Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
2.
Leukemia ; 35(3): 835-849, 2021 03.
Article in English | MEDLINE | ID: mdl-32595214

ABSTRACT

In the current World Health Organization (WHO)-classification, therapy-related myelodysplastic syndromes (t-MDS) are categorized together with therapy-related acute myeloid leukemia (AML) and t-myelodysplastic/myeloproliferative neoplasms into one subgroup independent of morphologic or prognostic features. Analyzing data of 2087 t-MDS patients from different international MDS groups to evaluate classification and prognostication tools we found that applying the WHO classification for p-MDS successfully predicts time to transformation and survival (both p < 0.001). The results regarding carefully reviewed cytogenetic data, classifications, and prognostic scores confirmed that t-MDS are similarly heterogeneous as p-MDS and therefore deserve the same careful differentiation regarding risk. As reference, these results were compared with 4593 primary MDS (p-MDS) patients represented in the International Working Group for Prognosis in MDS database (IWG-PM). Although a less favorable clinical outcome occurred in each t-MDS subset compared with p-MDS subgroups, FAB and WHO-classification, IPSS-R, and WPSS-R separated t-MDS patients into differing risk groups effectively, indicating that all established risk factors for p-MDS maintained relevance in t-MDS, with cytogenetic features having enhanced predictive power. These data strongly argue to classify t-MDS as a separate entity distinct from other WHO-classified t-myeloid neoplasms, which would enhance treatment decisions and facilitate the inclusion of t-MDS patients into clinical studies.


Subject(s)
Biomarkers, Tumor/analysis , Myelodysplastic Syndromes/classification , Myelodysplastic Syndromes/diagnosis , Neoplasms, Second Primary/classification , Neoplasms, Second Primary/diagnosis , Risk Assessment/methods , Aged , Aged, 80 and over , Female , Follow-Up Studies , Humans , Male , Middle Aged , Myelodysplastic Syndromes/therapy , Neoplasms, Second Primary/therapy , Prognosis , Retrospective Studies , Survival Rate
3.
Ann Hematol ; 99(10): 2303-2313, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32856141

ABSTRACT

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare hematologic disease characterized by a deregulated complement system, chronic Coombs-negative, intravascular hemolysis, and a variable clinical course with substantial risk to develop thromboembolic events. We analyzed diagnostic and prognostic parameters as well as clinical endpoints in 59 adult patients suffering from PNH in 5 hematology centers in Austria (observation period: 1978-2015). Median follow-up time was 5.6 years. The median clone size at diagnosis amounted to 55% and was higher in patients with classical PNH (81%) compared to patients with PNH associated with aplastic anemia (AA) or myelodysplastic syndromes (MDS) (50%). The clone size also correlated with lactate dehydrogenase (LDH) levels. In one patient, anemia improved spontaneously and disappeared with complete normalization of LDH after 16 years. Seventeen patients received therapy with eculizumab. The rate of thromboembolic events was higher in the pre-eculizumab era compared with eculizumab-treated patients but did not correlate with the presence of age-related clonal hematopoiesis or any other clinical or laboratory parameters. Peripheral blood colony-forming progenitor cell counts were lower in PNH patients compared with healthy controls. Only two patients with classical PNH developed MDS. Overall, 7/59 patients died after 0.5-32 years. Causes of death were acute pulmonary hypertension, Budd-Chiari syndrome, and septicemia. Overall survival (OS) was mainly influenced by age and was similar to OS measured in an age-matched healthy Austrian control cohort. Together, compared with previous times, the clinical course and OS in PNH are favorable, which may be due to better diagnosis, early recognition, and eculizumab therapy.


Subject(s)
Hemoglobinuria, Paroxysmal/epidemiology , Acute Kidney Injury/blood , Acute Kidney Injury/etiology , Adult , Anemia, Aplastic/epidemiology , Antibodies, Monoclonal, Humanized/therapeutic use , Austria/epidemiology , Bone Marrow/pathology , Cause of Death , Clone Cells/pathology , Colony-Forming Units Assay , Combined Modality Therapy , Complement Inactivating Agents/therapeutic use , Creatinine/blood , Disease Progression , Female , Follow-Up Studies , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Hemoglobinuria, Paroxysmal/complications , Hemoglobinuria, Paroxysmal/drug therapy , Hemoglobinuria, Paroxysmal/therapy , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Myelodysplastic Syndromes/epidemiology , Pregnancy , Pregnancy Complications, Hematologic/epidemiology , Prognosis , Thromboembolism/etiology
4.
Leuk Res ; 72: 27-33, 2018 09.
Article in English | MEDLINE | ID: mdl-30075323

ABSTRACT

The IPSS-R proved to be a powerful tool for the assessment of prognosis in MDS patients. We aimed at a validation of the IPSS-R for patients with MDS harboring deletion (5q) isolated or accompanied by additional aberrations. The study was based on 444 MDS patients from MDS centers in Europe. 67% of the patients were female, median age was 69 years. 43.5% had MDS del(5q), 5.9% were diagnosed with RCUD, 2.0% RARS, 18.4% RCMD, 14.6% RAEB-I and 15.5% RAEB-II. According to the IPSS-R, there were 9.9% very low, 39.6% low, 16.6% intermediate, 12.8% high, 20.9% very high risk patients. For very low risk patients survival was 7.5 years, low 9.0 years, intermediate 6.5 years, high 1.5 years and very high 0.7 years (p < 0.001). For low and intermediate risk, the probability of AML evolution was significantly different (p = 0.03) as well as for high versus very high risk groups (p = 0.002). The IPSS-R proved to be an appropriate prognostic tool for MDS with del(5q).


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/mortality , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Female , Humans , Male , Middle Aged , Myelodysplastic Syndromes/therapy , Risk Factors , Survival Rate
5.
Leukemia ; 29(7): 1502-13, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25721895

ABSTRACT

A risk-adapted treatment strategy is mandatory for myelodysplastic syndromes (MDS). We refined the World Health Organization (WHO)-classification-based Prognostic Scoring System (WPSS) by determining the impact of the newer clinical and cytogenetic features, and we compared its prognostic power to that of the revised International Prognostic Scoring System (IPSS-R). A population of 5326 untreated MDS was considered. We analyzed single WPSS parameters and confirmed that the WHO classification and severe anemia provide important prognostic information in MDS. A strong correlation was found between the WPSS including the new cytogenetic risk stratification and WPSS adopting original criteria. We then compared WPSS with the IPSS-R prognostic system. A highly significant correlation was found between the WPSS and IPSS-R risk classifications. Discrepancies did occur among lower-risk patients in whom the number of dysplastic hematopoietic lineages as assessed by morphology did not reflect the severity of peripheral blood cytopenias and/or increased marrow blast count. Moreover, severe anemia has higher prognostic weight in the WPSS versus IPSS-R model. Overall, both systems well represent the prognostic risk of MDS patients defined by WHO morphologic criteria. This study provides relevant in formation for the implementation of risk-adapted strategies in MDS.


Subject(s)
Myelodysplastic Syndromes/classification , Myelodysplastic Syndromes/diagnosis , World Health Organization , Adolescent , Adult , Aged , Aged, 80 and over , Combined Modality Therapy , Cytogenetic Analysis , Female , Follow-Up Studies , Humans , International Cooperation , Male , Middle Aged , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/therapy , Neoplasm Staging , Prognosis , Research Design , Risk Assessment , Survival Rate , Young Adult
6.
Leukemia ; 27(10): 1988-95, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23787396

ABSTRACT

Monosomal karyotype (MK) is associated with an adverse prognosis in patients in acute myeloid leukemia (AML). This study analyzes the prognostic impact of MK in a cohort of primary, untreated patients with myelodysplastic syndromes (MDS). A total of 431 patients were extracted from an international database. To analyze whether MK is an independent prognostic marker in MDS, cytogenetic and clinical data were explored in uni- and multivariate models regarding overall survival (OS) as well as AML-free survival. In all, 204/431 (47.3%) patients with MK were identified. Regarding OS, MK was prognostically significant in patients with ≤ 4 abnormalities only. In highly complex karyotypes (≥ 5 abnormalities), MK did not separate prognostic subgroups (median OS 4.9 months in MK+ vs 5.6 months in patients without MK, P=0.832). Based on the number of abnormalities, MK-positive karyotypes (MK+) split into different prognostic subgroups (MK+ and 2 abnormalities: OS 13.4 months, MK+ and 3 abnormalities: 8.0 months, MK+ and 4 abnormalities: 7.9 months and MK+ and ≥ 5 abnormalities: 4.9 months; P<0.01). In multivariate analyses, MK was not an independent prognostic factor. Our data support the hypothesis that a high number of complex abnormalities, associated with an instable clone, define the subgroup with the worst prognosis in MDS, independent of MK.


Subject(s)
Chromosome Aberrations , Monosomy/genetics , Myelodysplastic Syndromes/mortality , Adult , Aged , Aged, 80 and over , Cohort Studies , Female , Follow-Up Studies , Humans , Karyotyping , Male , Middle Aged , Multivariate Analysis , Myelodysplastic Syndromes/classification , Myelodysplastic Syndromes/genetics , Prognosis , Survival Rate , Young Adult
7.
Leukemia ; 26(6): 1286-92, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22289990

ABSTRACT

Myelodysplastic syndromes (MDS) with del(5q) are considered to have a benign course of the disease. In order to address the issue of the propensity of those patients to progress to acute myeloid leukemia (AML), data on 381 untreated patients with MDS and del(5q) characterized by low or intermediate I International Prognostic Scoring System (IPSS) risk score were collected from nine centers and registries. Median survival of the entire group was 74 months. Transfusion-dependent patients had a median survival of 44 months vs 97 months for transfusion-independent patients (P<0.0001). Transfusion need at diagnosis was the most important patient characteristic for survival. Of the 381 patients, 48 (12.6%) progressed to AML. The cumulative progression rate calculated using the Kaplan-Meier method was 4.9% at 2 years and 17.6% at 5 years. Factors associated with the risk of AML transformation were high-risk World Health Organization adapted Prognostic Scoring System (WPSS) score, marrow blast count >5% and red-cell transfusion dependency at diagnosis. In conclusion, patients with MDS and del(5q) are facing a considerable risk of AML transformation. More detailed cytogenetic and molecular studies may help to identify the patients at risk of progression.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Myelodysplastic Syndromes/genetics , Preleukemia/genetics , Adult , Aged , Aged, 80 and over , Disease Progression , Female , Humans , Male , Middle Aged , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/mortality , Preleukemia/diagnosis , Preleukemia/mortality , Prognosis , Registries , Retrospective Studies , Risk Factors , Survival Rate
8.
Leukemia ; 25(1): 110-20, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20882045

ABSTRACT

This cooperative study assessed prognostic factors for overall survival (OS) and risk of transformation to acute myeloid leukemia (AML) in 541 patients with de novo myelodysplastic syndrome (MDS) and deletion 5q. Additional chromosomal abnormalities were strongly related to different patients' characteristics. In multivariate analysis, the most important predictors of both OS and AML transformation risk were number of chromosomal abnormalities (P<0.001 for both outcomes), platelet count (P<0.001 and P=0.001, respectively) and proportion of bone marrow blasts (P<0.001 and P=0.016, respectively). The number of chromosomal abnormalities defined three risk categories for AML transformation (del(5q), del(5q)+1 and del(5q)+ ≥ 2 abnormalities) and two for OS (one group: del(5q) and del(5q)+1; and del(5q)+ ≥ 2 abnormalities, as the other one); with a median survival time of 58.0 and 6.8 months, respectively. Platelet count (P=0.001) and age (P=0.034) predicted OS in patients with '5q-syndrome'. This study demonstrates the importance of additional chromosomal abnormalities in MDS patients with deletion 5q, challenges the current '5q-syndrome' definition and constitutes a useful reference series to properly analyze the results of clinical trials in these patients.


Subject(s)
Chromosome Aberrations , Myelodysplastic Syndromes/genetics , Adult , Aged , Aged, 80 and over , Anemia, Macrocytic/genetics , Anemia, Macrocytic/mortality , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Female , Humans , Karyotyping , Male , Middle Aged , Myelodysplastic Syndromes/mortality , Prognosis , Retrospective Studies
9.
Ann Oncol ; 21(1): 114-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19605505

ABSTRACT

BACKGROUND: The evaluation of comorbidity is of increasing importance in patients with hematologic disorders. PATIENTS AND METHODS: In the present study, the influence of comorbidity on survival and acute myeloid leukemia (AML) evolution was analyzed retrospectively in 419 patients with de novo myelodysplastic syndromes (MDS) (observation period: 1985-2007). The median age was 71 years (range 24-91 years). Two different scoring systems, the hematopoietic stem-cell transplantation-specific comorbidity index (HCT-CI) and the Charlson comorbidity index (CCI) were applied. RESULTS: The HCT-CI was found to be a significant prognostic factor for overall survival (OS, P < 0.05) as well as event-free survival (EFS, P < 0.05) in our patients, whereas the CCI was of prognostic significance for OS (P < 0.05), but not for EFS. For AML-free survival, neither the HCT-CI nor the CCI were of predictive value. A multivariate analysis including age, lactate dehydrogenase, ferritin, karyotype, number of cytopenias, French-American-British groups, and comorbidity was applied. Comorbidity was found to be an independent prognostic factor in patients with low- or int-1-risk MDS (P < 0.05) regarding OS and EFS. CONCLUSIONS: Together, our data show that comorbidity is an important risk factor for OS and EFS in patients with MDS.


Subject(s)
Comorbidity , Leukemia, Myeloid, Acute/epidemiology , Myelodysplastic Syndromes/epidemiology , Precancerous Conditions/epidemiology , Adult , Aged , Aged, 80 and over , Austria , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Retrospective Studies , Young Adult
10.
Ann Oncol ; 21(1): 120-5, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19605511

ABSTRACT

BACKGROUND: The International Prognostic Scoring System (IPSS) is the golden standard to assess prognosis in myelodysplastic syndromes (MDS). The aim of this analysis was to study age and gender as interacting variables for individualized prognostication. PATIENTS AND METHODS: In all, 897 patients with primary MDS treated with supportive care only were examined in a retrospective multicenter study. A Cox model was developed to determine the prognostic impact of age and gender on survival and to examine their modulating influence on IPSS results. Based on main effects and interactions of these variables, we established an individualized age- and gender-adapted scoring system to improve prognostication in MDS. RESULTS: While the risk of a patient in the IPSS is best represented by the values 0 (low), +1 (intermediate-1), +2 (intermediate-2), and +3 (high), these values were found to vary between -1.9 and +3.5 in the same patients when including age and gender. Whereas in low-risk MDS, male patients were found to have a less favorable survival, a particularly high risk (+3.5) was found in younger (< or = 66 years) high-risk female patients. CONCLUSION: The inclusion of age and gender and their respective interactions contribute to improved and individualized prognostication in MDS.


Subject(s)
Myelodysplastic Syndromes/mortality , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Proportional Hazards Models , Risk Factors , Sex Factors , Young Adult
11.
Eur J Clin Invest ; 39(5): 406-11, 2009 May.
Article in English | MEDLINE | ID: mdl-19320908

ABSTRACT

BACKGROUND: Transfusion-related morbidity is an emerging problem in chronically transfused patients with myelodysplastic syndromes (MDS). Although several iron-chelating drugs are available, the optimal way of treatment of iron-overload remains uncertain. A major disadvantage of deferoxamine is that the drug has to be applied as continuous subcutaneous infusion. Therefore, novel oral agents have been developed. One of these drugs is deferasirox (Exjade). MATERIALS AND METHODS: We report on 14 MDS patients who were treated with deferasirox (500-1500 mg daily) for up to 24 months. In these patients, treatment responses were recorded by determining serum ferritin levels before and during therapy and by applying recently established response criteria. RESULTS: In all patients except one, ferritin levels decreased during therapy. Four patients showed a complete response, one a minor response and five a stable iron load. In the responding patients, initially elevated liver enzymes decreased substantially. No substantial change in transferrin saturation or transfusion frequency was recorded. Side effects were mild and tolerable in most patients. In one patient, treatment with deferasirox was stopped because of impaired kidney function. CONCLUSION: Our data show that treatment with deferasirox is a reasonable approach to counteract iron overload in patients with MDS.


Subject(s)
Benzoates/therapeutic use , Iron Chelating Agents/therapeutic use , Iron Overload/drug therapy , Myelodysplastic Syndromes/drug therapy , Triazoles/therapeutic use , Administration, Oral , Aged , Deferasirox , Female , Humans , Iron/analysis , Male , Middle Aged , Treatment Outcome
12.
Eur J Clin Invest ; 38(1): 43-52, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18173550

ABSTRACT

BACKGROUND: Recent data suggest that the mammalian target of rapamycin (mTOR) is involved in the regulation of growth of neoplastic cells in chronic myeloid leukaemia (CML). PATIENTS AND METHODS: We treated six patients with imatinib-resistant CML in haematological relapse (leukocytes > 20,000 microL(-1)) with rapamycin at 2 mg per os daily for 14 consecutive days, with dose-adjustment allowed to reach a target rapamycin serum concentration of 10-20 pg mL(-1). RESULTS: A major leukocyte response with decrease to less than 10,000 microL(-1) was obtained in two patients, and a minor transient response was seen in two other patients. In responding patients, we also observed a decrease in vascular endothelial growth factor (VEGF) mRNA levels in circulating leukaemic cells. Side effects during rapamycin treatment were mild in most patients. In one patient, pneumonia developed. Rapamycin was also found to counteract growth of CML cells in vitro as determined by (3)H-thymidine incorporation. Moreover, rapamycin inhibited the growth of Ba/F3 cells exhibiting various imatinib-resistant mutants of BCR/ABL, including the T315I variant that exhibits resistance against most currently available BCR/ABL kinase inhibitors. CONCLUSIONS: Rapamycin shows antileukaemic effects in imatinib-resistant CML in vitro and in vivo. Larger trials with rapamycin or rapamycin-derivatives in combination with other targeted drugs are warranted to further determine clinical efficacy in CML.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Chronic-Phase/drug therapy , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Sirolimus/therapeutic use , Aged , Benzamides , Drug Evaluation , Drug Resistance, Neoplasm , Female , Humans , Imatinib Mesylate , Male , Middle Aged , Pilot Projects , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism
13.
Eur J Clin Invest ; 38(1): 53-60, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18173551

ABSTRACT

BACKGROUND: Presenting the same histological diagnosis, multiple myeloma (MM) shows a large genomic variety, resulting in variable times of overall survival. MATERIALS AND METHODS: To investigate major cytogenetic categories (any 14q-translocation, t(11;14), t(4;14), 13q-deletions, 17p-deletions) and their clinical consequences in MM after a pre-existing monoclonal gammopathy (MM post-MGUS), we performed a comparative analysis of 41 patients with MM post-MGUS and 287 patients with unknown prior history MM (U-MM). RESULTS: In MM post-MGUS, a t(11;14) was found to be more frequent than in U-MM (24% vs. 14%) and it was associated with significantly shortened survival (24 months vs. 70 months in U-MM; P = 0.01). MM post-MGUS was further characterized by a higher frequency of 13q-deletions only (absence of all other specific abnormalities; 28% vs. 12% in U-MM; P = 0.02). A 13q-deletion only was an indicator of long survival in MM post-MGUS (median not yet reached) as opposed to U-MM (median survival, 29 months; P = 0.001). 17p-deletions were infrequent in MM post-MGUS (3% vs. 16% in U-MM; P = 0.04). Survival times for patients with t(4;14) and/or 17p-deletions and other abnormalities were similar in both MM patient cohorts. CONCLUSIONS: Our data suggest that t(11;14) and 13q-deletions have distinct prognostic implications in the context of MM post-MGUS.


Subject(s)
Chromosome Deletion , Monoclonal Gammopathy of Undetermined Significance/complications , Multiple Myeloma/genetics , Translocation, Genetic , Adult , Aged , Aged, 80 and over , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 14 , Female , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Multiple Myeloma/complications , Prognosis , Survival Rate
14.
Eur J Clin Invest ; 38(3): 143-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18218040

ABSTRACT

Transfusion-related morbidity is an emerging challenge in chronically transfused patients with low-risk myelodysplastic syndromes (MDS). In these patients, transfusion-induced iron overload may represent a leading medical problem. However, although iron-chelating drugs are available, little is known about optimal diagnostic tools, predisposing factors, and the optimal management of these patients. In the current article, we provide recommendations for the diagnosis, prevention and treatment of iron overload in MDS and propose treatment response criteria. Consensus criteria and resulting recommendations were discussed and formulated by members of the MDS platform of the Austrian Society of Haematology and Oncology in a series of meetings and conferences in 2006 and 2007. These recommendations should facilitate and assist in recognition of iron overload, selection of patients, timing of treatment, drug selection and the measurement of treatment responses.


Subject(s)
Chelation Therapy/methods , Erythrocyte Transfusion/adverse effects , Iron Chelating Agents/therapeutic use , Iron Overload/therapy , Myelodysplastic Syndromes/therapy , Ferritins/blood , Guidelines as Topic , Humans , Iron Overload/physiopathology , Iron Overload/prevention & control , Myelodysplastic Syndromes/complications
15.
Leukemia ; 19(12): 2223-31, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16193087

ABSTRACT

The international prognostic scoring system (IPSS) is considered the gold standard for risk assessment in primary myelodysplastic syndromes (MDS). This score includes several prognostic factors except serum lactate dehydrogenase (LDH). We evaluated the prognostic power of LDH as an additional variable in IPSS-based risk assessment. For this purpose, a total of 892 patients with primary MDS registered by the Austrian-German cooperative MDS study group was analyzed retrospectively. Multivariate analysis confirmed the value of established parameters such as medullary blasts, karyotype and peripheral cell counts and showed that elevated LDH was associated with decreased overall survival (P<0.00005) and increased risk of AML development (P<0.00005), independent of the system used to classify MDS (FAB or WHO). Moreover, elevated LDH was found to be a significant predictor of poor survival within each IPSS risk group and within each FAB group except RAEB-T. To exploit these results for refined prognostication, each IPSS risk group was split into two separate categories (A=normal LDH vs B=elevated LDH). Using this LDH-assisted approach, it was possible to identify MDS patients with unfavorable prognosis within the low and intermediate IPSS risk groups. We propose that the IPSS+LDH score should improve clinical decision-making and facilitate proper risk stratification in clinical trials.


Subject(s)
Clinical Enzyme Tests , L-Lactate Dehydrogenase/blood , Myelodysplastic Syndromes/diagnosis , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Models, Theoretical , Multivariate Analysis , Myelodysplastic Syndromes/classification , Myelodysplastic Syndromes/mortality , Prognosis , Retrospective Studies , Risk Assessment , Survival Rate
16.
Leukemia ; 18(11): 1879-82, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15385925

ABSTRACT

Molecular and genetic events associated with the transition from monoclonal gammopathy of undetermined significance (MGUS) to multiple myeloma (MM) are still poorly characterized. We investigated serial bone marrow specimens from 11 patients with MGUS who eventually progressed to MM (MM post-MGUS) by interphase fluorescence in situ hybridization for immunoglobulin heavy-chain gene (IgH) translocations and chromosome 13q deletions (del(13q)). In nine patients, IgH translocations were present both in MGUS and MM post-MGUS plasma cells, including three t(11;14)(q13;q32) and one t(4;14)(p16;q32), which was observed already 92 months prior to MM. Similarly, all five MM patients with del(13q) had this aberration already at the MGUS stage. Two patients without IgH translocation and del(13q) had chromosomal gains suggesting hyperdiploidy, but IgH translocations and/or del(13q) did not emerge at MM post-MGUS. IgH translocations and del(13q) are early genetic events in monoclonal gammopathies, suggesting that additional events are required for the transition from stable MGUS to progressive MM.


Subject(s)
Chromosomes, Human, Pair 13 , Gene Deletion , Immunoglobulin Heavy Chains/genetics , Multiple Myeloma/genetics , Paraproteinemias/genetics , Translocation, Genetic , Aged , Bone Marrow , Disease Progression , Humans , In Situ Hybridization, Fluorescence , Interphase , Middle Aged
17.
Blood ; 98(10): 2935-41, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11698274

ABSTRACT

In 1999 a working group of the World Health Organization (WHO) published a revised classification for myelodysplastic syndromes (MDS): RA, RARS, refractory cytopenia with multilineage dysplasia (RC+Dys), RAEB I and II, del (5q) syndrome, and MDS unclassifiable. Chronic myelomonocytic leukemia (CMML) and RAEB-t were excluded. Standard French-American-British (FAB) and new WHO classifications have been compared in a series of patients (n = 431) from a single center, analyzing morphologic, clinical, and cytogenetic data. According to the WHO findings, dysgranulocytopoiesis or dysmegakaryocytopoiesis only were found in 26% of patients with less than 5% medullary blasts. These patients are thus unclassified and should remain in the subgroups RA and RARS. Splitting of heterogeneous RAEB into 2 subgroups according to blast count was supported by a trend to a statistically significant difference in the single-center study population. Patients with CMML whose white blood cell counts are above 13 000/microL may be excluded from the MDS classification, as warranted by WHO, but a redistribution of patients with dysplastic CMML according to medullary blast count leads to more heterogeneity in other WHO subgroups. Although the natural courses of RAEB-T and acute myeloid leukemia (AML) with dysplasia are different, comparable median survival durations after treatment in patients with RAEB-T and AML were in favor of the proposed 20% medullary blast threshold for AML. The homogeneity of subgroups was studied by evaluating prognostic scores. A significant shift into lower IPSS risk groups was evident in the new classification. These data cannot provide evidence for the new WHO proposal, which should not be adopted for routine clinical use at present. Some of its aspects can provide a starting point for further studies involving refined cytogenetics and clinical results.


Subject(s)
Myelodysplastic Syndromes/classification , Adult , Aged , Aged, 80 and over , Bone Marrow/pathology , Cell Count , Female , Humans , Leukemia, Myelomonocytic, Chronic/blood , Leukemia, Myelomonocytic, Chronic/classification , Leukemia, Myelomonocytic, Chronic/mortality , Leukemia, Myelomonocytic, Chronic/pathology , Leukocyte Count , Life Tables , Male , Middle Aged , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/pathology , Retrospective Studies , Survival Analysis , World Health Organization
18.
Leuk Res ; 25(9): 741-7, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11489467

ABSTRACT

In chronic myelomonocytic leukemia (CMML) segregation of two subtypes has been suggested depending on WBC count-myelodysplastic (MD-CMML) and myeloproliferative (MP-CMML). In a retrospective analysis of 91 (60/31) previously untreated CMML patients, we compared the presenting clinical, haematological, laboratory and bone marrow features and examined the clinical impact of this reclassification. LDH values and bone marrow cellularity were significantly increased in MP-CMML. Median survival was significantly longer for patients with MD-CMML, progression rate was higher for MP-CMML. Patients with MD-CMML had longer median preleukemic duration; after transition to AML, MP-CMML patients had longer median survival. In MDS phase anemia was more common in MP-CMML and thrombocytopenia more common in MD-CMML whereas transfusion rates showed no difference. Evaluation of prognostic scoring systems for both groups confirmed that patients' characteristics and outcome could be well compared. Our data suggest that segregation into MD-CMML and MP-CMML is justified.


Subject(s)
Leukemia, Myelomonocytic, Chronic/complications , Myeloproliferative Disorders/etiology , Neural Tube Defects/etiology , Aged , Aged, 80 and over , Disease Progression , Female , Humans , Leukemia, Myelomonocytic, Chronic/mortality , Leukemia, Myelomonocytic, Chronic/pathology , Male , Middle Aged , Myeloproliferative Disorders/mortality , Myeloproliferative Disorders/pathology , Neural Tube Defects/mortality , Neural Tube Defects/pathology , Prognosis , Retrospective Studies , Survival Analysis , Time Factors
19.
Stem Cells ; 19(4): 321-8, 2001.
Article in English | MEDLINE | ID: mdl-11463952

ABSTRACT

The granulocyte-derived hemoregulatory peptide pyroGlu-Glu-Asp-Cys-Lys = pEEDCK is known to keep hematopoietic cells quiescent. When oxidized to its dimeric form (pEEDCK)2, it activates growth of hematopoietic progenitors in association with stroma-derived cytokines. (pEEDCK)2 has a Cys-Cys motif which is also a typical feature of the macrophage inflammatory protein (MIP-1alpha). The present study was designed to analyze differences between the response of normal and leukemic progenitor cells to (pEEDCK)2 or MIP-1alpha. When long-term bone marrow cultures (LTBMCs) were incubated with (pEEDCK)2 or MIP-1alpha and/or cytokines, the stimulatory effect on colony-forming units-granulocyte/erythroid/macrophage/megakaryocyte of LTBMC from chronic myeloid leukemia (CML) patients was less than 50% compared to LTBMC from healthy humans. No difference in oncogene expression could be observed in LTBMC from CML patients regarding reduction of Philadelphia chromosome-associated transcription of the BCR-ABL gene. With respect to the expression of growth and differentiation-associated genes (Galpha16, 5-lipoxygenase, phospholipaseA2, c-kit, and CD34), which were analyzed from LTBMC by semiquantitative reverse transcriptase-polymerase chain reaction, the same transcription rate was observed in CML patients and healthy donors. However, two isoforms of a key enzyme of oxidative metabolism, carnitine palmitoyltransferase (CPT1A and CPT1B), showed 50-fold higher expression rates in LTBMC cells of healthy donors compared to CML patients. It is known that a decrease in oxidative metabolism is associated with an increase in redox equivalents in malignancy. This might result in a reduction of disulphide bonds in (pEEDCK)2 or MIP-1alpha, thus inducing a downregulation of these factors in bone marrow from CML patients.


Subject(s)
Bone Marrow Cells/drug effects , Hematopoietic Stem Cells/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology , Macrophage Inflammatory Proteins/pharmacology , Oligopeptides/pharmacology , Animals , Bone Marrow Cells/physiology , Cells, Cultured , Chemokine CCL3 , Chemokine CCL4 , Colony-Forming Units Assay , Cytokines/pharmacology , Growth Inhibitors/pharmacology , Hematopoietic Stem Cells/physiology , Humans , Mice , Pyrrolidonecarboxylic Acid/analogs & derivatives
20.
Am J Hematol ; 64(3): 156-60, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10861809

ABSTRACT

The hematopoiesis-specific G protein alpha subunit Galpha16 is a specific element in the signal transduction of the early hematopoietic cytokine network. As Galpha16 mRNA can be detected in early hematopoietic progenitor cells, RT-PCR for Galpha16 can be used as a sensitive marker of hematopoietic activity. The aim of this study was to test the possible use of Galpha16 determinations for monitoring cytokine effects on hematopoietic recovery after chemotherapy in patients. We correlated presence of Galpha16 mRNA and CD34 surface antigen with hematopoietic recovery in six lymphoma patients undergoing salvage therapy with different cytokine support (IEV followed by G-CSF, IL-3, or placebo). Regardless of different cytokine schedules with different time courses, hematopoietic recovery was always preceded by transcription of Galpha16. Monitoring the expression of Galpha16 mRNA by RT-PCR is a highly sensitive diagnostic tool for analyzing hematopoietic recovery after chemotherapy and for characterizing the effects of cytokines on hematopoiesis.


Subject(s)
Antigens, CD34/analysis , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cytokines/pharmacology , Hematopoiesis/drug effects , Heterotrimeric GTP-Binding Proteins/genetics , Lymphoma/drug therapy , Adult , Antigens, CD34/biosynthesis , Epirubicin/administration & dosage , Etoposide/administration & dosage , Female , GTP-Binding Protein alpha Subunits, Gq-G11 , Gene Expression , Hematopoiesis/immunology , Humans , Ifosfamide/administration & dosage , Male , Middle Aged , RNA, Messenger/analysis , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...