Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
Filter
  • American Society of Hematology  (116)
  • 1
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 2688-2688
    Abstract: Introduction: Several recent publications have advanced our knowledge of the prognostic significance of clonal cytogenetic abnormalities in MDS, yet the genetic risk assessment of the rare karyotypic aberrations in MDS patients (pts) remains unknown. Using the German-Austrian (G-A) Cytogenetics Database, we previously defined 24 cytogenetic prognostic subgroups; however, 12 subgroups characterized by non-complex (isolated or one additional abnormality only) karyotypes with del(9q), del(15q), t(15q), del(12p), −X, t(1q), t(7q), t(17q), −21, t(11q23), +19, t(5q) were observed infrequently ( & lt;10 pts) and considered too few for an informative risk assessment. To increase the number of informative pts and expand/validate the statistical robustness of these rare cytogenetic subgroups, an international collaboration was initiated and promoted under the auspices of the MDS Foundation. Patients and Methods: A total of 90 new MDS pts with rare recurring abnormalities was collected from 12 MDS Foundation Centers of Excellence, of which 66 pts fulfilled the non-complex requirement. Survival was estimated by Kaplan-Meier analysis and restricted to pts treated by supportive care only. The final analysis included 108 pts: del(9q)(n=10); del(12p)=17; −X=10; t(1q)=13; t(7q)=14; −21=12; t(11q23)=12; +19=10; t(5q)=10. The frequency of non-complex del(15q), t(15q) and t(17q) remained below ten pts and deemed ineligible for further analysis at this time. Results: The pooled international data showed an excellent correlation with the G-A data set in 6 of 10 cytogenetic subgroups: t(7q): 34.7 months (mo.) median survival (G-A) vs. 34.7 mo. (new data), del(9q): not reached vs. 63.1 mo., t(11q23): 20.0 vs. 28.0 mo, del(12p): 108.0 mo. vs. not reached, +19: 19.8 vs. 21.7 mo., −21: 32.0 vs. 35.0 mo. A moderate correlation was found for t(1q): 34.7 mo. vs. not reached and for del(15q): not reached vs. 26.7 mo. Discordant median survival was observed for t(5q): 4.4 mo. vs. not reached and −X: 56.4 mo. vs. 15.7 mo. Conclusions: Through an international collaboration, we were able to define the prognostic impact of nine distinct yet infrequent, recurring cytogenetic aberrations observed in MDS pts. The discordant median survival differences observed in two subgroups may be attributed to various translocation partners and/or secondary abnormalities. Even though data collection remains ongoing, the current results expand and refine the cytogenetic prognostic classification system for pts suffering from MDS and underscore the need for standardized cytogenetic testing in MDS for further international collaborations.
    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 ...
  • 2
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 1658-1658
    Abstract: Myelodysplastic syndromes have been reclassified by new WHO proposals published in 2008. So far, there are no data on haematological features of the newly composed subtypes. We analyzed 2063 patients with MDS diagnosed according to the WHO 2008 classification. Patients were characterized by their initial haematological features. We especially payed attention towards isolated cytopeniae with regard to the new classification of MDS subcategories. Analyses were performed by the use of the MDS registry Duesseldorf and included hemoglobin level, platelets, leukocytes and karyotype (available in 1091 patients) as well as age, gender, IPSS and WPSS at the time of diagnosis. All items have been assessed in our haematological laboratory and in the department of human genetics, respectively. The table shows characteristic features of MDS patients according to their WHO subtype. The patients’ median age did not differ substantially. There was a preponderance of females in the del(5q), RARS and RC group. Median hemoglobin level ranked lowest in the del(5q) and RAEB II group. Median thrombocyte, leukocyte as well as granulocyte level progressively decreased with more advanced WHO risk groups. Chromosomal aberrations were more frequent with increasing medullary blasts and degree of dysplasia. Likewise, patients with these characteristics were categorized into more advanced WPSS subtypes. About 90% of all patients were anemic, that is patients with a hemoglobin level of under 12 g/dl. Leukocytopenia as well as thrombocytopenia were more severe with increasing medullary blast counts. Of interest, 46% of RC patients showed a leukocytopenia ( & lt;4000/μl) and 37% had thrombocytopenia ( & lt;150.000/μl). 26% of this group showed an isolated anemia, whereas only 1–3% of the patients presented with an isolated leukocytopenia or thrombocytopenia. The majority of MDS patients presents with bicytopenia, regardless of subtypes. The degree of hematopoietic insufficiency is strongly correlated with the amount of medullary blasts, but as well with the degree of dysplasia in blood and marrow. An inclusion of the number of cytopenia into the WHO classification would lead to an alteration of subcategories and result in a shift towards a higher percentage of multilineage MDS. RC RARS MDS with del(5q) RCMD RAEB I RAEB II Frequency 184 162 68 950 306 393 Age 70 70 64 72 69 67 Gender (%) M 48 48 35 55 58 56 W 52 52 65 45 42 44 Hemoglobin (g/dl) 9,5 9,5 9 9,2 9,2 9 Thrombocytes (1000/μl) 124 293 284 146 96 80 Leukocytes (/μl) 4800 5500 4300 3900 3100 2800 Granulocytes 1950 3200 2400 2200 1400 1200 Chromosomal Low 70 87 96 63 57 51 risk by IPSS Intermediate 19 10 2 18 16 19 (%) High 11 3 2 19 27 30 Initial IPSS (%) 0 48 83 72 34 0 0 1 42 14 26 53 2 7 2 10 3 2 13 55 50 3 0 0 0 0 40 43 Initial WPSS 0 48 73 40 30 0 0 (%) 1 30 27 60 40 0 0 2 22 0 0 30 33 0 3 0 0 0 0 50 59 4 0 0 0 0 18 41 Cytopenia (%) Anemia ( & lt;12 g/dl) 85 97 98 89 86 90 Leukopenia 46 18 39 50 64 89 ( & lt;4000/μl) Thrombocytopenia 37 3 8 35 52 80 ( & lt;150.000/μl) Isolated (%) Anemia 26 78 51 27 14 7 Leukocytopenia 1,5 0 1 1 2 1 Thrombocytopenia 3 0 0 2 3 2 Pancytopenia 22 3 6 24 35 44 (%) Survival (mths.) 561 69 80 36 19 10 AML after 3 3 15 15 32 63 3 yrs. (%)
    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 ...
  • 3
    In: Blood, American Society of Hematology, Vol. 110, No. 13 ( 2007-12-15), p. 4385-4395
    Abstract: We have generated a large, unique database that includes morphologic, clinical, cytogenetic, and follow-up data from 2124 patients with myelodysplastic syndromes (MDSs) at 4 institutions in Austria and 4 in Germany. Cytogenetic analyses were successfully performed in 2072 (97.6%) patients, revealing clonal abnormalities in 1084 (52.3%) patients. Numeric and structural chromosomal abnormalities were documented for each patient and subdivided further according to the number of additional abnormalities. Thus, 684 different cytogenetic categories were identified. The impact of the karyotype on the natural course of the disease was studied in 1286 patients treated with supportive care only. Median survival was 53.4 months for patients with normal karyotypes (n = 612) and 8.7 months for those with complex anomalies (n = 166). A total of 13 rare abnormalities were identified with good (+1/+1q, t(1q), t(7q), del(9q), del(12p), chromosome 15 anomalies, t(17q), monosomy 21, trisomy 21, and −X), intermediate (del(11q), chromosome 19 anomalies), or poor (t(5q)) prognostic impact, respectively. The prognostic relevance of additional abnormalities varied considerably depending on the chromosomes affected. For all World Health Organization (WHO) and French-American-British (FAB) classification system subtypes, the karyotype provided additional prognostic information. Our analyses offer new insights into the prognostic significance of rare chromosomal abnormalities and specific karyotypic combinations in MDS.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 113, No. 8 ( 2009-02-19), p. 1866-1868
    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 ...
  • 5
    In: Blood, American Society of Hematology, Vol. 138, No. Supplement 1 ( 2021-11-05), p. 3700-3700
    Abstract: Introduction: Iron storage in patients (pts) with myelodysplastic syndromes at the time of diagnosis may vary from normal to iron overload. Even before the first blood transfusion, storage iron can be increased due to down-regulation of hepcidin and subsequent increase in duodenal iron uptake. Iron overload is known to worsen the prognosis of MDS patients, partly due to iron-related organ damage after long-term transfusion therapy, and partly due to an increased risk of infections. However, it is unclear whether increased storage iron at the time of diagnosis already has a prognostic influence. We assessed bone marrow iron stores at the time of MDS diagnosis and correlated them with clinical outcome. Methods: In a retrospective analysis of 3762 adult MDS patients from the Düsseldorf MDS Registry, Prussian blue staining of marrow smears was performed in our cytology lab to assess iron stores according to the following categories: normal or decreased iron stores versus increased iron stores versus iron overload. Patients were followed up for survival and AML evolution until June 2021. Median time of follow-up was 20 months. 67.4% of the patients died during the course of the disease. Results: The study included 3.762 adult patients who received their initial diagnosis of MDS between 1970 and 2021. 58% were diagnosed as non-blastic MDS ( MDS SLD (RS) (n=240), MDS MLD (RS) (n=350), MDSdel(5q) (n=107), and MDS-U (n=25). Iron stores were decreased in 8% of the patients, normal in 44%, increased in 41%, and strongly increased in 7% (massive iron overload). In 282 cases, histologic assessment of storage iron was available. When comparing cytologic and histologic assessment, we found a strong correlation (p & lt;0.0005), since 87% of the patients with increased iron on cytomorphology also showed increased iron as assessed by histopathology. However, 37% of the patients who cytologically showed normal iron stores, were reported to have slightly increased iron as assessed by histopathology. Median and mean serum ferritin values of patients with normal or decreased iron stores were 295 and 629 µg/l, respectively, as compared to 548 and 902 µg/l, respectively, in patients with increased iron stores. The cumulative risk of AML evolution was not associated with the results of iron staining. Regarding survival, we found that patients with decreased or normal storage iron had a median survival of 31 months, whereas those with increased iron had a median survival of 28 months (p=0.007). Focusing on patients with non-blastic MDS, the difference was not significant (46 vs 44 ms). However, patients who presented as EB I (n=435), EBII (n=510), AML MRC (n=264), CMML I (n=254), or CMML II (n=77), showed a prognostic impact of storage iron; patients with increased iron had a median survival of 11 months, as compared to 16 months in patients with normal or decreased iron (p & lt;0.0005). Conclusion: Increased tissue iron in the bone marrow at the time of diagnosis is associated with inferior survival in patients with MDS, primarily in patients with higher risk MDS. At diagnosis, patients are not yet transfusion-dependent. This suggests that increased iron reflects a prolonged period of increased duodenal iron uptake as a consequence of ineffective erythropoiesis. Therefore, increased marrow iron at the time of MDS diagnosis seems to be a surrogate parameter of hematopoietic insufficiency, which is the real cause of inferior prognosis. Disclosures Nachtkamp: Jazz: Honoraria; Bsh medical: Honoraria; Celgene: Other: Travel Support. Gattermann: Novartis: Honoraria; Takeda: Research Funding; Celgene: Honoraria. Germing: Jazz Pharmaceuticals: Honoraria; Celgene: Honoraria; Bristol-Myers Squibb: Honoraria, Other: advisory activity, Research Funding; Janssen: Honoraria; Novartis: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2021
    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. 134, No. Supplement_1 ( 2019-11-13), p. 2882-2882
    Abstract: Background: Patients (pts) with R/R aggressive large B cell NHL who fail first-line therapy with immunochemotherapy and are ineligible for high-dose chemotherapy and hematopoietic stem cell transplantation (HSCT) have a poor prognosis. Available treatment options include platinum/gemcitabine-based or bendamustine-based regimens in combination with rituximab, with or without radiotherapy, or clinical trials. However, long-term outcomes remain poor due to lack of a curative option. Liso-cel is an investigational, anti-CD19, defined composition, 4-1BB CAR T cell product administered at target doses of CD4+ and CD8+ CAR T cells. In the ongoing TRANSCEND NHL 001 study of liso-cel as third- or later-line treatment for pts with R/R large B cell NHL, preliminary data showed high overall response rates with a low incidence of grade ≥3 cytokine release syndrome (CRS) and neurological events (NEs) (Abramson et al, ASCO 2018). The open-label, phase 2 PILOT study is assessing the safety and efficacy of liso-cel as second-line therapy in TNE pts (NCT03483103). PILOT is the first study evaluating CAR T cell therapy focusing on this pt population. Methods: Eligible pts had R/R large B cell NHL (diffuse large B cell lymphoma [DLBCL], not otherwise specified [NOS] , de novo or transformed indolent NHL, high-grade lymphoma with MYC and BCL2 and/or BCL6 [double/triple-hit lymphoma], or follicular lymphoma (FL) grade 3B) and had received only 1 prior line of immunochemotherapy containing an anthracycline and a CD20-targeted agent (eg, R-CHOP). Pts had to be deemed ineligible for high-dose chemotherapy followed by HSCT by meeting at least 1 of the following TNE criteria while still fulfilling the criteria for CAR T cell therapy: age ≥70 years, ECOG PS of 2, and/or impaired pulmonary (DLCO ≤60% but SaO2 ≥92% on room air and CTCAE ≤1 dyspnea), cardiac (LVEF ≥40% and 〈 50%), renal (creatinine clearance 〉 30 and 〈 60 mL/min), or hepatic function (AST/ALT 〉 2 and ≤5 ×ULN). Liso-cel was administered at a target dose of 100×106 CAR+ T cells after lymphodepletion (LD) with fludarabine/cyclophosphamide for 3 days. Pts could be treated as outpatients at the investigator's discretion. Results: At data cutoff, 10 pts had been leukapheresed, and 9 pts had LD followed by liso-cel infusion; 1 pt is awaiting liso-cel treatment. Liso-cel was manufactured successfully in all pts. Five pts were infused and monitored as outpatients. Median age was 71 (range, 64-79) years; 5 pts were male. Histology included DLBCL NOS (n=7) and transformed FL (n=2); 2 pts had triple-hit, one of whom had transformed from FL. Five pts had relapsed from, and 4 pts had disease refractory to, prior therapy. Median SPD and LDH were 26.6 cm2 and 201 U/L, respectively. Four pts had high tumor burden with SPD ≥50 cm2 (n=4) and/or LDH ≥500 U/L (n=1). The median Hematopoietic Cell Transplantation Comorbidity Index (HCT-CI) score was 3 (range, 0-3). Six pts had 1 or more treatment-emergent adverse events (TEAEs) grade ≥3, which were primarily cytopenias. Three pts had prolonged grade ≥3 cytopenias at Day 29. Two pts had infections of any grade; no pts had grade ≥3 infections. No pts had CRS or NEs, and no pts received tocilizumab, corticosteroids, or vasopressors. There were no cases of macrophage activation syndrome, tumor lysis syndrome, infusion reactions, or grade 5 TEAEs. Among the 5 pts treated and monitored as outpatients, none were admitted to hospital for adverse events within the first 29 days post liso-cel infusion. All 9 pts achieved an objective response. Four pts achieved complete response; all are ongoing. Five pts achieved partial response (PR), with 2 PRs ongoing. Results were similar in inpatient vs outpatient pts. Median follow-up was 3.5 months. Median (range) time to peak CAR T cell expansion was 10 (7-21) days. Conclusions: These preliminary safety and efficacy data from the ongoing phase 2 PILOT study suggest that liso-cel can be successfully administered, including in the outpatient setting, as second-line therapy in pts with R/R aggressive B cell NHL who were ineligible for high-dose chemotherapy and HSCT by prespecified criteria. Updated safety and efficacy data with longer follow-up will be presented. Disclosures Sehgal: Kite/Gilead: Research Funding; Merck: Research Funding; Juno/Celgene: Research Funding. Pribble:Celgene/Juno: Employment. Wang:Celgene Corporation: Employment. Thorpe:Juno Therapeutics, a Celgene Company: Employment, Equity Ownership. Hildebrandt:Axim Biotechnologies: Equity Ownership; Abbvie: Equity Ownership; GW Pharmaceuticals: Equity Ownership; Endocyte: Equity Ownership; Clovis Oncology: Equity Ownership; Kite Pharma: Equity Ownership, Membership on an entity's Board of Directors or advisory committees, Other; CVS Health: Equity Ownership; Celgene: Equity Ownership; Axim Biotechnologies: Equity Ownership; Pharmacyclics: Research Funding; Sangamo: Equity Ownership; Cellectis: Equity Ownership; Bluebird Bio: Equity Ownership; Bristol-Myers-Squibb: Equity Ownership; crispr therapeutics: Equity Ownership; IDEXX laboratories: Equity Ownership; Johnson & Johnson: Equity Ownership; Pfizer: Equity Ownership, Membership on an entity's Board of Directors or advisory committees, Other: Travel; Procter & Gamble: Equity Ownership; Vertex: Equity Ownership; Scotts-Miracle: Equity Ownership; Takeda: Research Funding; Bayer: Equity Ownership; Astellas: Other: Travel; Kite Pharma: Equity Ownership, Membership on an entity's Board of Directors or advisory committees, Other: Travel; Novartis: Equity Ownership; Aetna: Equity Ownership; Juno Therapeutics: Equity Ownership; Cardinal Health: Equity Ownership; Novartis: Equity Ownership; Insys Therapeutics: Equity Ownership; Incyte: Membership on an entity's Board of Directors or advisory committees, Other: Travel; Jazz Pharmaceuticals: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, Research Funding; Immunomedics: Equity Ownership.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    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. 112, No. 11 ( 2008-11-16), p. 3668-3668
    Abstract: The cancer-testis (CT) class of tumor antigens is a group of proteins, the expression of which is characteristically restricted to cancer and the human germline. Based on their immunogenicity and restricted tissue expression, CT antigens seem ideal targets for immunotherapeutic approaches. This is particularly the case in multiple myeloma (MM) where these tumor antigens are frequently and strongly expressed. Unfortunately, very little is known about the biological function of CT antigens which seems surprising given the strong impact of CT antigen expression on the prognosis of a variety of malignancies including MM. We examined for the first time the impact of the expression of two CT antigens frequently found in MM, MAGE-C1/CT7 and MAGE-A3, on the biology of the disease conducting knock-down experiments using RNA interference (RNAi) technology. As read-out assays, Western Blots as well as a large variety of tests measuring proliferation, cytotoxicity, cell adhesion, and clonogenic growth were performed. Transfecting myeloma cell line MOLP-8 with RNAi specific for MAGE-C1/CT7 and MAGE-A3 we observed a down-regulation of CT antigen protein expression by 80% and 70%, respectively. Importantly, transfection with MAGE-A3 RNA stealth led to a specific decrease in MAGE-A3 expression while the protein expression of other MAGE genes (MAGE-A4, MAGE-C1/CT7, MAGE-C2/CT10) and non-MAGE CT antigens such as Ropporin-1 was not affected. The same was true for knock-down experiments targeting MAGE-C1/CT7 with the exception of a slight decrease in expression of the highly homologous gene MAGE-C2/CT10. Comparing myeloma cell lines treated with gene-specific versus nonsense RNA stealth, we found that knocking down MAGE-C1/CT7 and MAGE-A3 led to the induction of apoptosis in in about 70% of cells at 72h post transfection as indicated by MTT assays, LDH release assays and Annexin V expression analyzed by flow cytometry. No such effect was observed when transfection was performed with nonsense RNAi (p 〈 0.01). Importantly, CD138-negative myeloma precursors, which also expressed the CT antigens investigated, were also affected by downregulation of CT antigen expression as indicated by an 48% decrease in clonogenic growth (p 〈 0.01). Applying chemotherapy with melphalan to the cell cultures we observed that knock-down of MAGE-C1/CT7 or MAGE-A3 increased the chemosensitivity of MM cell lines by 67% (p 〈 0.01). Importantly, knock-down of MAGE-C1/CT7 or MAGE-A3 specifically affected cell survival and did not alter the proliferative capacity of myeloma cells or cell adhesion. In conclusion, we show here for the first time that CT antigens MAGE-C1/CT7 and MAGE-A3, which are also expressed in the majority of patient myeloma samples, are important for the survival of myeloma cell lines and clonogenic myeloma precursors. In addition, the expression of these CT antigens most likely contributes to promoting resistance to chemotherapy in multiple myeloma. Targeting CT antigens expressed by myeloma cells, either by applying CT antigen-specifric immunotherapy or other targeted therapies, might significantly improve myeloma therapy, i.e. by eliminating chemotherapy-resistant clones.
    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 ...
  • 8
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 252-252
    Abstract: In the IPSS the variables bone marrow blasts, cytogenetics and cytopenias were found to be most relevant for clinical outcome by multivariate analysis. Comparing cytogenetics and blasts scoring points were assigned as follows: 0 (good cytogenetics and less than 5% blasts), 0.5 points (intermediate cytogenetics, and 5–10% blasts), 1.0 points (poor cytogenetics), 1.5 points (11–20% blasts), 2.0 (21–30% blasts). In order to examine the correctness of weighting of cytogenetics in comparison to blast counts we compared the survival curves, median survival times (mst) and differences of mst (mst diff.) related to the mst of 37.5 months (mo) of our entire study population (on the basis of 2124 pts. with MDS from our German-Austrian database) between patient subgroups. The results in the subgroups were as follows: Blasts below 5% (n=609) (mst: 58 mo, mst diff.: +20.5 mo), good cytogenetics (n=768) (mst: 55.3 mo, mst diff.: +17.8 mo), blasts 5–10% (n=231) (mst: 28.0 mo, mst. diff.: −9.5 mo), intermediate cytogenetics (n=222) (mst: 28.0 mo, mst diff.: −9.5 mo), blasts: 11–20% (n=160) (mst: 16.5 mo, mst diff.: −21 mo), poor cytogenetics (n=212) (mst: 11.1 mo, mst diff.: −26.3 mo), blasts 21–30% (n=92) (mst: 11.7, mst diff.: −25.7 mo). Our results clearly show that within the IPSS poor cytogenetics are significantly underweighed. Referring to the survival data unfavorable cytogenetics resulted in a survival disadvantage at the same scale as compared to 21–30% blasts. Thus, in a revised IPSS this cytogenetic feature should get the same scoring points as compared to 21–30% blasts when using the FAB-classification. For a scoring system based on the WHO-classification unfavorable cytogenetics should get an even higher scoring value as compared to the maximum blast count of 19%. Further statistical analyses are on the way to substantiate our conclusions.
    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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 4056-4056
    Abstract: Abstract 4056 Background: Multiple myeloma (MM) is a malignancy characterized by the expansion of a plasma cell (PC) clone that localizes to the bone marrow (BM). Myeloma cells and BM stromal cells both produce soluble factors promoting the survival and progression of MM. Interleukin-(IL)-16 is involved in regulating migration and proliferation of normal leukocytes, however, it has been unclear whether IL-16 also plays a role in the pathophysiology of human cancers. Methods: Using an antibody array we screened supernatants of myeloma cell lines for the presence of a variety of cytokines/chemokines. We confirmed IL-16 expression in myeloma cell lines as well as in malignant PC and BM plasma from MM patients applying real-time PCR, western blots, ELISA, and flow cytometry. We applied inhibitory RNA to analyze IL-16 function and we used anti-IL-16 antibodies to evaluate possible therapeutic options for MM. Results: We found IL-16 to be strongly overexpressed in the BM of myeloma patients. Myeloma cell lines as well as primary tumor cells from MM patients constitutively expressed IL-16 RNA and protein and spontaneously secreted soluble IL-16. Functional analyses revealed that IL-16 supports the proliferation of myeloma cells. Accordingly, silencing of IL-16 expression had an anti-proliferative effect on the tumor cells. Most importantly, the application of a monoclonal antibody directed against IL-16had a strong growth-inhibiting influence on myeloma cells. Conclusions: These findings suggest that cytokine IL-16 is an important growth-promoting factor in MM and might represent a novel diagnostic and therapeutic target for this incurable human malignancy. 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: 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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 2373-2373
    Abstract: Abstract 2373 NK-cell alloreactivity determined by donor killer-cell immunoglobulin-like receptors (KIRs) has been shown to be predictive for relapse-free survival in myeloid leukemia after allogeneic stem cell transplantation. The role role of NK-cell allo reactivity in patients with multiple myeloma is unclear. Although the ligands for activating KIRs remain elusive, inhibitory KIRs on donor derived NK-cells and HLA class I molecules on the recipient cell determine NK cell alloreactivity. KIR genes on chromosome 19 segregate independently from HLA genes KIR haplotypes can be distinguished in two groups: group A has a fixed number of inhibitory receptors and only one activating receptor (KIR2DS4), while group B haplotype includes more activating receptor genes. Therefore individuals can be categorised as having A-KIR haplotype (A/A), or B-KIR haplotype which contains either A/B heterocygotes or B/B homocygotes KIRs. To analyse the effect of donor KIR-haplotype on outcome in multiple myeloma, we analysed 118 patients with a median age of 51 years (r: 29 – 68) who underwent allogeneic stem cell transplantation. Gender of the study population was predominantly male (n = 75, female: n = 33). Stem cell source was mainly peripheral blood stem cells (n = 105) and derived from unrelated donors (n = 81) or HLA-identical sibling (n = 37). 46 patients received graft from HLA-mismatched donor. The majority of the patients received a melphalan (100 – 140 mg/m2)/fludarabine-based reduced conditioning regimen (N = 106), either as part of an auto-allo-tandem protocol or as an allogeneic salvage protocol after relapse to an autograft. 46 patients had experienced relapse to an autograft prior to allogeneic stem cell transplantation. 79 patients were transplanted from KIR haplotype Bx and 39 from KIR haplotype A. Both groups were well balanced regarding age, HLA match, stem cell source, CMV seropositivity, remission status prior to transplantation, del 13q14, and relapse to prior autograft. After a median follow-up of 5 years the 5-year estimated disease-free and overall survival was better for KIR haplotype B donors in comparison to KIR haplotype A donors (30 % vs. 14 %, p = 0.008, and 49 % vs. 36 %, p = 0.07). The disease-free survival benefit for KIR haplotype B was mainly seen in HLA-matched patients (4y DFS: 39 % vs. 18 %, p = 0.005), while in HLA mismatch transplantation no difference according to KIR haplotype could be detected (26 % vs. 18 %, p = 0.5).The difference in survival in the HLA-matched group was due to higher risk of relapse for KIR haplotype A in the HLA-matched group (cumulative incidence at 1 year 46 % vs. 17 %, p = 0.005). In a multivariate Cox model KIR haplotype A remained a significant factor for worse disease-free (HR: 1.82, p = 0.008) and overall survival (HR: 1.69, p = 0.042). Other significant factors in the multivariate analysis were female donor sex (HR: 1.67, p = 0.04) and chemo-refractory disease at transplantation (HR: 1.76, p = 0.03) for OS and factors for DFS were unrelated donor (HR: 1.83, p = 0.02) and patient's age as continous variable (HR: 1.03, p =0.008) and donor female sex (HR: 1.62, p = 0.03). We conclude that female donor sex and KIR haplotype A are important risk factors for outcome of allogeneic SCT in multiple myeloma and should be considered for donor selection. 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: 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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages