Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 31, No. 21 ( 2013-07-20), p. 2662-2670
    Abstract: Myelodysplastic syndromes (MDS) are clonal hematopoietic disorders that are more common in patients aged ≥ 60 years and are incurable with conventional therapies. Reduced-intensity conditioning (RIC) allogeneic hematopoietic stem-cell transplantation is potentially curative but has additional mortality risk. We evaluated RIC transplantation versus nontransplantation therapies in older patients with MDS stratified by International Prognostic Scoring System (IPSS) risk. Patients and Methods A Markov decision model with quality-of-life utility estimates for different MDS and transplantation states was assessed. Outcomes were life expectancy (LE) and quality-adjusted life expectancy (QALE). A total of 514 patients with de novo MDS aged 60 to 70 years were evaluated. Chronic myelomonocytic leukemia, isolated 5q– syndrome, unclassifiable, and therapy-related MDS were excluded. Transplantation using T-cell depletion or HLA-mismatched or umbilical cord donors was also excluded. RIC transplantation (n = 132) stratified by IPSS risk was compared with best supportive care for patients with nonanemic low/intermediate-1 IPSS (n = 123), hematopoietic growth factors for patients with anemic low/intermediate-1 IPSS (n = 94), and hypomethylating agents for patients with intermediate-2/high IPSS (n = 165). Results For patients with low/intermediate-1 IPSS MDS, RIC transplantation LE was 38 months versus 77 months with nontransplantation approaches. QALE and sensitivity analysis did not favor RIC transplantation across plausible utility estimates. For intermediate-2/high IPSS MDS, RIC transplantation LE was 36 months versus 28 months for nontransplantation therapies. QALE and sensitivity analysis favored RIC transplantation across plausible utility estimates. Conclusion For patients with de novo MDS aged 60 to 70 years, favored treatments vary with IPSS risk. For low/intermediate-1 IPSS, nontransplantation approaches are preferred. For intermediate-2/high IPSS, RIC transplantation offers overall and quality-adjusted survival benefit.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2013
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 115-115
    Abstract: Abstract 115 Reduced-intensity conditioning (RIC) allogeneic hematopoietic stem cell transplantation is a curative approach for older patients with myelodysplastic syndrome (MDS), but is associated with early mortality risk. We previously used decision modeling to define the role of myeloablative transplantation in younger MDS patients aged 〈 60 years, stratified by IPSS risk (Cutler et al, Blood 2004). We thereafter undertook a similar analysis to define the role of RIC transplantation in older MDS patients. We constructed a Markov decision model in de-novo MDS patients aged 60–70 years to evaluate life expectancy after a strategy of early T-replete RIC transplantation versus standard-of-care non-transplantation approaches. Patients with chronic myelomonocytic leukemia, isolated 5q-, unclassifiable and therapy-related MDS were excluded. Patients undergoing transplantation with anti-thymocyte globulin or alemtuzumab-based conditioning or HLA mismatched or umbilical cord donors were also excluded. We undertook adjustments for quality-of-life (QoL) using standard utility estimates for different MDS and transplantation states (0.84: RBC transfusion-independent low/intermediate-1 MDS; 0.6: RBC transfusion-dependent low/intermediate-1 MDS; 0.53: intermediate-2/high MDS; 0.92: post transplantation-overall; 0.6: post transplantation-GVHD) We evaluated survival of 92 patients after HLA matched RIC transplantation (Dana-Farber, Fred Hutchinson and CIBMTR datasets) stratified by IPSS risk state, versus survival with: best supportive care for 183 non-anemic low/intermediate-1 IPSS patients (IMRAW and Pavia datasets); hematopoietic growth factors for 78 anemic low/intermediate-1 IPSS patients (Nordic MDS Group and GFM datasets); and hypomethylating agents for 160 intermediate-2/high IPSS risk patients (Celgene AZA-001, GFM compassionate use azacytidine and M. D. Anderson decitabine datasets). For older patients with low/intermediate-1 IPSS risk, early RIC transplantation impaired life expectancy compared to non-transplantation approaches (Table). Excluding patients transplanted beyond 12 months from diagnosis did not alter the conclusion. QoL adjustment narrowed the gap in quality adjusted life expectancy (QALE), but sensitivity analysis did not support RIC transplantation as the preferred strategy across a broad range of possible utility estimates (0.5–1), indicating that MDS-associated morbidity (e.g. RBC transfusion-dependence) does not favor early transplantation unless its QoL impact is substantial (state utility 〈 0.4). For older patients with intermediate-2/high IPSS risk, early RIC transplantation improved life expectancy compared with hypomethylating agents (Table). Excluding patients transplanted beyond 12 months from diagnosis did not alter the conclusion. However, transplantation is associated with early mortality such that the life expectancy benefit was apparent only after modeling survival beyond 5 years. Importantly, QoL adjusted survival benefit of transplantation was apparent both at 5 years and beyond, highlighting the morbidity of higher-risk MDS. In sensitivity analyses, QoL adjustment across the range of plausible transplantation-associated utility estimates (0.5–1) did not change the conclusion of QALE benefit. We conclude that for de-novo MDS patients aged 60–70 years with low/intermediate-1 disease, early transplantation is not the preferred strategy unless MDS-associated QoL impairment is substantial. For intermediate-2/high IPSS risk, early RIC transplantation offers a life expectancy benefit, with quality adjusted survival benefit detectable earlier.Table.Early RIC transplantationNo early RIC transplantationLow/intermediate-1 IPSSOverall Life Expectancy (months)3877QALE: Transfusion-independent MDS morbidity (months)3565QALE: Transfusion-dependent MDS morbidity (months)3546Intermediate-2/high IPSSOverall Life Expectancy (months)3628QALE: Higher-risk MDS morbidity (months)3315QALE: GVHD morbidity (months)2215 Disclosures: Fenaux: Celgene: Honoraria, Research Funding. Hellstrom-Lindberg:Celgene: Consultancy, Research Funding. Gale:celgene: Employment; unitedhealthcare: Consultancy; oxford health plans: Consultancy. Beach:celgene: Employment. Greenberg:novartis: Consultancy, Research Funding; glaxosmithkline: Research Funding; onconova: Research Funding; amgen: Consultancy, 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 ...
  • 3
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 4613-4613
    Abstract: Introduction: Azacytidine (Aza) is first-line treatment for patients with higher-risk MDS but only around 50% of patients respond to therapy. Overall survival for this patient group is short and clinical decision-making tools are highly warranted. As Aza may improve survival also in patients with hematologic improvement or stable disease, survival may be a better response predictor than response rate. Methods: We evaluated the impact of clinical parameters (n=134), mutations (n=90) and DNA methylation profiles (n=42) on response and survival in a cohort of consecutive patients with higher-risk MDS treated with Aza. Targeted sequencing of 42 genes involved in myeloid disease and Illumina 450 methylation arrays were applied for mutational assessment and methylation profiling, respectively. The IWG criteria were used for response scoring. Results: Patients were eligible for analysis if they had received ≥1 dose of Aza. Median number of cycles given was 6 (range 1-29). Responses were scored as CR (22%), mCR (11%), PR (3%), HI (13%), SD (27%) and PD (13%). Fifteen patients (11%) were not evaluated for response due to early death. Disease duration was negatively associated with both response (p=0.035) and survival (p=0.001). Adverse cytogenetics and high absolute neutrophil count was associated with shorter survival (p=0.03 and p=0.02) but not with response. No single mutation or group of mutations was associated with response although there was a weak positive trend for TET2 and ASXL1. When using survival as endpoint, ASXL1 showed a strong trend towards prolonged survival (median 29 vs 14 months, p=0.07) and, importantly, the group of patients with any mutation in histone modulators (ASXL1, EZH2, MLL) had a significant longer survival (median 28 vs 13 months, p=0.01). This remained significant in the cox regression model (HR 0.3223 (0.16-0.70 95% CI); p=0.002). No other mutations or group of mutations were associated with survival. Interestingly, previously reported negative prognostic factors including RUNX1 (p=0.82), TP53 (p=0.54), and the number of mutations (p=0.37), were not associated with survival in this Aza-treated cohort DNA methylation profiling identified 233 differentially methylated regions (DMRs) between responders and non-responders, corresponding to 200 genes, including six HOX-genes, which were highly enriched for gene ontology pathways involved in development and differentiation. High methylation of HOXA5, the most significant DMR, was associated with prolonged survival (22 vs 12 months, p=0.03). We also studied the methylation level of HOXA5 in CD34+ cells from patients with high-risk MDS and sorted compartments during myeloid differentiation in normal bone marrow. The methylation profile in responding patients was closer to that of differentiated cells while non-responding cells were closer to progenitor cells. Discussion: Single mutations have a limited impact on response rates. Howver, we demonstrate a clear survival benefit for patients with mutations in histone modulators, which previously have been reported as negative prognostic factors (Bejar, NEJM 2013; Haferlach, Leukemia 2014). Moreover, several negative risk factors, such as RUNX1, TP53, and the number of mutations were neutralized by Aza. Histone modulation mutations may therefore be used in the clinical decision-making for higher-risk MDS. We demonstrate for the first time that methylation profiles in genes involved in differentiation and development differ between responders and non-responders and that hypermethylation of HOXA5 is positively associated with survival (p=0.03). Since methylation pattern in HOXA5 is linked to differentiation status, we hypothesize that non-responding patients are skewed towards more immature differentiation. Figure 1: Survival curves Figure 1:. Survival curves Figure 2: DNA methylation levels at the HOXA5 locus. Squares represent gene location with light green=TSS-1500; Dark green=TSS-200; Red=Gene body; Magenta=1st Exon; Dark blue=5’UTR; Cyan=3’UTR and diamonds represent sample values. A=Median methylation level of responders illustrated with orange diamonds (MNCs) and non-responders with blue diamonds (MNCs). B=Added CD34+ cells with red diamonds. C=All patients. D=Normal bone marrow with PMN illustrated with brown diamonds and CMP with green diamonds. Figure 2:. DNA methylation levels at the HOXA5 locus. Squares represent gene location with light green=TSS-1500; Dark green=TSS-200; Red=Gene body; Magenta=1st Exon; Dark blue=5’UTR; Cyan=3’UTR and diamonds represent sample values. A=Median methylation level of responders illustrated with orange diamonds (MNCs) and non-responders with blue diamonds (MNCs). B=Added CD34+ cells with red diamonds. C=All patients. D=Normal bone marrow with PMN illustrated with brown diamonds and CMP with green diamonds. Figure 3 Figure 3. 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: 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 ...
  • 4
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 3857-3857
    Abstract: Abstract 3857 The diagnosis of patients with myelodysplastic syndromes (MDS) is largely dependent on morphologic examination of bone marrow aspirates. Several criteria that form the basis of the classifications and scoring systems most commonly used in clinical practice are affected by operator-dependent variation. In order to identify more standardized molecular markers that would allow a more reliable prediction of prognosis, we have used gene expression profiling (GEP) data on CD34+ cells from MDS patients to determine the relationship between gene expression levels and prognosis in this disorder. GEP data on CD34+ cells from 125 MDS patients with a minimum 12-month follow-up since date of bone marrow sample collection were included in this study. Prediction for overall survival was performed using supervised principal components (“SuperPC”) and lasso penalized Cox proportional hazards regression applying the “Coxnet” algorithm. Supervised principal components analysis was performed on patients randomly split in a training set (n=84) and a test set (n=41), and 139 genes were identified the expression of which was significantly associated with MDS patient survival, including LEF1, CDH1, WT1 and MN1. In order to identify a smaller set of genes associated with patient survival, a second approach aiming at building sparse prediction models was used. A model was generated using the Coxnet algorithm and a predictor consisting of 20 genes was identified. Eight genes identified by the supervised principal components method were in common with the genes identified by the Coxnet model: ADHFE1, BTBD6, CPT1B, LEF1, FRMD6, GPR114, C7orf58 and LOC100286844. The Coxnet predictor outperformed other predictors including one which additionally used clinical information. To validate our findings, we evaluated the performance of our prognostic Coxnet gene signature in an independent gene expression profiling dataset on MDS bone marrow mononuclear cells (Mills et al, Gene Expression Omnibus series GSE15061). Our Coxnet gene signature based on CD34+ cells significantly identified a low-risk patient group in this independent GEP dataset based on unsorted bone marrow mononuclear cells, demonstrating that our signature is robust and may be applicable to bone marrow cells without the need to isolate CD34+ cells. These GEP-based signatures correlating with clinical outcome may significantly contribute to a refined risk classification of 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: 2012
    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. 116, No. 21 ( 2010-11-19), p. 298-298
    Abstract: Abstract 298 The myelodysplastic syndromes (MDS) are a heterogeneous group of clonal hematopoietic stem cell malignancies that are characterized by ineffective hematopoiesis resulting in peripheral cytopenias and a hypercellular bone marrow. Approximately 40% of patients with MDS will develop an acute myeloid leukemia. It is important to establish prognosis of MDS patients since the treatment options vary from supportive care to bone marrow transplantation. In order to determine the relationship of gene expression levels to prognosis and so identify new molecular markers, we have used gene expression profiling to study the transcriptome of the hematopoietic stem cells of 125 MDS patients with a minimum 12 month follow up. The CD34+ cells obtained from MDS patients and healthy individuals were analyzed using Affymetrix U133 Plus2.0 arrays. The patients were split randomly in a training set (n=84) and a test set (n=41). Supervised principal components analysis was used to identify genes correlated with survival. Using the 84 patients in the training set, the Cox scores were computed for each gene, and the principal components calculated on the genes with the highest Cox scores. The first of the principal components was then used to generate a regression model to predict the survival in the test set. Finally, for each probe set an importance score was calculated equal to its correlation with the supervised principal component predictor. This approach returned a list of 150 top ranked probe sets correlated with survival. Patients in the training set were split into tertiles based on the predictor (low, medium and high score) and patients in the test set were assigned to their predicted class, and Kaplan-Meier plots were generated for both training and test set. The differences in survival for both training and test set were statistically significant (Figure 1). Top ranked genes showing lower expression levels in patients with shorter survival include CDH1, LEF1 and AKAP12/Gravin. Top ranked genes showing higher expression levels in patients with shorter survival include IL23A, WT1 and PTHR2. Figure 2 shows survival of patients divided into tertiles of expression for the individual genes CDH1, LEF1 and WT1. It is probable that the genes identified in this study will become the first validated molecular markers for MDS prognosis. Multivariate analysis is currently being performed. Figure 1 Figure 1. Figure 2 Figure 2. Disclosure: 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: 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 ...
  • 6
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 2626-2626
    Abstract: The myelodysplastic syndromes (MDS) are a heterogeneous group of hematopoietic malignancies. We have used Affymetrix microarray technology to determine the gene expression profiles in CD34+ cells of 84 MDS patients (25 RA, 28 RARS and 31 RAEB) and 16 healthy controls. Twenty-five of 84 patients had a del(5q). CD34+ cells were isolated from bone marrow samples using MACS magnetic columns. Extracted total RNA was amplified using the Two-Cycle Target Labelling kit (Affymetrix) and samples were hybridized to Affymetrix U133 Plus2.0 chips (representing 39,000 human genes). Cell intensity calculation and scaling was performed using GeneChip Operating Software and data analysis using GeneSpring 7.3. The expression profiles of MDS CD34+ cells showed many similarities to reported interferon-γ induced gene expression in normal CD34+ cells. Indeed the two most up-regulated genes, IFIT1 and IFITM1, are interferon-stimulated genes. IFIT1 and IFITM1 were up-regulated by 〉 2-fold in 58/84 and 53/84 MDS patients respectively. Genes down-regulated by 〉 2-fold in the majority of MDS patients include the putative tumor suppressor gene Gravin/AKAP12, ARPP-21, CD24 and MME. The association of distinct gene expression profiles with specific FAB and cytogenetic groups was determined using data from 55 MDS patients as a training set. Hierarchical clustering performed using 457 significantly different genes between different FAB subtypes showed that MDS patients with RARS constitute a homogeneous group, while MDS patients with RA and RAEB show more overlap. CD34+ cells from patients with RARS showed up-regulation of mitochondrial-related genes, and in particular of those of heme synthesis (e.g. ALAS2). Statistical analysis showed that 889 probe-sets could discriminate MDS patients with a del(5q) from those without a del(5q). MDS patients with the del(5q) showed distinctive down-regulation of genes mapping to chromosome 5q, and up-regulation of the histone HIST1 gene cluster at chromosome 6p21 and of genes related to the actin cytoskeleton. In order to identify genes differentially expressed between early and advanced MDS, a comparison was made between the 18 patients with RA and the nine MDS patients with RAEBII. 762 significantly different probe sets were identified that could group together MDS patients with RAEBII. The most significant genes identified include CASP3 and FLT3, and represent potential prognostic markers or markers of disease progression. The remaining 29 MDS patients were used as a test set for class prediction using support vector machines. The FAB subtype was correctly predicted for 83% of the test samples. The presence or absence of a del(5q) was predicted correctly for 93% of the test samples. Finally, 94% of the test samples were predicted correctly as RA or RAEBII. This study provides important and new insights into the pathophysiology of MDS.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    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. 108, No. 11 ( 2006-11-16), p. 855-855
    Abstract: The immunomodulatory drug lenalidomide induces cytogenetic remissions in 75% of patients with myelodysplastic syndrome (MDS) and del(5)(q31) through unknown mechanisms. We investigated the in vitro effects of lenalidomide on growth and maturation in differentiating erythroblasts from MDS patients with del(5)(q31) (n=13) and from healthy controls (n=10). Lenalidomide selectively inhibited growth of del(5q) erythroblasts, while not affecting normal cells, including cytogenetically normal cells from MDS del(5q) patients. The inhibitory effect was more pronounced in erythroid than in myeloid cells. In order to gain insight into the mode of action of lenalidomide and to identify the molecular targets of this drug, we have investigated the gene expression profiles of the lenalidomide-treated and untreated intermediate erythroblasts from MDS del(5q) patients (n=9) and from healthy controls (n=8). GeneChip Human Genome U133 Plus 2.0 arrays (Affymetrix), covering over 47,000 transcripts representing 39,000 human genes, were used. Treatment with lenalidomide significantly influenced the pattern of gene expression in del(5q) intermediate erythroblasts, with up-regulation of VSIG4, PPIC, TPBG, and SPARC in all samples, and down-regulation of many genes involved in erythropoiesis, including HBA2, GYPA, and KLF1, in most samples. Up-regulation of SPARC (median 4.4-fold, range 2.4–9.5) is of particular interest since SPARC, a gene with known tumor suppressor functions, is both anti-proliferative and anti-angiogenic, and is located at 5q31–q32, within the commonly deleted region in MDS 5q- syndrome. Activin A was one of the most significant differentially expressed genes between lenalidomide-treated cells of MDS del(5q) patients and healthy controls. Activin A is a member of the transforming growth factor-beta superfamily, with pleiotropic functions including apoptosis of hemopoietic cells. We conclude that lenalidomide specifically inhibits growth of del(5q) erythroid progenitors, while not affecting cytogenetically normal cells. These novel findings suggest that up-regulation of SPARC and Activin A may underlie the potent effects of lenalidomide, in particular growth inhibition and anti-angiogenesis, in MDS with del(5)(q31). The localization of the SPARC gene to the CDR of the 5q- syndrome is intriguing and, in relation to the findings of the present study, we suggest that SPARC may well play a role in the molecular pathogenesis of the 5q- syndrome.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    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, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 3642-3642
    Abstract: The myelodysplastic syndromes (MDS) are a heterogeneous group of hematopoietic malignancies, characterized by blood cytopenias, ineffective hematopoiesis and a hypercellular bone marrow. We have investigated the gene expression profiles of a large group of patients with MDS in order to better understand the molecular pathogenesis of this disorder. The CD34+ cells obtained from 154 MDS patients and 17 healthy individuals were analyzed using Affymetrix U133 Plus2.0 arrays. 38 genes were up-regulated by 〉 2-fold in at least 77 MDS patients, and pathway analysis using these genes showed that the interferon signalling pathway was significantly deregulated (p=0.0006). Indeed IFIT1, the most up-regulated gene (up-regulated in 110 of 154 MDS patients), is an interferon-stimulated gene (ISG). Other ISGs, which mediate growth inhibitory effects of interferon, such as IFITM1, IFI44L and IFIT3, were markedly up-regulated in the majority of MDS patients. Up-regulation of ISGs is a major feature of MDS and may be responsible for some of the hematological characteristics of this disorder, such as peripheral blood cytopenias. We investigated differences in gene expression that could distinguish MDS patients according to their FAB subtype classification (48 patients with RA, 44 patients with RARS and 62 patients with RAEB). Hierarchical clustering performed using the 773 significantly differentially expressed probe sets identified showed that MDS patients with RARS constitute the most homogeneous group, while MDS patients with RA and RAEB show more overlap. RARS gene expression profile was characterized by up-regulation of mitochondrial-related genes and by down-regulation of ABCB7, a gene mutated in the rare inherited X-linked sideroblastic anemia with ataxia (XLSA/A). Moreover, a good separation between the 20 patients with RARS and the 20 patients with RCMD-RS was obtained by hierarchical clustering using the 86 significantly differentially expressed genes between these two WHO subgroups. One of the most significant genes was MFN1, which is essential for mitochondrial fusion and maintenance of mitochondrial morphology. The association of distinct gene expression profiles with specific cytogenetic groups was also determined, and we were able to separate by hierarchical clustering MDS patients with del(5q), patients with −7/del(7q) and patients with trisomy 8. The expression profile of patients with the del(5q) was characterized by down-regulation of genes mapping to chromosome 5q. Genes differentially expressed in patients with −7/del(7q) include LOX and UBE2H, while genes differentially expressed in patients with trisomy 8 include HRSP12 and TPM4. These findings suggest distinct molecular pathogenetic pathways for MDS patients with del(5q), −7/del(7q) and trisomy 8. In order to identify differences in gene expression associated with MDS disease progression, we compared the 48 patients with early MDS (RA) and the 35 patients with advanced MDS (RAEB2). Hierarchical clustering performed using 1081 significantly differentially expressed probe sets resulted in a good separation between MDS patients with RA and patients with RAEB2. LEF1, a regulator of neutrophilic granulopoiesis, was the most significant differentially expressed gene with higher expression levels in patients with RA and decreasing in patients with RAEB2. Other genes showing higher expression levels in patients with RA, decreasing in patients with RAEB2, include CASC5, a cancer susceptibility candidate gene, and RBBP8, a gene that plays a role in DNA-damage-induced cell cycle checkpoint control. Several genes mapping to the cell cycle pathway were significantly deregulated between early and advanced MDS. This study provides new important insights into the pathophysiology of MDS and represents a first step towards determining pathway signatures in MDS as a guide to targeted therapies.
    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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 140, No. Supplement 1 ( 2022-11-15), p. 6968-6970
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2022
    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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 3438-3438
    Abstract: The immunomodulatory drug lenalidomide induces 82% complete cytogenetic remissions in patients with myelodysplastic syndrome (MDS) and 5q31 deletion. Lenalidomide has multiple effects, but the causal mechanisms of action are unknown. In this study we assessed the direct effect of lenalidomide on hematopoietic cells from eight MDS patients with del(5)(q31) and from five healthy controls. Lenalidomide titrated up to 500 μM caused no growth inhibition of mononuclear cells from healthy controls. Selected CD34+ hematopoietic stem cells were then cultured with or without 10 μM of lenalidomide in a 14-day model for pure erythroblast differentiation in a medium containing IL-3, IL-6, and SCF, and with addition of Epo during the second week. Cell count and viability was monitored regularly, and FISH and FACS analyses were performed at day 0, 7, and 14. The median proportion of 5q- cells by FISH at day 7 was 98% (range 86–99), dropping to 88% (range 35–98) at day 14 due to a variable outgrowth of cytogenetically normal cells. Day 7 cells, the majority still being 5q-, were used for gene expression analyses. In erythroblast cultures with cells from healthy controls, lenalidomide had no inhibitory effect on fold increase of cell counts (P=0.92). However, in cultures with cells from 5q- patients, the clone with 5q deletion showed significant inhibition of fold increase at day 14 (P=0.009), while the cytogenetically normal progenitors were not inhibited (P=0.83). Gene expression profiling was performed using Affymetrix Human Genome U133 Plus 2.0 Arrays. A group of genes was found whose expression was affected by the addition of lenalidomide to the cultures of both normal erythroblasts and 5q- erythroblasts. Furthermore, lenalidomide decreased the proportion of cells expressing late erythroid markers on FACS analysis at day 14. We conclude that lenalidomide selectively inhibits in vitro growth of 5q- hematopoietic stem cells, while not affecting growth of cytogenetically normal cells from MDS patients with 5q deletion or from healthy controls. In addition, we see that lenalidomide affects cell differentiation and induces changes in gene expression.
    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
    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