and iron stores in pts with haematological disorders undergoing HCT. Disclosures: Al-Ali: Novartis: Consultancy, Honoraria, Research Funding. Westerman:Intrinsic Life Sciences: Employment, Membership on an entity's Board of Directors or advisory committees. Hehme:Novartis: Employment. Niederwieser:Bristol-Myers Squibb: Speakers Bureau; Novartis: Speakers Bureau.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 396-396
    Abstract: Mastocytosis is a hematopoietic disorder characterized by abnormal growth and accumulation of neoplastic mast cells (MC) in various organ systems. Using updated WHO criteria and the proposal of the consensus group, the disease can be divided into cutaneous mastocytosis (CM), indolent systemic mastocytosis (ISM), smouldering SM (SSM), SM with an associated hematologic neoplasm (SM-AHN), aggressive SM (ASM), and mast cell leukemia (MCL). In adult patients with skin involvement but unknown/unavailable bone marrow (BM) studies, the provisional diagnosis ´mastocytosis in the skin (MIS)´ is appropriate. Although this classification has been validated repeatedly and is of prognostic significance, additional prognostic parameters have been identified in recent years (yrs). We have established a patient-registry in the ECNM where over 1,000 cases with confirmed mastocytosis are included. The aim of this study was to identify and validate new prognostic variables predicting survival in patients with mastocytosis and to prepare a simple prognostic scoring system applicable in daily practice. Using the data set of the ECNM registry we analyzed overall survival (OS) and event-free survival (EFS; i.e. until death or progression) in 1,088 patients with mastocytosis (median age: 45.7 yrs; range: 0.1-83.3 yrs, f:m ratio, 1:0.79), including CM (n=152), MIS (n=126), ISM (n=650), SSM (n=26), SM-AHN (n=89), ASM (n=35), and MCL (n=10). The median observation period was 3.5 yrs (75-25% percentile: 1.5-6.9 yrs, maximum 34.1 yrs). In the entire cohort, the median OS was not reached. The probability to be alive after 5, 10, and 20 yrs was 89%, 83%, and 70%, respectively. As expected, the WHO classification turned out to be of utmost predictive significance (Figure 1A; p 〈 0.005). In patients with non-advanced disease, namely CM, MIS, ISM, and SSM, the median survival was not reached, and the survival at 5 yrs was 100%, 97%, 98%, and 85%, respectively, whereas in advanced SM, namely SM-AHN, ASM, and MCL, the median survival was 2.8, 4.1, and 0.8 yrs, respectively. Patients with advanced SM were found to be older, to have higher serum tryptase- and alkaline phosphatase (aPhos) levels, higher white blood counts (WBC), lower hemoglobin (Hb) and platelet (PLT) counts, and more frequently presented with organomegaly (hepatomegaly, splenomegaly, or lymphadenopathy). Moreover, the male/female ratio was higher in advanced mastocytosis. To define the relative impact of the identified risk factors we randomly divided the total cohort (50:50) into a learning set and a validation set, performed uni- and multivariate analyses, and subsequently calculated cut off values for optimal prognostication. In these studies, the poor prognosis of patients with advanced SM was confirmed. In patients with non-advanced disease, all variables tested were significant concerning OS in univariate analyses. However, WBC, Hb, and lactate dehydrogenase had to be excluded due to variance inflation. In multivariate analyses, age 〉 70 yrs, PLT 〈 80 G/L (not related to SM), and aPhos ≥240 U/L were significant predictors concerning OS. Based on these parameters, we established a simple prognostic scoring system. In this score, patients with non-advanced disease (CM, MIS, ISM, SSM) without additional risk factors comprised the low risk group, those with non-advanced disease and presence of one or more risk factors (age 〉 70 yrs, PLT 〈 80 G/L, aPhos ≥240 U/L) the intermediate risk group, and those with advanced disease (with or without additional risk factors) the high risk group. The median OS in the low risk group was not reached, in the intermediate risk group it was 13.5 yrs, and in the high risk group 3.5 yrs (p 〈 0.005; Figure 1B). Significant differences were also observed regarding EFS (p 〈 0.005, Figure 1C). In conclusion, the WHO classification remains the gold-standard of prognostication in patients with mastocytosis, but additional factors, namely age, PLT, and aPhos, are powerful additional variables predicting OS in these patients. Based on in-depth analyses of the ECNM registry data-set, a simple prognostic scoring system for mastocytosis was established and is recommended to define the probability of OS and EFS in patients with mastocytosis in daily practice. Disclosures Sperr: Amgen: Honoraria, Research Funding; Novartis: Honoraria. Gotlib:Incyte Corporation: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Valent:Amgen: Honoraria; Celgene: Honoraria, Research Funding; Novartis: Honoraria, Research Funding; Deciphera Pharmaceuticals: Research Funding; Ariad: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 5223-5223
    Abstract: The significance of host- and disease-related prognostic factors on outcome in patients (pts) with MDS and AML with MDS-related changes (sAML) depends on the treatment given. The impact of therapy opposed to prognostic variables on the heterogeneity of MDS and sAML was investigated. Patients and methods From January, 2004-August, 2012, 367 pts (MDS, n=208; sAML, n=159) consecutively treated (median age 63y) at the University of Leipzig were included. Patients (84%) with marrow blasts 〉 10% received induction chemotherapy (CT; 59%) or azacitidine (AZA; 25%) (after its approval in the EU in January, 2009) with the intention of performing a subsequent allogeneic hematopoietic cell transplantation (HCT) in pts 〈 70y. Up-front HCT was scheduled if blasts were 〈 10% (n=56). Cytogenetics were categorized according to Schanz et al, JCO 2012 for MDS and the WHO classification for sAML. As confounders in the estimation of therapy, host- and disease-related features were investigated in a multistep process. 38% of pts had 〉 2 comorbidities with no difference between MDS and sAML. The sAML group (median blasts 40%) included 69 and 81 pts with previous MDS and MDS-related cytogenetic abnormality respectively. Cytogenetics were poor and very poor in 34% of MDS. Outcome at two years are presented. Results Median interval between diagnosis of MDS and therapy was 3.6 months. Median survival time for sAML was 15 vs 72 months for MDS (p 〈 0.0005). Overall, age was higher (median 68y) and blasts lower (median 13%) in the AZA group compared to CT (62y and 27%) (p 〈 0.0005). Cytogenetics and the comorbidity burden (CB) were comparable. OS with AZA was similar to up-front HCT (68%) and superior to CT (48%) (p=0.01). OS was 50% if HCT was performed after CT (136 pt) compared to CT alone (p=0.01). In the 20% of pts 〉 70y, AZA was given to 52% and CT to 48%. OS was 55% and best with AZA (p=0.01). Median survival times were 30 for AZA/MDS, 27 for AZA/AML, 15 for CT/AML, and 5 months for CT/MDS. Of the 110 pts 〈 70y with MDS, AZA was given to 50 (45%) and CT to 60 (55%). The IPSS, cytogenetics, CB, BM blasts (10% vs 11.5%) were similar in both groups. With a median age of 63y, the AZA/MDS group was older than the CT/MDS group (median age 60y) (p=0.005). OS for both groups was 68%. NRM (16%) and RI (38% vs 34%) were alike. For the 114 pts 〈 70y with sAML (median age 62%; median blasts 44%) treated with CT, OS was 40% and inferior to MDS (AZA/MDS, p=0.007; CT/MDS, p=0.01) due to higher RI (57%) (p=0.008). Overall, 218 (78%) pts 〈 70y received HCT (after a median of 3 AZA cycles for AZA/MDS). Ferritin, cytogentics, CB, type of donor, and blasts at HCT (median 4%) were comparable in the transplant groups (AZA/MDS, CT/MDS, HCT up-front, CT/sAML). Irrespective of prior therapy (p=0.6), interval between therapy and HCT, and blasts 〈 5 vs 〉 5- 〈 10%, outcome in the MDS groups (OS, 60%, NRM 29%, RI, 32%) was similar. In multivariate analysis, 〉 2 comorbidities and very poor cytogenetics were associated with an inferior OS (p=0.001)and a higher RI (p=0.003). With a median survival time of 11 months for sAML and a RI of 49%, outcome after HCT for sAML was inferior to MDS (p=0.005). In multivariate analysis, blasts 〈 5%, 〉 2 comorbidities were associated with a poor outcome. For MDS/CT and sAML/CT, a complex karyotype (38%) tended to decrease OS (p=0.06) and increase RI (p=0.01) after HCT. Conclusions Treatment was able to reduce the significance of most negative host- and disease-specific prognostic factors on outcome in MDS. AZA is superior to CT in elderly patients and equal to CT in younger patients with MDS and seems to have no negative impact on outcome after HCT. Despite the improvement achieved with allogeneic HCT, AML with MDS-related changes remains a distinct clinic-pathologic entity associated with a worse outcome. Genetics rather than marrow blasts are an important determinant of prognosis after treatment including allogeneic HCT. 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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 2723-2723
    Abstract: Introduction: Bortezomib is a novel proteasome inhibitor that has shown important clinical efficacy either as a single agent or in combination with other cytostatic agents in relapsed/refractory multiple myeloma (MM). The combinated treatment of bortezomib with bendamustine and prednisone (BPV) was assessed to determine the efficacy and toxicity of this regiment in patients with advanced MM. Methods: Between January 2005 and July 2007, 46 patients (median age 63; range 31–77 years) with relapsed or refractory MM (29 patients stage III a, 17 patients III b) were treated with bendamustine 60 (−80) mg/qm on day 1 and 2, bortezomib 1,3 mg/qm on day 1, 4, 8 and 11, and prednisone 100 mg on day 1, 2, 4, 8 and 11. Cycles were repeated every 21 days until maximum response or progressive disease. The time from first diagnosis ranged from 1 to 183 (median 36) months. The duration of the last remission before beginning the BPV-therapy was 6 (range 0–36) months. Previous therapy lines (median 2, range 1–6) included 18 × thalidomide, 10 × autologous PBSCT, and 9 × autologous/allogeneic PBSCT. 16 patients were refractory to the last treatment. 22 patients had preexistent severe thrombocytopenia, leukocytopenia or anemia (WHO grade 3 or 4). Response was assessed using EBMT criteria modified to include near complete remission (nCR) and very good partial remission (VGPR). Results: 36 patients (78%) responded after at least one cycle of chemotherapy with 2 CR, 5 nCR, 6 VGPR, 15 PR and 8 MR. 4 patients had stable disease and 6 patients had a progress. With a median follow up of 13 months, EFS, and OS at twelve months for patients without severe haematological toxicities due to previous treatments (n=24) were 46% and 79%, respectively. Outcome for these patients was significantly better compared to patients with severe haematological toxicities (grade 3 or 4, n=22) where EFS, and OS were 10% and 22%, respectively (p 〈 0,01). The median number of the BPV-treatment was 2 (1–7) cycles. 20 of 36 responding patients showed a rapid decrease of the myeloma protein and reached the best response after the first cycle and 12 after the second cycle. The regimen was well-tolerated with few significant side effects reported. New cytopenias occured infrequently (four patients had a thrombocytopenia grade 3, and two patients had a grade 4 thrombocytopenia). 1 patient had a moderate new polyneuropathy (grade 2). Summary: These results indicate that the combination of bortezomib, bendamustine and prednisone is effective and well tolerated in a heavily pretreated population of patients with relapsed or refractory MM.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 1860-1860
    Abstract: In retrospective studies, CALR mutation is found to be a favorable prognostic variable in myelofibrosis (MF) compared with JAK2, or MPL mutations. With the availability of the JAK1/JAK2 inhibitor ruxolitinib (RUX) for treatment of MF, it is not yet known whether response varies across mutational subgroups and whether the prognostic implication of the driver mutations in terms of survival (OS) changes under RUX. We studied the prevalence and prognostic weight of clinical parameters in the IPSS and DIPSS-plus and the impact of RUX on OS in the context of mutation status. Patient and Methods: As of November 2009, RUX for treatment of splenomegaly and/or constitutional symptoms became an option at the University of Leipzig. All patients (pt) with MF seen since then were included (n=127; f=43%, median age 58 years) with the exception of pt with MPL mut+ because of small number. Screening for the JAK2V617F and CALR mutations was performed as previously published. Cytogenetic-risk categorization was conducted as published (Caramazza et al. Leukemia 2011). Results: JAK2 mut+ (group A; 60.6%) constituted the largest group, followed by CALR mut+ (group B; 19.7%), and triple-negative (group C; 19.7%). The median duration of MF was 22 months with no difference in the 3 groups. Higher-risk IPSS was present in the majority [Int-II 34.4%; high-risk 42.4%]. Constitutional symptoms were present across all groups, although more frequent in group A which was also characterized by prominent splenomegaly (Table). Group B pt were younger with low WBC in contrast to goup C pt who were older, anemic, with high WBC and low platelets, unfavorable cytogenetics and high-risk IPSS. Hb 〈 100 g/L (54%), and transfusion-dependency (32%) were frequently present in group B and was similar to group A. RUX was given to 72 (57%) pt [graoup A (66%), group B (56%), group C (29%)] with a median exposure of 8 months. The response of 80% was not influenced by the mutation status, cytogenetics, or IPSS variables. Leukemic transformation was documented in 22 pt [14 (64%) pt had no RUX exposure] after a median of 18 months after diagnosis. The incidence was highest in group C (36%) and lowest in group B (4%) (p=0.004). Unfavorable cytogenetics were 75%, and 27% in group C and A respectively. After a median follow-up of 31 months, OS at 3-years was 82% and worst in group C (72%) vs 80% for group A (p=0.05) vs 96% in goup B (p=0.003). In univariate analysis, non-mutated status (p=0.02), Int-2/high-risk IPSS (p=0.06), anemia (p=0.03), transfusion dependency (p=0.04), and platelets 〈 100 x109/L (p=0.06) but not unfavorable cytogenetics were associated with an inferior OS. In multivariate analysis, advanced IPSS only (p=0.04) but not the mutation status (p=0.4) retained its negative impact on OS. Generally, in group A pt treated with RUX (n=50), OS was only marginally inferior to group B (p=0.08). In pt with int-2/high-risk IPSS, OS in group A with RUX (n=41) was similar to that of group B (p=0.4). Conclusions: The mutation status in MF bestows distinct clinical phenotypes and has crucial prognostic and therapeutic implications. Patients with non-mutated MF had the worst prognosis, unfavourable cytogenetics, and the highest rate of leukemic transformation. Although the IPSS retains its value, the prognostic power of certain factors such as anemia and constitutional symptoms in CALR-mutated pt needs to be re-evaluated. Response to ruxolitinib seems to be independent of the mutation status. More importantly, a JAK1/JAK2 inhibition appears to be capable of attenuating the prognostic implication of the different mutation profiles by improving the less-favorable survival associated with non-CALR-mutated MF through a disease-modifying effect. The full potential of a sustained JAK1/JAK2 inhibition in modifying survival in the context of the various mutational profiles needs to be further studied in a larger cohort of patients. Table Clinical variables in patients with different driver mutations Variable JAK2 mut+ CALR mut+ Triple-negative p N (%) 77 (60.6) 25 (19.7) 25 (19.7) Median age (years) 59 51 61 0.02 Constitutional symptoms (%) 69 52 46 0.08 Median palpable spleen (cm) 10 4 3 0.001 Int-2/high-risk IPSS (%) 79 52 96 0.01 WBC 〉 25 x109/L (%) 21 11.5 44 0.008 Median peripheral blasts (%) 1 0.5 2 ns Median Hb (g/L) 106 99 86 0.008 Hb 〈 100 g/L (%) 42 54 80 0.03 Transfusion dependency (%) 33 32 67 0.009 Platelets 〈 100 x109/L (%) 30 8 52 0.04 Unfavorable cytogenetics (%) 31 14 48 0.06 Disclosures Al-Ali: Novartis: Honoraria, Research Funding. Jaekel:Novartis: Honoraria. Lange:Novartis: Honoraria. Roskos:Novartis: Honoraria. Niederwieser:Novartis: Honoraria, 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: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood Advances, American Society of Hematology, Vol. 7, No. 14 ( 2023-07-25), p. 3403-3415
    Abstract: In order to elucidate mechanisms for severe acute respiratory syndrome coronavirus 2 vaccination success in hematological neoplasia, we, herein, provide a comprehensive characterization of the spike-specific T-cell and serological immunity induced in 130 patients in comparison with 91 healthy controls. We studied 121 distinct T-cell subpopulations and the vaccination schemes as putative response predictors. In patients with lymphoid malignancies an insufficient immunoglobulin G (IgG) response was accompanied by a healthy CD4+ T-cell function. Compared with controls, a spike-specific CD4+ response was detectable in fewer patients with myeloid neoplasia whereas the seroconversion rate was normal. Vaccination-induced CD4+ responses were associated to CD8+ and IgG responses. Vector-based AZD1222 vaccine induced more frequently detectable specific CD4+ responses in study participants across all cohorts (96%; 27 of 28), whereas fully messenger RNA-based vaccination schemes resulted in measurable CD4+ cells in only 102 of 168 participants (61%; P  & lt; .0001). A similar benefit of vector-based vaccination was observed for the induction of spike-specific CD8+ T cells. Multivariable models confirmed vaccination schemes that incorporated at least 1 vector-based vaccination as key feature to mount both a spike-specific CD4+ response (odds ratio, 10.67) and CD8+ response (odds ratio, 6.56). Multivariable analyses identified a specific CD4+ response but not the vector-based immunization as beneficial for a strong, specific IgG titer. Our study reveals factors associated with a T-cell response in patients with hematological neoplasia and might pave the way toward tailored vaccination schemes for vaccinees with these diseases. The study was registered at the German Clinical Trials Register as #DRKS00027372.
    Type of Medium: Online Resource
    ISSN: 2473-9529 , 2473-9537
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2023
    detail.hit.zdb_id: 2876449-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Environmental Management, Springer Science and Business Media LLC, Vol. 37, No. 2 ( 2006-2), p. 247-257
    Type of Medium: Online Resource
    ISSN: 0364-152X , 1432-1009
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2006
    detail.hit.zdb_id: 1478932-2
    detail.hit.zdb_id: 131372-1
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Allergy, Wiley, Vol. 75, No. 8 ( 2020-08), p. 1927-1938
    Abstract: In indolent systemic mastocytosis (ISM), several risk factors of disease progression have been identified. Previous studies, performed with limited patient numbers, have also shown that the clinical course in ISM is stable and comparable to that of cutaneous mastocytosis (CM). The aim of this project was to compare the prognosis of patients with ISM with that of patients with CM. Methods We employed a dataset of 1993 patients from the registry of the European Competence Network on Mastocytosis (ECNM) to compare outcomes of ISM and CM. Results We found that overall survival (OS) is worse in ISM compared to CM. Moreover, in patients with typical ISM, bone marrow mastocytosis (BMM), and smoldering SM (SSM), 4.1% of disease progressions have been observed (4.9% of progressions in typical ISM group, 1.7% in BMM, and 9.4% in SSM). Progressions to advanced SM were observed in 2.9% of these patients. In contrast, six patients with CM (1.7%) converted to ISM and no definitive progression to advanced SM was found. No significant differences in OS and event‐free survival (EFS) were found when comparing ISM, BMM, and SSM. Higher risk of both progression and death was significantly associated with male gender, worse performance status, and organomegaly. Conclusion Our data confirm the clinical impact of the WHO classification that separates ISM from CM and from other SM variants.
    Type of Medium: Online Resource
    ISSN: 0105-4538 , 1398-9995
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2020
    detail.hit.zdb_id: 2003114-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages