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. 114, No. 22 ( 2009-11-20), p. 2783-2783
    Abstract: Abstract 2783 Poster Board II-759 Myelodysplastic syndromes are a heterogeneous group of bone marrow (BM) disorders characterised by clonal proliferation. The diagnosis of MDS can be challenging, especially in cases with minimal dysplastic changes by morphology and without characteristic karyotypic abnormalities. Recent investigations show that multi parameter analysis of BM samples of MDS patients by flow cytometry (FCM) might be of value in the diagnosis of MDS and for discriminating MDS from other disease entities. Detection of immunophenotypic abnormalities in the immature and the maturing myelomonocytic compartment is even included as a co-criterion in the minimal diagnostic criteria for MDS (Valent et al., Leuk Res 2009,31:727–36). Integration of FCM as a diagnostic tool in MDS requires a simple method with high sensitivity and specificity with low inter laboratory variation. Recently, reduced CD38 expression was demonstrated to discriminate between MDS and other disease entities (Goardon et al., Haematologica 2009, 94:1160–63).To validate the value of CD38 in the diagnosis of MDS, we analysed BM samples of 27 MDS patients (refractory anemia n=4; refractory anemia with multi lineage dysplasia n=15; refractory anemia with excess blasts [RAEB] -1 n=4 and RAEB-2 n=4), age-matched healthy volunteers (n=16) and pathologic controls (n=39). The pathologic control group consisted of aplastic anemia (n=3), iron deficiency (n=6), Multiple Myeloma (n=8), B-cell non-Hodgkin lymphoma (n=6) and autoimmune thrombocytopenia (n=3) patients. We found that CD38 RMFI on CD34+ cells was significantly decreased in MDS with less than 5% blasts (mean±SD: 216.5±95.4) as compared to pathologic controls (mean±SD: 377.2±246.1, P=0.01) and healthy volunteers (mean±SD: 322.8±138.1, P=0.02). In RAEB-1/-2, CD38 RMFI (mean±SD: 111.3±74.9, P=0.001) was even more decreased in comparison with both pathologic controls and healthy volunteers. Expression of CD38 on CD34+ cells is heterogeneous and the most immature cells or hematopoietic stem cells are considered to be negative for CD38 expression. By using the median fluorescence intensity (RMedFI), the skewed distribution of CD38 expression on CD34+ cells is taken into account. In our cohort, RMedFI is significantly decreased in MDS with 〈 5% blasts (mean±SD: 368.1±210.3, P=0.03) and MDS RAEB -1/-2 (mean±SD: 139.7±108.5, P 〈 0.001) as compared to pathologic controls (mean±SD: 521.6±314.2). However, no significant differences were observed between RMedFI of CD38 of MDS with less than 5% blasts and healthy volunteers. In conclusion, we show that CD38 expression is reduced in CD34+ cells of MDS patients and might be of value in the diagnosis of MDS. However, there is considerable overlap in CD38 expression between the MDS with less than 5% blasts group and pathologic controls group and healthy volunteers group. Differentiation of MDS from other disease entities by FCM is based on an accumulation of aberrancies rather than a single aberrancy which might reflect the degree of deviation from normal. The number of aberrancies in the myelomonocytic and blast compartment can be translated by a flow cytometric scoring system into a flow score (Wells et al., Blood, 2003,102:394–403, van de Loosdrecht et al., Blood, 2008,111:1067–77). A flow score above 3 is suggestive for MDS. The clinical impact of CD38 expression as a single flow cytometric parameter or as part of a multi parameter analysis requires validation with emphasis on inconclusive cases such as idiopathic cytopenia of unknown significance and idiopathic dysplasia of unknown significance. Flow cytometry in MDS is highly promising and prospective studies are warranted. 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: 2009
    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 ...
  • 2
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 3798-3798
    Abstract: Abstract 3798 Poster Board III-734 Myelodysplastic syndromes (MDS) can be classified into low risk and high risk categories, with evolution to acute myeloid leukemia (AML) predominantly in the latter cases. In AML, survival of leukemia-initiating cells, often referred to as leukemic stem cells, after chemotherapy is thought to result in minimal residual disease leading to relapse. A larger size of the stem cell compartment in de novo AML is predictive for poor survival [Van Rhenen et al.,Clin Cancer Res 2005,11]. A monoclonal antibody against the cell surface antigen C-type lectin-like molecule-1, CLL-1, together with lineage infidelity markers enables discrimination of normal and malignant stem cells in AML [Van Rhenen et al., Blood 2007, 110; Van Rhenen et al., Leukemia 2007, 21] . Previously, we showed that the size of the total stem cell compartment including both normal and malignant cells, defined as CD45dimCD34+CD38−, were significantly more prevalent within the blast cell fraction in high risk MDS as compared to low risk MDS (median 0.77% (n=15) and 0.25% (n=73), respectively, p=0.040) [Westers et al. Blood 2008, 112, abstract 1661]. This might reflect the differences in clinical course in these patients. It could be hypothesized that aberrant marker expression on MDS stem cells may predict leukemic evolution. Therefore, stem cells in MDS bone marrow samples were analyzed by flow cytometry for expression of CLL-1 and aberrant lineage markers. A reliable number of stem cells ( 〉 20) could be examined in 22 low risk and 14 high risk MDS patients; patients were classified by WHO2001 as refractory anemia w/o ring sideroblasts (RS; n=6) or refractory cytopenia with multilineage dysplasia w/o RS (n=16) and 14 refractory anemia with excess of blasts type 1 or 2 (n=14). Median CLL-1 expression on the CD34+CD38− stem cells was 0.0% (range 0-50, 3 out of 22 clearly ( 〉 10%) positive cases) in low risk and 2.2% (range 0-27, 3 out of 14 clearly positive cases) in high risk MDS. For comparison, median CLL-1 expression on stem cells in normal controls was 0.0% (range 0-4.7, not significantly different from MDS cases: p=0.55 and 0.20 as compared to low and high risk categories, respectively). Eleven of the low risk MDS cases were tested for aberrant lineage infidelity marker expression on their myeloid blasts and stem cells. In these cases, myeloid blasts expressed either CD5 or CD7 (n=2 and n=7, respectively; median 33% of blast cells). Median expression of these antigens on stem cells was only 1.2% (range 0-47). There was only one case with high expression (47%); the rest was below 7%. Only four of the high risk cases could be analyzed for lineage infidelity marker expression. In these patients, median 51% of the myeloid blasts expressed either CD5 (n=1), CD7 (n=1) or CD56 (n=2). Median expression of these antigens on stem cells in these high risk MDS cases was 44% (range 3-83); three of four cases had high expression ( 〉 40%). Overall, expression of CLL-1 and lineage infidelity markers was higher in high risk MDS as compared to the low risk category (p=0.047 and p=0.036 for CLL-1 and lineage infidelity markers, respectively). Of note, the only low risk case with a high percentage (47%) of aberrant stem cells as assessed by flow cytometry rapidly progressed towards AML. To conclude, CLL-1 is virtually absent on stem cells in low and high risk MDS. Nevertheless, expression of lineage infidelity markers, such as CD5, CD7 and CD56, on CD34+CD38− stem cells in some cases strongly suggests that a considerable part of these stem cells was malignant. Aberrant stem cell phenotype as assessed by flow cytometry might discriminate normal from MDS stem cells. This enables further analysis of MDS stem cells and, moreover, makes them amenable for application of targeted therapy. Future analysis might reveal whether the existence of MDS stem cells with an aberrant profile by flow cytometry predicts leukemic evolution. 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: 2009
    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 ...
  • 3
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 1338-1338
    Abstract: Myelodysplastic syndromes (MDS) represent a heterogeneous group of clonal hematopoietic disorders, characterized by ineffective hematopoiesis resulting in cytopenias and a highly variable clinical course. Cytogenetics are routinely used as one of the diagnostic, prognostic and therapeutic markers in the clinical management of MDS (Greenberg et al. Blood 120:2454,2012). Although karyotyping is generally considered as the gold standard in the cytogenetic characterization of MDS, 40-60% of the patients exhibit a normal karyotype. Intrinsically, the resolution of karyotyping is limited by its capacity to detect only those copy number changes that are microscopically visible (5-10 Mb in size). In contrast, microarray-based genomic profiling analyses allow a genome-wide detection of copy number alterations (CNAs), down to 100 kb in size, and regions of copy neutral loss of heterozygosity (CNLOH). Such analyses also overcome some of the other major limitations of karyotyping such as low success rate due to inadequate metaphase yield and/or poor banding quality. We have compared karyotyping and fluorescence in situ hybridization (FISH) with microarray-based genomic profiling with respect to the detection yield for genetic abnormalities in bone marrow samples from lower risk MDS patients. We used the HOVON89 study-cohort, a prospective phase II randomized multicenter study to assess the efficacy of lenalidomide with or without erythropoietin and granulocyte-colony stimulating factor in patients with low/intermediate-1 risk MDS; www.trialregister.nl; NTR1825; EudraCT nr.: 2008-002195-10. Inclusion target of the study is 200 low/intermediate-1 risk MDS patients (134 enrolled, inclusion ongoing). Data regarding cytogenetics, FISH and microarray were obtained in a fully blinded fashion for 68 MDS patients. For microarray-based genomic profiling we used the recently launched high resolution CytoScan HD Array (Affymetrix) platform. The following interpretation criteria were applied: (i) the threshold for CNAs was set at 〉 5 Mb, (ii) inclusion of 〈 5 Mb CNAs segments that coincide with known cancer genes as reported on www.sanger.ac.uk and (iii) the threshold for CNLOH was set at 〉 10 Mb and to telomere. Karyotyping and interphase FISH were performed using standard cytogenetic methods. In all 4 patients where karyotyping revealed no metaphases interphase FISH was performed. Thirty-six of the 68 (53%) MDS patients had an abnormal microarray profile. Of interest, in 13 of these 36 patients no abnormalities were observed by karyotyping and/or FISH. All these abnormalities observed by microarray only, involved focal 〈 5 Mb CNAs (containing e.g. the TET2, RUNX1 and DNMT3A genes) and regions of CNLOH (coinciding with 1p, 2p, 4q, 7q, 11p, 11q, 12q, 14q and 18q), which are all out of the scope of karyotyping and FISH. All CNAs identified by karyotyping and FISH were also observed by microarray-based genomic profiling, including a case with loss of 5q in 5% of the cells and loss of 7q in 9% of the cells as observed by interphase FISH and another case with loss of 5q in 3 of 20 the analyzed metaphases by karyotyping. These observations demonstrate the high sensitivity of the CytoScan HD array platform for the identification of CNAs in (small) subclones. As expected balanced translocations such as t(3;3)(q21;q26) and t(2;14)(q37;q22) present in 2 of the patients in our cohort were not identified by microarray-based genomic profiling. This study will be extended to all patients to be included in the HOVON89 trial. In conclusion, we demonstrate that in the present cohort of 68 patients, microarray-based genomic profiling allows the identification of almost all copy number abnormalities also observed by karyotyping and FISH. In addition, we show that microarray-based genomic profiling allows the detection of potential prognostic relevant abnormalities (focal CNAs and CNLOH) which would have remained undetected by karyotyping and FISH. The predictive and/or prognostic value of these novel CNAs and CNLOH will be evaluated within the ongoing prospective clinical HOVON89 trial in lower risk MDS. 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 ...
  • 4
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 1521-1521
    Abstract: Myelodysplastic syndromes (MDS) constitute a heterogeneous group of hematopoietic stem cell disorders, characterized by ineffective hematopoiesis resulting in cytopenias and variable risk of acute myeloid leukemia (AML). To make an accurate distinction between specific risk categories in MDS, especially in low and intermediate risk MDS, a multi-diagnostic approach is recommended. To verify the efficacy of multiple diagnostic tools in MDS we used the HOVON89 study-cohort (a prospective phase II randomized multicenter study to assess the efficacy of lenalidomide with or without erythropoietin and granulocyte-colony stimulating factor in patients with low-intermediate-1 risk MDS; trial registered at www.trialregister.nl as NTR1825; EudraCT nr.: 2008-002195-10. Inclusion target of the study is 200 low/intermediate-1 risk MDS patients (134 enrolled, inclusion ongoing). We collect data on cytomorphology (CM), conventional cytogenetics (CCG), fluorescence in situ hybridization (FISH) and microarray-based genomic profiling. In addition, we performed flow cytometric (FC) analysis according to European LeukemiaNet guidelines (Van de Loosdrecht et al., Haematologica 2009 and Leukemia 2012). Current CM results (N=98) identified: 8 refractory anemia (RA); 16 refractory anemia with ringed sideroblasts (RARS); 43 refractory cytopenia with multilineage dysplasia with/without ringed sideroblasts (RCMD/RCMD-RS); 16 refractory anemia with excess blast-1 (RAEB-1), 5 chronic myelomonocytic leukemia-1 (CMML-1) and 10 patients with isolated del(5q). CCG analysis (N=101) indicated 2 very good risk, 84 good risk, 13 intermediate risk and 2 poor risk patients according to the IPSS-R risk categories (Greenberg et al., Blood 2012). In addition, interphase FISH analysis (N=72) was normal in 15 patients, in 6 patients the del(5q) was confirmed. From 68 MDS patients data from both CCG and microarray were available. Microarray-based genomic profiling identified genomic abnormalities such as copy neutral loss of heterozygosity and small ( 〈 5 Mb) copy number alterations coinciding with a cancer gene in 13 patients with normal CCG. As expected in one patient the balanced translocation t(3;3)(q21;q26) was not identified by microarray-based genomic profiling. FC analysis (N=82) evaluated aberrancies with regard to count, marker expression level and lineage infidelity marker expression on myeloid progenitors, B cell progenitors, maturing myeloid/monocytic and erythroid cells. Current, validated FC-scoring systems identified 60/81 (Della Porta et al., Haematologica 2012) and 61/81 (Wells et al., Blood 2003) patients with MDS. Both scoring systems do not evaluate dyserytropoiesis and dysmegakaropoiesis, thereby possibly not recognizing RA, RARS or RCMD patients with only dyserytropoiesis and dysmegakaropoiesis. We integrated both scoring systems into one score (Van de Loosdrecht et al., Leukemia 2012, and JNCCN 2013). Patients were divided into 3 categories: not likely MDS (A), signs of dysmyelopoiesis (B) and fitting MDS (C). This score diagnosed 69/78 patients as probably or likely MDS by FC (B or C, figure 1). Remarkably, patients scored as ‘category A’ only displayed dyserytropoiesis and/or dysmegakaryopoiesis by CM. FC identified dyserytropoiesis in these patients, however, a consensus erytroid flow score is not yet validated. In addition, FC identified different risk categories within the patient group with no genetic abnormalities (based on CCG, FISH and microarray-based genomic profiling; data not shown). In conclusion, this is the first prospective study in low/int-1 risk MDS that validates FC as a valuable diagnostic tool in MDS (sensitivity of FC in this cohort: 88%). FC only failed to recognize some patients with only dyserytropoiesis and dysmegakaryopoiesis by CM, not evaluated by the current scoring system. Thirteen patients with unilineage dysplasia by CM had multilineage dysplasia by FC. We postulate that RA/RARS patients with multi-lineage dysplasia by FC may have clinical features of RCMD patients and therefore a higher risk on transformation to AML. Clinical follow-up data are expected within 1.5 year. In near future, a multi-diagnostic approach may i) identify risk categories within well defined IPSS-R subgroups, ii) predict risk on transformation and iii) select patients who might benefit from new emerging drugs for low-int-1 risk MDS. 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 ...
  • 5
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 2697-2697
    Abstract: It is generally accepted that myelodysplastic syndromes (MDS) most often originate in a multipotent, myelorestricted progenitor population, although primary transformation may occur at the hematopoietic stem cell level. MDS can be classified into low risk and high risk with evolution to acute myeloid leukemia (AML) predominantly in the latter cases. In AML, survival of leukemia-initiating cells, often referred to as leukemic stem cells, after chemotherapy is thought to lead to minimal residual disease and relapse. Hence, in de novo AML a larger size of the stem cell compartment is predictive for poor survival. [Van Rhenen et al.,Clin Cancer Res 2005,11] The monoclonal antibody against the cell surface antigen C-type lectin-like molecule-1, CLL-1, together with lineage infidelity markers enables discrimination of normal and malignant stem cells. [Van Rhenen et al.,Blood 2007,110; Van Rhenen et al.,Leukemia 2007,21] It could be hypothesized that CLL-1 and aberrant marker expression on MDS stem cells together with size of the stem cell compartment may predict leukemic evolution. Therefore, stem cells, defined as CD45dimCD34+CD38−, were analyzed for expression of CLL-1 and aberrant lineage markers in bone marrow samples from 88 MDS patients classified by WHO as 16 RA w/o RS, 42 RCMD w/o RS, 3 MDS-U, 5 hypoplastic MDS, 6 MDS/MPD and CMML, 15 RAEB-1 and 2, 20 AML patients with a known MDS history and 26 healthy controls. Analysis of the CD34+CD38− frequency in all MDS patients and normal controls revealed no significant differences (median 0.0061% vs. 0.0074%, respectively), whereas the frequency of CD34+CD38− cells was 17-fold higher in high risk MDS (RAEB-1 and 2, median: 0.076%) as compared to low risk MDS (median: 0.0046%, p & lt;0.001). Similar as in AML, stem cells were significantly more prevalent within the blast cell fraction (CD45dimSSCint/low) of high risk MDS as compared to low risk MDS (median 0.77% and 0.25%, respectively), reflecting the differences in clinical course in these patients (p=0.040). Regarding CLL-1 expression, a reliable number of stem cells ( & gt;20) could be tested in 11/15 high risk RAEB-1 and 2 cases and in 16/73 of the remaining low risk MDS cases. In these cases, median CLL-1 expression on the CD34+CD38− cells was 1.6% (range 0–50) in low risk and 2.0% (range 0–27) in high risk MDS. Median CLL-1 expression on stem cells was 0.0% (range 0–4.7) in normal controls. Nevertheless, expression of lineage infidelity markers, such as CD5, CD7 and CD56, on CD34+CD38− stem cells in MDS strongly suggests that a considerable part of these stem cells is malignant (median 35% in 7/16 patients tested). Our data show that CLL-1 is virtually absent on stem cells in MDS. Remarkably, median CLL-1 expression on stem cells in AML cases that evolved from MDS (7%, range 0–53, n=9) was manifold lower than in de novo AML (median 45% when excluding non de novo AML [Van Rhenen et al.,Blood 2007,110], p=0.034). Detailed analysis of CLL-1 expression in AML had already revealed that CLL-1 expression increases with differentiation (CD34− & gt; CD34+CD38− & gt; CD34+CD38+). [Bakker et al.,Cancer Res 2004,64;Van Rhenen et al.,Blood 2007,110] Thus, our data suggest that the CD34+CD38− cells in high risk MDS and AML with antecedent MDS are more immature than in most de novo AML, which might explain poor prognosis of AML cases with MDS history. To conclude, our data indicate that CLL-1 is a specific marker of de novo AML, while CLL-1-negative AML may have been evolved from a MDS pre-phase that is further characterized by an increasing size of the stem cell compartment upon progression towards AML.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    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
    Online Resource
    Online Resource
    American Society of Hematology ; 2010
    In:  Blood Vol. 116, No. 21 ( 2010-11-19), p. 1860-1860
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 1860-1860
    Abstract: Abstract 1860 Immune editing is recognized as being important in the pathogenesis of myelodysplastic syndrome (MDS). In low risk MDS, inappropriate immune reactions are thought to contribute to the initiation and progression of the disease. IL-17 producing CD4+ T-helper cells have been associated with various autoimmune diseases and are found in increased frequencies in low-risk MDS. These increased Th17 frequencies are accompanied by a reduced FoxP3+ T-regulatory (Treg) frequency. On the other hand, in high-risk MDS, the elevated number of bone marrow blasts was associated with increased numbers of Treg. These increased Treg may inhibit immune responses directed against the dysplastic blasts and as such permit progression of the disease. The demethylating agents ‘5-aza-2’-deoxycytidine and ‘5-azacitidine (Aza) have shown significant clinical benefits in the treatment of MDS. Although aberrant DNA methylation patterns appear to play a role in the pathogenesis of MDS, clinical responses to DNA demethylating agents could not be related to changes in DNA methylation patterns. Cytokine production and FoxP3 expression are also regulated by epigenetic mechanisms, and FoxP3 expression is increased upon in vitro treatment with Aza. Since cytokines and FoxP3+ Treg may play a role in the pathogenesis of MDS, we set out to investigate the effects of Aza (Vidaza®) treatment on CD4+ T-helper subset frequencies in high-risk MDS patients. Nine patients treated with 75 mg/m2/day subcutaneously for 7 consecutive days in a 28 day cycle were included in this study. Peripheral blood samples were drawn before, 15 days after start of treatment and at the end of the third cycle. There was no change in absolute or relative CD3+ and CD3+CD4+ T-cell numbers during treatment. The proportion of Treg was slightly but not significantly increased (paired student's T-test, p=0.08) on day 15 (11.9 % of CD4+) compared to levels before treatment (9.6 % of CD4+) while numbers dropped to pretreatment levels after the third cycle(9.6 % of CD4+). Cytokine producing cells were enumerated after stimulation with phorbol-12-myristate-13-acetate (PMA) and ionomycin in the presence of Brefeldin A. No significant changes in IFNγ (23.8%, 22.5% and 17.9% of CD4+ cells pretreatment, 15d post treatment and after the third cycle respectively) TNFα (38.3, 35.9 and 39.3% of CD4+ cells) or IL-4 (0.6, 1.0 and 0.6% of CD4+ cells) producing cells were observed. However, the proportion of IL-17 producing cells progressively decreased during treatment (1.1, 0.7 and 0.6% of CD4+ cells; ANOVA, p=0.006). In conclusion, Aza may modulate the immune response and in particular reduce IL-17 responses. This effect of Aza may provide a rationale for expanding the group of patients eligible for Aza treatment with low risk MDS patients, because low risk MDS has been associated with elevated, potential pathogenic, Th17 cells. Disclosures: Ossenkoppele: Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding. Van de Loosdrecht:Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding.
    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 ...
  • 7
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 3841-3841
    Abstract: Abstract 3841 The efficacy of azacitidine in the treatment of high risk myelodysplastic syndromes (MDS), chronic myelomonocytic leukaemia (CMML) and acute myeloid leukaemia (AML) (20–30% blasts) has been demonstrated. To investigate the efficacy of azacitidine in daily clinical practice and to identify predictors for response, we analyzed a cohort of 90 MDS, CMML and AML patients who have been treated in a Dutch compassionate patient named program. Patients received azacitidine for a median of 5 cycles (range 1–19). The overall response rate (CR/PR/HI) was 57% in low risk MDS, 53% in high risk MDS, 50% in CMML, and 39% in AML patients. Median overall survival (OS) was 13.0 (9.8–16.2) months. In multivariate analysis we confirmed that circulating blasts (HR 0.48, 95% CI 0.24–0.99; p=.05) and poor risk cytogenetics (HR 0.45, 95% CI 0.22–0.91; p=.03) are independent predictors for OS. Interestingly, in this analysis we also identified platelet doubling after the first cycle of azacitidine as a simple and independent positive predictor for OS (HR 5.4, 95% CI 0.73–39.9; p=.10). Of the 90 treated patients, 14 (16%) had an at least two-fold increase in platelet count after the first cycle of azacitidine, which was associated with significant better OS (p=.01, according to logrank test) (figure). Of these 14 patients 13 could be classified according the azacitidine prognostic scoring system for OS as recently proposed by Itzykson et al. (Blood:2011;117:403); 6 patients belonged to the low risk and 7 to the intermediate risk group. Median baseline platelet count of these patients was 35 x109/L (range 2–290 x109/L). Characteristics of this subgroup of patients were not significantly different from the patients without platelet doubling. Interestingly, platelet doubling was observed in all cytogenetic risk groups, in patients with and without circulating blasts, and in patients who are transfusion dependent and independent. In conclusion, routine administration of azacitidine to patients with variable risk groups of MDS, CMML and AML is feasible and subgroups with distinct efficacy of azacitidine treatment can be identified. Disclosures: Wijermans: Centocor Ortho Biotech Research & Development: Research Funding.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    S. Karger AG ; 2014
    In:  Neuropsychobiology Vol. 69, No. 3 ( 2014), p. 159-164
    In: Neuropsychobiology, S. Karger AG, Vol. 69, No. 3 ( 2014), p. 159-164
    Abstract: 〈 b 〉 〈 i 〉 Aims: 〈 /i 〉 〈 /b 〉 We aimed to investigate plasma omentin concentrations in non-obese, drug-free patients with schizophrenia in comparison with healthy volunteers. 〈 b 〉 〈 i 〉 Method: 〈 /i 〉 〈 /b 〉 Thirty-two patients with schizophrenia and 33 control subjects were recruited. Plasma omentin levels were determined by enzyme-linked immunosorbent assay. 〈 b 〉 〈 i 〉 Results: 〈 /i 〉 〈 /b 〉 Plasma levels of omentin (ng/ml) were found to be markedly lower in patients with schizophrenia (median = 7.7, 25th percentile = 6.3, 75th percentile = 604.9) than in controls (median = 486, 25th percentile = 326, 75th percentile = 794.2, p 〈 0.01). No significant difference was found between drug-free (n = 23) and drug-naive (n = 9) patients with respect to plasma omentin levels. Omentin concentrations correlated negatively with severity of illness, suggesting that patients with more severe pathology had lower fasting levels of omentin (n = 32; r = -0.387; p = 0.029). 〈 b 〉 〈 i 〉 Conclusion: 〈 /i 〉 〈 /b 〉 The present results suggest that plasma omentin levels are decreased in physically healthy, non-obese, antipsychotic-free patients with schizophrenia when compared with physically and mentally healthy individuals. To our knowledge, this is the first study that demonstrated the association between omentin and schizophrenia.
    Type of Medium: Online Resource
    ISSN: 0302-282X , 1423-0224
    Language: English
    Publisher: S. Karger AG
    Publication Date: 2014
    detail.hit.zdb_id: 1483094-2
    SSG: 5,2
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    Elsevier BV ; 2009
    In:  Leukemia Research Vol. 33, No. 2 ( 2009-2), p. 209-211
    In: Leukemia Research, Elsevier BV, Vol. 33, No. 2 ( 2009-2), p. 209-211
    Type of Medium: Online Resource
    ISSN: 0145-2126
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2009
    detail.hit.zdb_id: 2008028-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Annals of Hematology, Springer Science and Business Media LLC, Vol. 100, No. 7 ( 2021-07), p. 1711-1722
    Type of Medium: Online Resource
    ISSN: 0939-5555 , 1432-0584
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2021
    detail.hit.zdb_id: 1458429-3
    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