GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: The Lancet Child & Adolescent Health, Elsevier BV, Vol. 6, No. 6 ( 2022-06), p. 367-383
    Type of Medium: Online Resource
    ISSN: 2352-4642
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2022
    detail.hit.zdb_id: 2900829-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 89, No. 12 ( 1997-06-15), p. 4531-4536
    Abstract: The immune-mediated graft-versus-leukemia effect is important to prevent relapse after allogeneic progenitor cell transplantation. This process requires engraftment of donor immuno-competent cells. The objective of this study was to assess the feasibility of achieving engraftment of allogeneic peripheral blood or bone marrow progenitor cell after purine analog containing nonmyeloablative chemotherapy. Patients with advanced leukemia or myelodysplastic syndromes (MDS) who were not candidates for a conventional myeloablative therapy because of older age or organ dysfunction were eligible. All patients had an HLA-identical or one-antigen–mismatched related donor. Fifteen patients were treated (13 with acute myeloid leukemia and 2 with MDS). The median age was 59 years (range, 27 to 71 years). Twelve patients were either refractory to therapy or beyond first relapse. Eight patients received fludarabine at 30 mg/m2/d for 4 days with idarubicin at 12 mg/m2/d for 3 days and ara-c at 2 g/m2/d for 4 days (n = 7) or melphalan at 140 mg/m2/d (n = 1). Seven patients received 2-chloro-deoxyadenosine at 12 mg/m2/d for 5 days and ara-C 1 at g/m2/d for 5 days. Thirteen patients received allogeneic peripheral blood stem cells and 1 received bone marrow after chemotherapy. Graft-versus-host disease (GVHD) prophylaxis consisted of cyclosporine and methyl-prednisolone. Treatment was generally well tolerated, with only 1 death from multiorgan failure before receiving stem cells. Thirteen patients achieved a neutrophil count of greater than 0.5 × 109/L a median of 10 days postinfusion (range, 8 to 17 days). Ten patients achieved platelet counts of 20 × 109/L a median of 13 days after progenitor cell infusion (range, 7 to 78 days). Eight patients achieved complete remissions (bone marrow blasts were 〈 5% with neutrophil recovery and platelet transfusion independence) that lasted a median of 60 days posttransplantation (range, 34 to 170+ days). Acute GVHD grade ≥2 occurred in 3 patients. Chimerism analysis of bone marrow cells in 6 of 8 patients achieving remission showed ≥90% donor cells between 14 and 30 days postinfusion, and 3 of 4 patients remaining in remission between 60 and 90 days continued to have ≥80% donor cells. We conclude that purine analog-containing nonmyeloablative regimens allow engraftment of HLA-compatible hematopoietic progenitor cells. This approach permits us to explore the graft-versus-leukemia effect without the toxicity of myeloablative therapy and warrants further study in patients with leukemia who are ineligible for conventional transplantation with myeloablative regimens either because of age or concurrent medical conditions.
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 1997
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 104, No. 11 ( 2004-11-16), p. 885-885
    Abstract: Background: IFI is the most frequent cause of mortality in pts with AML and MDS undergoing chemotherapy. None of the antifungal prophylactic regimens used since 1992 at MD Anderson Cancer Center appeared to be significantly superior in the prevention of IFI. Study Aims: To compare the efficacy and safety of IV-VORI versus IV-ITRA as antifungal prophylaxis in AML and MDS pts receiving chemotherapy Patients and Methods: Pts older than 18 years old receiving induction or salvage chemotherapy, without documentation of prior IFI were eligible. Pts were randomized on day 1 of chemotherapy to receive IV-VORI 400 mg q12 h x 2 days followed by 300 MG IV twice per day or IV-ITRA 200 mg q12 h x 2 days followed by 200 mg IV once per day. Prophylaxis continue until recovery from neutropenia, developed possible/proven IFI, complete remission, declared resistant or up to 35 days for induction pts and up to 42 days for salvage pts. Results: 114 pts were evaluable (49 on IV-ITRA, 65 on IV-VORI). Baseline characteristics were similar in both groups. 102 were induction pts and 12 were on first salvage. Median time on prophylaxis was 21 (induction) and 17 days (salvage) for both groups. 45% pts on IV-ITRA and 48% pts on IV-VORI completed prophylaxis without modification (p=ns). Two pts on IV-ITRA developed IFI (1 disseminated C. glabrata and 1 disseminated Fusarium) as oppose to none on the IV-VORI arm (p=0.101). No significant differences were seen in the number of pts that required empiric antifungal therapy due to persistent fever or possible fungal pneumonia (14% on IV-VORI, 18% on IV-ITRA). 15/49 pts on IV-VORI (23%) versus 4/49 (8%) on IV-ITRA discontinued prophylaxis due to side effects (p=0.036). Reversible increase in the liver function tests (9 on IV-VORI, 4 on IV-ITRA) and hallucinations (5 on IV-VORI) were the most frequent adverse events. Response to induction chemotherapy, overall induction mortality and survival were similar in both groups. Conclusions: 1) IV-VORI appears to be more efficacious than IV-ITRA in preventing IFI in AML and MDS pts receiving chemotherapy. More pts are needed to confirm this finding. 2) IV-VORI tends to be more toxic than IV-ITRA. The usage of high dose IV-VORI may explain the incidence of side effects.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2004
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Society of Hematology ; 1997
    In:  Blood Vol. 90, No. 7 ( 1997-10-01), p. 2843-2845
    In: Blood, American Society of Hematology, Vol. 90, No. 7 ( 1997-10-01), p. 2843-2845
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 1997
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 3235-3235
    Abstract: Background: Chronic myelomonocytice leukemia is characterized by peripheral blood/bone marrow monocytosis and variable degree of dysplasia. The World Health Organization classification includes it in a myelodysplastic/myeloproliferative disorder category. We have reported earlier on a predictive model (MDAPS) based on hemoglobin, presence of circulating immature cells, absolute lymphocyte count and blast percentage and this has been validated in prospective group (Beran,M. et al. Leuk Lymphoma.2007;48:1150). In recent times hypomethylating agents are reported to have activity in this disease. Five year estimated survival with stem cell transplant is in the 20% range. Patients and Method: We performed an analysis of 279 patients seen at MD Anderson Cancer Center between the years 1999–2007. We also included 26 (9%) patients who underwent SCT in the analysis. Patients were assigned a score according to the International Prognostic Scoring System (IPSS) used for patients with myelodysplastic syndrome. Cox proportional hazard regression models were used for each variable. Log transformations were used for WBC, platelet counts, lactate dehydrogenase levels and absolute monocyte counts. Stepwise model selection was employed to fit a multivariable model. Results: Median age of patients is 68 years (range, 30–89), WBC count 14.5 x106/ml (range, 1.7–99), hemoglobin 10.3 gm/dL (range, 5.9–16.4), platelet count 98 x106/ml, beta 2 micrglobulin 4.1 mg/L (range, 1.2–20), absolute monocyte count 2.9 x106/ml (range, 1–40.2). Median survival is 15.7 months [95% confidence interval (CI); 13.9–18.8]. In univariate analysis patients undergoing stem cell transplantation survived longer than the others (hazards ratio 0.59, 95% CI 0.3–0.9) (Fig.1). Survivals at 2 and 5 years were 40.5% (95% CI, 0.22–0.72) and 24.3 (95% CI, 0.1–0.6) respectively in patients undergoing transplant versus 34.3% (95% CI, 0.27–0.42) and 8.9% (95% CI, 0.05–0.17) for others. Additionally WBC, creatinine, LDH, absolute monocyte count, hemoglobin and platelet count were laboratory parameters significantly associated with survival (Table 1). Splenomegaly, higher IPSS score (1.5 and above) and poor-risk cytogenetics (−5 and or −7, 8 abnormality) predicted for worse survival. Multivariate analysis indicated that lower hemoglobin level and platelet count and higher absolute monocyte count predicted for worse survival. Further analysis including treatment as a variable is in progress. Conclusion: Survival of patients with CMML has not improved despite availability of agents with activity in this disease. SCT may improve outcome but larger number of patients is needed for definitive answer. Table 1. Univariate analysis Variable HR(95%CI) p-value Age 1.01 (1.00–1.03) 0.10 WBC 1.44 (1.22–1.70) 〈 .0001 Hemoglobin 0.84 (0.78–0.91) 〈 .0001 Platelet 0.80 (0.70–0.92) 0.001 Creatinine 1.17 (1.06–1.28) 0.001 LDH 1.35 (1.04–1.77) 0.03 Absolute Monocyte count 1.47 (1.24–1.74) 〈 .0001 Bone marrow blast percentage 1.04 (1.01–1.07) 0.01 Normal spleen size 0.69 (0.49–0.96) 0.03 IPSS (1.5 or higher) 2.23 (1.35–3.67) 0.002 SCT 0.53 (0.30–0.93) 0.03 Cytogenetics (−5, −5 and −7, −7,+8) 2.09 (1.43–3.05) 0.0001 Fig. 1: Survival analysis according to stem cell transplant status Fig. 1:. Survival analysis according to stem cell transplant status
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 104, No. 11 ( 2004-11-16), p. 3003-3003
    Abstract: AMegL is a rare form of acute myeloid leukemia (AML) associated with poor prognostic features. Here, we report the experience at M.D. Anderson Cancer Center from 1987 to 2003. Among 1861 pts with newly diagnosed AML (excluding APL), 38 pts (2%) had AMegL. The diagnosis of AMegL was based on (i) morphological features defined by FAB classification and (ii) immunophenotypypical positivity for CD42 and CD61 whenever available. All 38 pts received induction chemotherapy containing cytarabine and anthracyclines. Sixteen (42%) were males; median age was 54 years (range 21–78); 11 (29%) had antecedent hematologic disorder (AHD) and/or MDS, and 4 (11%) had previously received chemotherapy for other malignancies. Median white blood cell count at diagnosis was 3.5x109/L (0.5–49.9), hemoglobin 7.8 g/dl (3.2–10.9), platelet count 34.5x109/L (5–2292), and the median bone marrow blast percentage was 30% (0–80). For pts with 〈 30% bone marrow blasts, diagnosis of AMegL was supported by peripheral blood blasts ≥30%. Significant bone marrow fibrosis was found in 24 pts (63%). Cytogenetic analysis was available in 30 pts (79%) due to insufficient material or insufficient metaphases. Twenty-seven pts (71%) showed abnormalities: deletion 7 in 15 pts (39%), deletion 5 in 14 (37%), chromosome 3 abnormality in 6 (16%) and trisomy 8 in 4 (11%). Philadelphia chromosome was detected in 2 pts (5%). Sixteen pts (42%) achieved a complete remission (CR). None of 22 pts with primary refractory disease achieved a complete remission from subsequent chemotherapies. Median overall survival of all AMegL pts (n=38) was 24 compared to 38 weeks for the control group (non AmegL AML pts, excluding APL,N=1823) (p 〈 0.01). Median disease free survival of pts with AMegL (n=16) and other AML (n=1031) was 23 and 52 weeks, respectively (p 〈 0.001). Two pts with primary refractory disease and three who developed relapsed disease eventually underwent allogeneic stem cell transplantation. Median survival after stem cell transplantation was 21 weeks. In summary, AMegL is a rare form of AML, which is observed in pts with AHD and/or MDS or as secondary leukemia. AMegL is highly associated with poor cytogenetic abnormalities, translating into a low CR rate and dismal outcome.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2004
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 104, No. 11 ( 2004-11-16), p. 2738-2738
    Abstract: The intensive combination chemotherapy program hyper-CVAD in newly diagnosed Ph+ ALL yields complete remission (CR) rates of 90%, but remissions are brief with median CR duration of 16 months [Kantarjian et al, JCO 18:547, 2000]. The activity of the tyrosine kinase inhibitor imatinib given as a single agent in relapsed or refractory Ph+ ALL or chronic myelogenous leukemia in lymphoid blast phase was 20% [Druker et al, NEJM 344:1084, 2001] . A phase II trial of imatinib and hyper-CVAD was conducted in newly diagnosed Ph+ ALL. Imatinib was given 400 mg days 1–14 of each course of therapy (fractionated cyclophosphamide, vincristine, doxorubicin and dexamethasone alternating with high dose methotrexate and ara-C). Preliminary results were published in the first 20 patients (pts) treated, with CR rates of 100% in the de novo group, and improvement of disease-free survival (DFS) compared with hyper-CVAD alone [Thomas et al, Blood 103:4396, 2004]. To date, 32 pts with Ph+ ALL have been treated from April 2001 to February 2004. Twenty-six patients had active disease, either untreated (n=21) or refractory (n=5) to one induction course without imatinib. Six pts were in CR at study entry after one induction course without imatinib mesylate. Median a ge was 48 years (range, 17–75); 59% were male. Five had CNS disease (16%). Twenty-five of 26 pts (96%) with active disease at study entry achieved CR (1 failed to meet platelet criteria for CR). Median days to response was 21 days. Two of 26 pts (8%) required 2 courses to achieve CR. Allogeneic stem cell transplant (SCT) was performed in 13 pts in CR within a median of 3 months from start of therapy (range, 1–12). After a median follow-up of 2 years (range, 4–36 months), 1 primary refractory pt relapsed at 12 mos (bcr-abl/abl RT-PCR ratio 〈 .05 at 9 mos), 1 pt relapsed day 149 after matched related SCT despite negative nested PCR for bcr-abl, and 2 pts changed therapy after 5 mos for persistent marrow Ph+ metaphases without overt leukemia relapse. Five pts died in CR, 3 older pts related to comorbid conditions (2 were negative for bcr-abl by nested PCR) and 2 related to complications of allogeneic SCT. Molecular response rate (negative bone marrow RT-PCR for bcr-abl confirmed by nested PCR) was approximately 50% in 19 pts who did not undergo allogeneic SCT. Outcome appears better with the hyper-CVAD and imatinib regimen with 2-year DFS rates of 87% (all pts) compared with 28% for hyper-CVAD alone or 12% for VAD (p 〈 .001). Unexpected toxicities related to the addition of imatinib mesylate were not observed. The hyper-CVAD and imatinib regimen with or without allogeneic SCT continues to appear promising with additional accrual and longer follow-up.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2004
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 107, No. 4 ( 2006-02-15), p. 1555-1563
    Abstract: KBM5 cells, derived from a patient with blast crisis Philadelphia chromosome-positive (Ph+) chronic myelogenous leukemia (CML), and imatinib-resistant KBM5 (KBM5-STI571) cells were found to express high levels of survivin. Inhibition of Bcr-Abl by imatinib significantly decreased survivin expression and cell viability in KBM5, but much less so in KBM5-STI571 cells. Inhibition of MEK, downstream of the Bcr-Abl signaling cascade decreased survivin expression and cell viability in both KBM5 and KBM5-STI571 cells. In addition, down-regulation of survivin by a survivin antisense oligonucleotide (Sur-AS-ODN) inhibited cell growth and induced maximal G2M block at 48 hours, whereas cell death was observed only at 72 hours in both KBM5 and KBM5-STI571 cells as shown by annexin V staining. Further, the combination of Sur-AS-ODN and imatinib induced more cell death in KBM5 cells than did either treatment alone. Down-regulating survivin also decreased colony-forming units (CFUs) in blast crisis CML patient samples. Our data therefore suggest that survivin is regulated by the Bcr-Abl/MAPK cascade in Ph+ CML. The facts that down-regulating survivin expression induced cell-growth arrest and subsequent cell death regardless of the cell response to imatinib and enhanced the sensitivity to imatinib suggest the potential therapeutic utility of this strategy in patients with CML, both imatinib sensitive and resistant.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 100, No. 3 ( 2002-08-01), p. 791-798
    Abstract: Myelodysplastic syndrome (MDS) is a disease characterized by ineffective hematopoiesis. There are significant biologic and clinical differences between MDS and acute myeloid leukemia (AML). We studied a cohort of 802 patients, 279 (35%) with newly diagnosed MDS and 523 (65%) with newly diagnosed AML, and compared clinical and biologic characteristics of the 2 groups. Complete clinical and cytogenetic data were available on all patients, and a subgroup of patients was studied for apoptosis, angiogenesis, proliferation, and growth factors. Our results demonstrate that MDS is a discrete entity that is different from AML and is characterized primarily by increased apoptosis in early and mature hematopoietic cells. Using cell sorting and loss of heterozygosity, we demonstrate that the leukemic cells from MDS patients are capable of differentiation into mature myeloid cells and monocytes. We also demonstrate that there is a significant overlap between AML and MDS when MDS is defined on the basis of an arbitrary percentage of blasts of 20% or 30%. These data suggest that despite similarities between AML and MDS in their responses to treatment and outcomes, MDS is biologically and clinically different from AML and should not be considered an early phase of AML. The data indicate that MDS must be better defined on the basis of its biology rather than the percentage of blasts; further, the data suggest that there is a need to develop therapeutic approaches that specifically address the biologic abnormalities of MDS.
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2002
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 3797-3797
    Abstract: Abstract 3797 Background: Since the time of initial proposal of the MD Anderson Prognostic Score (MDAPS) in 2000, there has been substantial development in diagnosis and treatment for patients (pts) with CMML. MDAPS did not incorporate cytogenetic abnormalities, which is one of the most important factors of prognostication in other myeloid malignancies. Therefore, we analyzed a large cohort of patients with CMML and developed new prognostic scoring system that also incorporates cytogenetic abnormalities (named MDAPS-R). Methods: From 2003 and 2012, we identified 358 pts with diagnosis of CMML, using standards strictly defined by World Health Organization (WHO) criteria. Potential prognostic factors were identified by log-rank test. Of those, independent prognostic factors were extracted after Cox proportional hazard regression. Based on the relative strength of hazard ratio (HR), MDAPS-R was developed and was verified by log-rank test. Result: Median age of the analyzed group was 68 years (range:23–89);113 (32%) pts were female. Two hundred twenty one (62%) pts were classified as CMML-1 and 104 (29%) were CMML-2 (unknown in 33 pts). Thirty nine (11%) pts had prior exposure to chemotherapy and/or radiation therapy. Mean (± SE) white blood cell count (WBC) was 24.5 ± 1.5 (x103/μL), hemoglobin (Hb) was 10.8 ± 0.1(g/dL), platelet count (Plt) was 132 ± 7.0 (x103/μL) and bone marrow blast count (BMBL) was 6.9 ± 0.3 (%), respectively. Cytogenetics was diploid in 224 (63%) pts. Trisomy 8 was detected in 14 (4%) pts, del 20q in 12 (3.4%), -Y in 13 (3.6%), del 7q/-7 in 25 (7%), and del 5q/-5 in 10 (2.8%) pts, respectively. Complex cytogenetic abnormality was detected in 16 (4.5%) pts. Two hundred eighty (78%) pts had RAS mutation analysis and 49 (18%) had NRAS mutation while 16 (5.7%) had KRAS mutation. FLT3 alteration was tested in 297 pts (83%):3 (1%) had D835 mutation while 10 (3.4%) had ITD. JAK2 mutation was tested in 161 (45%) pts of which 19 (12%) had V617F mutation. Less commonly occurring mutations included: NPM1 (5/88 tested), c-kit (3/156), CEBPA (6/83), IDH1 (1/59), IDH2 (3/58), and DNMT3a (1/4). During the median follow up duration of 15 months (range; 1–145), 53 (15%) pts transformed to acute leukemia and 182 (51%) pts died. Median transformation free survival (TFS) and overall survival (OS) of the analyzed group was 24.9 months (range; 1–145) and 26.8 months (range; 1–145), respectively. Log-rank test identified significant covariates in association with OS that include: BMBL ( 〈 10 vs. ≥10; P = 0.024), WBC (≤10 vs. 〉 10; P = 0.01), Hb ( 〈 12 vs. ≥12; P 〈 0.001), CMML subtype (CMML-1 vs. 2; P = 0.007), prior exposure to chemo and/or radiation (Yes vs. No; P 〈 0.001), cytogenetics (diploid vs. complex or del7q/-7 vs. others; P 〈 0.001), serum β2 microglobulin (β2MG) (≤4.0 vs 〉 4.0; P 〈 0.001), serum LDH (≤700 vs. 〉 700; P 〈 0.001), peripheral absolute lymphocyte count (ALC) (≤2.5 vs. 〉 2.5; P 〈 0.001), and peripheral absolute monocyte count (≤4.0 vs. 〉 4.0; P = 0.012). None of the molecular mutations had impact on OS. After being fitted into Cox proportional hazard regression, following covariates remained independently significant: BMBL ≥10 % (vs. 〈 10; HR = 1.6), Hb 〈 12 g/dL (vs. ≥12; HR = 1.9), LDH 〉 700 IU/L (vs. ≤700; HR = 1.5), ALC 〉 2.5 × 103/μL (vs. 〈 2.5; HR = 1.7), β2MG 〉 4.0 mg/L (vs. ≤ 4.0; HR = 1.6), and complex cytogenetics or del 7q/-7 (vs. diploid; HR = 2.3 and others vs. diploid; HR = 1.5). We developed MDAPS-R based on relative strength of HR in each of these above factors (1 point assigned to each of the following: BM BL '10 %, Hb 〈 12 g/dL, LDH 700 IU/L, ALC .2.5 × 103/μL, and β2MG 〉 4.0 mg/L; 0 points for diploid cytogenetics, 2 points for −7/del 7q or complex cytogenetics, and 1 point for all other abnormal karyotype). Among 358 pts, 282 (79%) were evaluable for analysis via MDAPS-R. MDAPS-R stratified pts into 4 distinct prognostic groups: score 0–1 = low risk (N = 70, median OS 56 months), 2–3 = intermediate-1 risk (N = 133, median OS 28 months), 4–5 = intermediate-2 risk (N = 68, median OS 18 months), and 6–7 = high risk (N = 11, median OS 7.5 months) (P 〈 0.001, Figure 1A). MDAPS-R also predicted TFS in the same cohort (median TFS: low = 54, int-1 = 26, int-2 = 15, and high = 7 months, P 〈 0.001, Figure 1B). Conclusion: We propose a refined version of MDAPS (MDAPS-R) specifically for pts with CMML that incorporates cytogenetic abnormalities. This model may help risk-stratified decision making in CMML pts. 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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...