Differential Prognostic Effect of IDH1 Versus IDH2 Mutations in Myelodysplastic Syndromes: A Mayo Clinic Study of 277 Patients

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 971-971
Author(s):  
Mrinal M. Patnaik ◽  
Curtis A. Hanson ◽  
Janice M Hodnefield ◽  
Terra L Lasho ◽  
Christy Finke ◽  
...  

Abstract Abstract 971 Unlike the case in acute myeloid leukemia, there is limited information on the prognostic impact of IDH mutations in myelodysplastic syndromes (MDS). In the current study of 277 patients with MDS, IDH mutations were detected in 34 (12%) cases: 26 IDH2 (all R140Q) and 8 IDH1 (six R132S and two R132C). Mutational frequency was 4% (2 of 56) in refractory anemia with ring sideroblasts (RARS), 12% (16 of 130) in refractory cytopenia with multilineage dysplasia, 14% (2 of 14) in MDS-unclassifiable, 14% (6 of 42) in refractory anemia with excess blasts (RAEB)-1, and 23% (8 of 35) in RAEB-2. Normal karyotype was noted in all but one IDH1-mutated cases and 13 IDH2-mutated cases. Multivariable analysis identified presence of mutant IDH1 (p=0.0004; HR 4.0, 95% CI 1.9–8.8), revised International Prognostic Scoring System (IPSS-R) risk category (p<0.0001), and red cell transfusion need (p=0.002) as independent predictors of inferior survival (Figure 1). In a similar multivariable analysis, mutant IDH1 was the only variable associated with shortened leukemia-free survival (p=0.001; HR 7.0, 95% CI 2.3–20.8). The presence of IDH2 R140Q did not affect overall (p=0.54) or leukemia-free (p=0.81) survival (Figure 2). The current study suggests a powerful adverse prognostic effect for mutant IDH1 in MDS. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2882-2882
Author(s):  
Naseema Gangat ◽  
Terra L. Lasho ◽  
Mrinal M Patnaik ◽  
Christy Finke ◽  
Mark R Litzow ◽  
...  

Abstract Background: Addition of sex combs-like 1 (ASXL1) is frequently mutated (mutational frequency 14-29%) and is prognostically relevant in myelodysplastic syndromes (MDS) (Bejar, NEJM 2011, Thol, JCO 2011). The prevalence and prognostic impact of ASXL1 mutation in MDS with ring sideroblasts (MDS-RS) is not known. MDS-RS is defined by the presence of ≥15% RS in bone marrow (BM), with refractory anemia with ring sideroblasts (RARS) being the prototype but may also be seen in other categories. Methods : Our institutional database was reviewed to identify patients with WHO-defined primary MDS with ≥1% BM RS. Pathology slides, including iron stains, were reviewed to accurately quantify BM RS percentage and confirm WHO morphologic categories. All patients were annotated for their mutational status including ASXL1, JAK2, MPL and IDH with a subset for SF3B1 by PCR sequencing performed on BM specimens obtained at diagnosis. Results : i) Patient characteristics: A total of 76 MDS patients displayed ≥1% BM RS (median age 72 years; 76% males); 51 (67%) patients had ≥15% BM RS. IPSS-R risk categories for the entire 76 study patients were 30% very low, 37% low, 14% intermediate, 11% high and 8% very high; IPSS-R karyotype was normal in 63%, very good/good risk 17%, intermediate risk 12%, and poor/very poor in 8%; 3% had monosomal karyotype. ii) Prevalence of ASXL1 mutations: Twenty-one (28%) of the 76 study patients were ASXL1 mutated; ASXL1 mutational frequencies were 25% (16/63 patients) in the absence and 38% (5/13 patients) in the presence of excess blasts (P =0.34). When considering only those patients with ≥15% BM RS (n =51), ASXL1 mutations were detected in 12 (24%) patients with mutational frequencies of 24% (11/46 patients) in the absence and 20% (1/5 patients) in the presence of excess blasts (P =0.84); ASXL1 mutational frequencies were 13% (3/23 patients) in RARS and 37% (7/19 patients) in RCMD-RS (P=0.07). In terms of other mutations, all 76 study patients were wild-type for JAK2 and MPL, whereas IDH2 R140Q mutations were present in 4 (5%) patients, including 3 with concomitant ASXL1 and IDH2 mutations. IDH1 mutation was seen in only 1 patient. 25 of 43 (58%) patients screened were mutated for SF3B1, including 4 (9%) who were mutated for both ASXL1 and SF3B1. Significant associations were evident between ASXL1 and IDH2 mutations (P =0.02) but not between ASXL1 and SF3B1 mutations (P =0.61). iii) Clinical correlates of ASXL1 mutations: Among all 76 study patients, presence of ASXL1 mutation did not correlate with age (P =0.30), hemoglobin level (P =0.17), platelet count (P =0.53), BM blast percentage (P =0.17), WHO morphologic category (P =0.34), transfusion dependence (P =0.84), IPSS-R cytogenetic categories (P =0.93), or IPSS-R risk group (P =0.33). The results were unchanged when analyzing the 51 patients with MDS-RS (i.e. ≥15% BM RS). Amongst the ASXL1 mutated patients (n =21) IPSS-R cytogenetic categories were as follows: 13 patients with normal karyotype (62%), 2 patients with trisomy 8 (10%), 1 patient each with -Y, del(11q), del(5q), del(20q), isochromosome 17 and monosomy 7 (5% each). iv) Prognostic impact of ASXL1 mutations: Median follow-up was 42.5 months, during which time 69 (91%) deaths and 8 (11%) leukemic transformations were documented. ASXL1 mutated patients had a median survival of 29 months, compared to 45 months in ASXL1 wild-type patients (P =0.04). However, the difference in median survival was no longer significant during multivariable analysis, which instead identified only IPSS-R and transfusion need as being independent predictors of inferior survival. Amongst those patients without excess blasts (n =63), presence of ASXL1 mutation predicted inferior survival in univariate analysis (P =.04) and significance was sustained during multivariable analysis that included IPSS-R cytogenetic categories (P =.04) but became borderline when WHO morphologic category was included (P =0.06); ASXL1 mutated patients had a shortened median survival of 43 months compared to 66 months in ASXL1 wild-type patients (P =.04). In the presence of ≥15% BM RS, ASXL1 mutations did not affect survival (P =0.48); the results were the same for RARS (P =0.9) and RCMD-RS (P =0.64). Conclusions : ASXL1 mutations might not affect survival in MDS patients with ≥15% BM RS, including those with RARS or RCMD-RS. Furthermore, an apparent survival disadvantage seen in ASXL1 -mutated MDS patients with ≥1% BM RS was accounted for by IPSS-R. Disclosures Pardanani: Stemline: Research Funding.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4117-4117
Author(s):  
Toshio Asayama ◽  
Mariko Ishibashi ◽  
Hideto Tamura ◽  
Yasuko Hamada ◽  
Namiko Okuyama ◽  
...  

Abstract Background: Galectin-9 (Gal-9), a member of the galectin family, plays a crucial role in inflammation and tumorigenesis involving angiogenesis, cell adhesion, immune escape, and cancer cell survival. In acute myeloid leukemia (AML), Gal-9 is highly expressed in some AML cell lines and binds with T-cell immunoglobulin- and mucin-domain-containing molecule-3 (Tim-3) on T cells and leukemic stem cells. The Tim-3/Gal-9 pathway may be associated with disease progression by inducing T-cell dysregulation and mediating autocrine proliferation of leukemic cells. However, the role of the pathway in myelodysplastic syndromes (MDS) remains unclear. In the current study, we investigated the prognostic impact of plasma Gal-9 levels and the pathophysiological roles of Gal-9 and Tim-3 in MDS. Methods and results: 1) To investigate the prognostic impact of Gal-9, plasma Gal-9 levels were measured by ELISA in 92 MDS patients: 30 with refractory anemia (RA); 3 with RA with ringed sideroblasts (RARS); 18 with RA with excess blasts (RAEB); 14 with RAEB in transformation (RAEB-t); and 29 with acute leukemia transformed from MDS (AL-MDS). The plasma Gal-9 level increased with disease progression (mean ± SD: control 3.4 ± 0.9; RA, 9.3 ± 5.7; RARS, 10.2 ± 6.8; RAEB, 9.7 ± 6.5; RAEB-t, 17.2 ± 9.6; AML-MDS, 20.1 ± 14.1 ng/ml). Furthermore, patients with AL-MDS in International Prognostic Scoring System (IPSS) high-risk categories (Intermediate-2/High) had significantly higher Gal-9 levels compared with IPSS low-risk (Low/Intermediate-1) patients (Low/Intermediate-1 vs Intermediate-2/High, p = 0.002; Low/Intermediate-1 vs AML-MDS, p = 0.004). The overall survival of MDS patients with plasma Gal-9 higher than the normal level (10 ng/mL) was significantly shorter than that of other patients (log-rank p < 0.001) even in lower-risk (RA, RARS) cases (log-rank p = 0.0369). Moreover, multivariate analysis showed that a high level of Gal-9 was an independent predictor of shorter survival in MDS patients as well as IPSS. 2) Bone marrow mononuclear cells were isolated from some MDS patients, and Tim-3 expression on granulocytes, monocytes, lymphocytes, and blasts was analyzed using flow cytometry (FCM). Tim-3 expression on blasts tended to be higher in AL-MDS patients than in MDS patients. 3) To investigate whether MDS cells can produce Gal-9, we analyzed Gal-9 levels in supernatants of cell cultures of 3 human AL-MDS cell lines, F-36P, SKM-1, and MDS-L, and MDS blasts obtained from an AL-MDS patient by ELISA. Gal-9 was detected in all cell culture supernatants and its concentration was higher when cultured in medium containing 10% fetal bovine serum (FBS) than in 2.5% FBS medium in F-36P and SKM-1 cells. The concentration of Gal-9 produced from MDS blasts was increased time dependently in 2-6 day culture. Furthermore, recombinant human Gal-9 enhanced the cell proliferation of F-36P cells. 4) After analyzing Tim-3 expression in MDS cell lines using real-time quantitative PCR and FCM, all cell lines expressed relatively low levels of Tim-3 mRNA. Extracellular expression of Tim-3 was detected and induced by adding cell culture supernatants of the human stromal cell line HS-5 in F-36P cells. To elucidate the signaling pathway inducing Tim-3 expression, we investigated Tim-3 mRNA expression in F-36P cells treated with HS-5 supernatant with several signal transduction inhibitors (STAT3 inhibitor, U0126; MAPK/ERK; AG490; JAK2, LY294002; PI3K, PDTC; NF-κB inhibitor). Upregulation of Tim-3 gene expression in F-36P cells treated with HS-5 supernatant was inhibited by the MEK inhibitor U0126. These results suggest that the MAPK/ERK pathway is involved in Tim-3 expression induced by HS-5 supernatant. Conclusions: In MDS, the elevation of plasma Gal-9 levels is associated with disease progression, including leukemic transformation, and with shorter survival. Furthermore, its receptor Tim-3 is upregulated on blasts in AL-MDS patients. Our data suggest that the Gal-9/Tim-3 pathway plays a key role in MDS disease progression. Although further study is required to clarify the detailed functions of the interaction between Gal-9 and Tim-3, the current study could lead to the development of a new immunotherapeutic strategy via the blockade of this pathway in MDS. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 119 (24) ◽  
pp. 5674-5677 ◽  
Author(s):  
Mrinal M. Patnaik ◽  
Curtis A. Hanson ◽  
Nanna H. Sulai ◽  
Janice M. Hodnefield ◽  
Ryan A. Knudson ◽  
...  

Abstract The presence of ≥ 15% bone marrow (BM) ring sideroblasts (RS) and < 5% blasts is required for a diagnosis of refractory anemia with ring sideroblasts. We examined the phenotypic and prognostic relevance of this “15%” RS threshold in 200 patients with myelodysplastic syndromes (MDS) without excess blasts and with ≥ 1% RS. The impact of RS% was assessed both as a continuous and categorical variable: < 5% (n = 56), 5%-14% (n = 32), 15%-50% (n = 79), and > 50% (n = 33). RS% correlated (P < .05) directly with age, platelet count, transfusion dependency, BM cellularity, and mutant SF3B1 and inversely with hemoglobin level, multilineage dysplasia, and high-risk karyotype; but did not correlate with IDH mutations. At a median follow-up of 33 months, 156 (73%) deaths and 24 (12%) leukemic transformations were documented. Neither univariate nor multivariable analysis showed significant effect for RS% on overall or leukemia-free survival, suggesting the limited prognostic value of quantifying BM RS in MDS.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1756-1756
Author(s):  
Animesh Pardanani ◽  
Terra L Lasho ◽  
Christy Finke ◽  
S. Vincent Rajkumar ◽  
Preet Paul Singh ◽  
...  

Abstract Abstract 1756 Background: We hypothesized that surrogate markers of host immune response to clonal myeloproliferation predict survival in myeloid malignancies. Because of immediate applicability to current practice, we chose plasma immunoglobulin free light chain (FLC) concentration (B-cell activation marker) as the biomarker of interest. Patients and Methods: Two independent cohorts of patients with primary myelofibrosis (PMF) or myelodysplastic syndromes (MDS) were studied. Both groups were fully annotated for karyotype. Kappa (κ) and lambda (λ) FLC were measured by a quantitative nephelometric assay. Patients with monoclonal FLC were excluded. Results: Above upper normal-limit values for κ or λ FLC was documented in 33% of 240 study patients with PMF and 46% of 74 patients with MDS. Multivariable analysis revealed significant associations between increased FLC and elevated creatinine, as well as advanced age in PMF (p=0.0004) and hemoglobin <10 g/dL in MDS (p=0.005). Increased FLC predicted shortened survival in both PMF and MDS, independent of age, creatinine, and other conventional risk factors. Receiver-operating characteristic analysis-based cutoff levels for κ plus λ total FLC delineated risk groups with highly significant differences in overall survival (Figure); International Prognostic Scoring System-adjusted HR (95% CI) in PMF was 1.9 (1.3-2.7) and in MDS 6.3 (2.7-16.6). No correlations were seen with leukemia-free survival, karyotype or JAK2, MPL or IDH mutations. Conclusions: Elevated plasma FLC concentration predicts inferior survival in both PMF and MDS. Its lack of correlation with leukemia-free survival and tumor-specific genetic markers suggests a primarily host-driven biological phenomenon that might be applicable to other malignant and non-malignant conditions. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4060-4060
Author(s):  
Ayalew Tefferi ◽  
Christy Finke ◽  
Terra L Lasho ◽  
Emnet A Wassie ◽  
Ryan A Knudson ◽  
...  

Abstract Background Spliceosome pathway gene mutations are recurrent in myeloid malignancies with the highest frequencies reported for myelodysplastic syndromes associated with ring sideroblasts (MDS-RS; 85%), MDS without RS (44%) and chronic myelomonocytic leukemia (CMML; 55%) (Nature 2011;478:64). SF3B1 mutations were the most frequent (75%) in MDS-RS and SRSF2 mutations in CMML (28%); U2AF1 mutational frequencies were 12% in MDS without RS and 8% in CMML. We have previously described SRSF2 (Blood. 2012;120:4168) and SF3B1 (Leukemia. 2012;26:1135) mutations in 17% and 6.5% of patients with primary myelofibrosis (PMF); prognostic relevance was shown for the former but not the latter. Objectives The objectives of the current study were to i) describe the incidence of U2AF1 mutations in PMF and their correlation with clinical features, karyotype and other mutations and ii) examine the prognostic significance of U2AF1 mutations in PMF, in the context of both conventional prognostic models and other prognostically-relevant mutations. Methods Information on clinical and laboratory parameters and karyotype was available in all study patients, at time of referral, which coincided with time of sample collection for mutation screening. Risk stratification was according to the Dynamic International Prognostic Scoring System (DIPSS)-plus system. U2AF1 and other mutations were analyzed using standard PCR techniques and bidirectional sequencing; for U2AF1, two hot spots that included residues S34 and Q157 were amplified. Results A total of 251 PMF patients (median age 63 years; 160 males) were studied. DIPSS-plus risk distribution was high in 32%, intermediate-2 in 38%, intermediate-1 in 17% and low in 13% of the patients. The frequency of each DIPSS-plus adverse feature was as follows: age >65 years (42%), transfusion need (32%), hemoglobin <10 g/dL (47%), leukocyte count >25 x 10(9)/L, (16%), platelet count <100 x 10(9)/L (22%), ≥1% blasts (57%), constitutional symptoms (35%), and unfavorable karyotype (9%). Karyotype was normal in 156 (63%) patients. At a median follow-up of 48 months, 158 (63%) deaths and 27 (11%) leukemic transformations were recorded. Mutational frequencies Forty-one (16.3%) patients harbored U2AF1 mutations: 16 (39%) Q157P, 10 (24%) Q157R, 8 (20%) S34F, 4 (10%) S34Y and one each for Q157P/E159A, Q157R/S34Y and Q157-Y158insYE. Frequencies for other mutations were 11% for SRSF2, 7.3% for SF3B1, 31% for ASXL1, 5.5% for EZH2, 5% for IDH1/2 and 58% for JAK2V617F. U2AF1 mutations were usually, but not always, exclusive of other spliceosomal mutations: one patient expressed all three spliceosomal mutations. The frequency of any one of the three spliceosomal mutations was 34%. Clinical correlates U2AF1 mutations were significantly associated with older age (p=0.02), JAK2V617F (p=0.002), mutant ASXL1 (p=0.04), transfusion need (p<0.0001), hemoglobin <10 g/dL (p<0.0001), platelets <100 x 10(9)/L (p<0.0001) and normal karyotype (p=0.006). The associations with anemia, thrombocytopenia, JAK2V617F and normal karyotype were inter-independent; the frequency of U2AF1 mutations in the presence of anemia was 29%, transfusion need 36%, thrombocytopenia 35%, JAK2V617F 23% and normal karyotype 21%. Prognostic interactions U2AF1 mutations were associated with inferior overall (p=0.004) but not leukemia-free (p=0.6) survival. However, the survival association was fully accounted for by the above-mentioned clustering of U2AF1 mutations with anemia and thrombocytopenia. Similarly, although multivariable analysis of U2AF1, SRSF2, ASXL1, EZH2 and IDH mutations identified the first three as being independently predictive of poor survival, only ASXL1 and SRSF2 remained significant when either anemia or thrombocytopenia was included as a co-variate. Conclusions U2AF1 mutations are the most frequent spliceosome pathway mutations in PMF, cluster with normal karyotype and JAK2V617F, and are strongly associated with anemia and thrombocytopenia; the latter suggests a pathogenetic contribution to ineffective hematopoiesis in PMF. The current study also suggests that more than one third of patients with PMF carry a spliceosome mutation. Disclosures: No relevant conflicts of interest to declare.


2017 ◽  
Vol 138 (3) ◽  
pp. 143-151 ◽  
Author(s):  
Na Wang ◽  
Fei Wang ◽  
Ningning Shan ◽  
Xiaohui Sui ◽  
Hongzhi Xu

Background: Genomic sequencing technologies have identified isocitrate dehydrogenase (IDH) mutations in haematological malignancies. The prognostic implications of somatic IDH mutation (mIDH) in myelodysplastic syndromes (MDS) remain controversial. Methods: Mutations in IDH1 and IDH2 were detected using genomic sequencing technologies in 97 patients with MDS. Results: Seven (7.2%) mutations were identified: 3 in IDH1 (all R132C) and 4 in IDH2 (3 R140Q and 1 R140L). The frequency of mutation was 16.6% (2/12) in refractory anaemia with excess blasts (RAEB)-1 and 14.7% (5/34) in RAEB-2. IDH1/2 mutations were closely associated with higher bone marrow blast counts (median 10.0 vs. 2.3%; p = 0.019) and lower absolute neutrophil counts (median 0.44 × 109/L vs. 1.21 × 109/L; p = 0.027). All IDH mutations were mutually exclusive and heterozygous. IDH mutations were not significantly correlated with any specific karyotype. Patients with IDH1 mutations exhibited shorter overall and progression-free survival (OS and PFS; p = 0.039 and p = 0.042, respectively), whereas IDH2 mutations did not affect OS or PFS (p = 0.560 and p = 0.218, respectively). Multivariate analysis indicated that IDH1 mutation (p = 0.018; hazard ratio [HR] 4.735; 95% confidence interval [CI] 1.299-17.264), karyotype risk (p = 0.036; HR 1.619; 95% CI 1.033-2.539) and the revised International Prognostic Scoring System risk category (p < 0.0001; HR 2.122; 95% CI 1.401-3.213) were independent inferior prognostic factors. Conclusions:IDH1 mutation is associated with a poor prognosis.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 423-423
Author(s):  
Naseema Gangat ◽  
Mrinal M. Patnaik ◽  
Kebede Begna ◽  
Taxiarchis Kourelius ◽  
Ryan A Knudson ◽  
...  

Abstract Abstract 423 Background: Cytogenetic abnormalities at diagnosis are seen in 20–70% of patients with myelodysplastic syndromes (MDS). The International Prognostic Scoring System -revised (IPSS-R) classifies these cytogenetic abnormalities into five prognostic groups: very good (del(11q), ΠY with overall survival (OS) of 61 months); good (normal, del(20q), del(5q), del(12p) with OS 49 months); intermediate (+8,7q-, i(17q),+19,+21 with OS 26 months); poor (der(3)q21/q26, −7, complex with 3 abnormalities with OS 16 months); and very poor (complex with >3 abnormalities with OS of 6 months) (Schanz J, JCO 2012, Greenberg P, Blood 2012). Monosomal karyotype (MK) is defined as the presence of two or more autosomal monosomies or a single monosomy associated with a structural abnormality. We have previously shown that MK identifies a prognostically worse subgroup of MDS patients with a complex karyotype (Patnaik M, Leukemia 2010). Objectives: Methods: The Mayo Clinic database for MDS was used to identify patients in whom bone marrow histologic and cytogenetic information was obtained at the time of diagnosis. WHO criteria were used for MDS diagnosis and leukemic transformation. Results: i) Patient characteristics: A total of 1,014 patients met the above-stipulated criteria. Median age of the cohort was 71 years (range: 18–98 years) with 684 (67%) males. 819 patients (81%) had primary MDS with 195 cases (19%) of therapy-related MDS. 759 patients (75%) are dead at a median follow-up of 18 months (range: 0–223 months) with leukemic transformation documented in 125 cases (12%). IPSS-R risk distribution was as follows: very low (n=149, 15%), good (n=261, 26%), intermediate (n=221, 22%), high (n=131, 13%), and very high (n=252, 25%). ii) IPSS-R karyotype (Entire cohort; n=1,014): The five cytogenetic groups by IPSS-R were as follows: very good (n=43, 4%), good (n=577, 57%), intermediate (n=187, 18%), poor (n=83, 8%), and very poor (n=124, 12%). We evaluated the prognostic impact of the above groups by Kaplan Meier and Cox proportional hazards analysis. OS for the five cytogenetic groups was as follows: good (36 months), very good (28 months), intermediate (20 months), poor (13 months), and very poor (6 months) (Figure 1). OS of patients with very good karyotype was significantly better than patients with very poor and poor karyotype (p<0.0001, RR 3.2 and p=0.04, RR 1.6 respectively) but similar to patients with intermediate and good karyotype (p=0.22 and 0.28 respectively). iii) IPSS-R karyotype (Primary MDS; n=819): Cytogenetic risk groups by IPSS-R were as follows: very good (n=38, 5%), good (n=512, 63%), intermediate (n=153, 19%), poor (n=46, 6%), and very poor (n=70, 9%). OS of the five groups was as follows: good (39 months), very good (23 months), intermediate (21.5 months), poor (14.5 months), and very poor (6.5 months). OS of patients with very good karyotype was significantly better than patients with very poor karyotype (p=0.0002, RR 2.5) but similar to patients with poor, intermediate and good karyotype (p=0.23, 0.26 and 0.18 respectively). iv) Monosomal karyotype: MK was identified in 153 (15%) cases of which 4 cases were intermediate, 42 poor, and 107 very poor by IPSS-R. OS of patients with MK was similar to those with very poor karyotype (6 months). In addition, OS of patients with very poor without MK was akin to patients with poor without MK (p=0.21). Furthermore, among primary MDS we identified 83 (8%) cases of MK of which 4 were intermediate, 21 were poor, and 58 very poor by IPSS-R. We confirmed that OS of patients with MK was similar to patients with very poor karyotype (6.5 months). Moreover, survival of patients with very poor without MK was similar to patients with poor without MK (p=0.51). Conclusions: In this independent analysis of 1,014 patients with MDS, IPSS-R cytogenetic risk characterization failed to discriminate between very good, good and intermediate prognostic groups. In addition, patients with very poor without MK have a similar prognosis to patients with poor without MK and should be included in the poor category whereas the presence of MK should warrant inclusion into the very poor risk category. Disclosures: No relevant conflicts of interest to declare.


2006 ◽  
Vol 24 (16) ◽  
pp. 2465-2471 ◽  
Author(s):  
Norbert Vey ◽  
Andre Bosly ◽  
Agnes Guerci ◽  
Walter Feremans ◽  
Herve Dombret ◽  
...  

Purpose Evaluation of the safety and efficacy of arsenic trioxide in patients with myelodysplastic syndromes (MDS). Patients and Methods MDS patients diagnosed according to standard French-American-British criteria received a loading dose of 0.3 mg/kg per day of arsenic trioxide for 5 days followed by a maintenance dose of 0.25 mg/kg arsenic trioxide twice weekly for 15 weeks. Patients were divided into two cohorts: lower-risk MDS (International Prognostic Scoring System risk category low or intermediate 1) and higher-risk MDS (International Prognostic Scoring System risk category intermediate 2 or high). Modified International Working Group criteria were used for response evaluation. Results Of 115 patients enrolled and treated in the study, 67% of patients were transfusion dependent at baseline; median age was 68 years. Most treatment-related adverse events were mild to moderate. The overall rate of hematologic improvement (intent-to-treat) was 24 (19%) of 115, including one complete and one partial response in the higher-risk cohort. The hematologic response rates were 13 (26%) of 50 and 11 (17%) of 64 in patients with lower-risk and higher-risk MDS, respectively. Major responses were observed in all three hematologic lineages; 16% of RBC transfusion-dependent patients and 29% of platelet transfusion-dependent patients became transfusion independent. At data cut off, the median response duration was 3.4 months, with responses ongoing in nine patients. Conclusion Arsenic trioxide treatment consisting of an initial loading dose followed by maintenance therapy has moderate activity in MDS, inducing hematologic responses in both lower- and higher-risk patients. This activity combined with a manageable adverse effect profile warrants the additional study of arsenic trioxide, particularly in combination therapy, for the treatment of patients with MDS.


Cancers ◽  
2020 ◽  
Vol 12 (6) ◽  
pp. 1689 ◽  
Author(s):  
Bianca Grosser ◽  
Meike Kohlruss ◽  
Julia Slotta-Huspenina ◽  
Moritz Jesinghaus ◽  
Nicole Pfarr ◽  
...  

We investigated the prognostic and predictive impact of p53 expression for gastric cancer (GC) patients treated without or with preoperative chemotherapy (CTx) and its relationship with specific molecular GC subtypes. Specimens from 694 GC patients (562 surgical resection specimens without or after CTx, 132 biopsies before CTx) were analyzed by p53 immunohistochemistry. High (H) and low (L) microsatellite instability (MSI) and Epstein–Barr virus positivity were determined previously. Our results show that aberrant p53 expression was a negative prognostic factor in uni- and multivariable analysis in the resection specimens cohort (each p < 0.01). Subgroup analysis showed the strongest prognostic effect for patients with distally located tumors or no CTx treatment. In the biopsy cohort before CTx, p53 did not predict response or survival. p53 expression was significantly different among the molecular subtypes in surgical resection and bioptic specimens with strong association of altered p53 with MSI-L. Patients with MSI-H and aberrant p53 showed the worst survival in the biopsy cohort. In conclusion, the prognostic impact of p53 in GC differs according to tumor localization and CTx. Altered p53 is characteristic for MSI-L, and the p53 status in biopsies before CTx delineates MSI-H subtypes with inverse prognostic impact.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4831-4831
Author(s):  
Dhatri Kodali ◽  
Hector Mesa ◽  
Ajay Rawal ◽  
Pankaj Gupta

Abstract Thrombocytosis at presentation is uncommon in the myelodysplastic syndromes (MDS), and its influence on the clinical course of the disease and on prognosis is uncertain. To determine the clinical course and long-term outcomes of patients (pts) with thrombocytosis at initial diagnosis of MDS, we conducted a retrospective analysis of 503 pts diagnosed with MDS between Jan 1966 and July 2006 at the Minneapolis VA Medical Center. Original bone marrow and peripheral blood slides and reports were reviewed with a hematopathologist (H.M.) in all pts with high platelet counts (&gt; 400 × 103/μL) and evidence of dysplasia. Clinico-pathological correlation was obtained by chart review. Patients with inadequate data, secondary causes of thrombocytosis, transient thrombocytosis, and those without evidence of dysplasia were excluded. Of 503 pts, 41 (8.2%) were found to have thrombocytosis at presentation. Their median age was 74 years. The spleen was enlarged (by imaging) in 6 pts. Peripheral blood counts (mean; range) at diagnosis were: hemoglobin (10.9 g/dL; 7.4 – 17.1), absolute neutrophils (7.9 × 103/μL; 0.8 – 30.7) and platelets (627 × 103/μL; 402 – 1231). The cases were re-classified according to the WHO classification of myeloid disorders as follows: chronic myelomonocytic leukemia-1 (CMML-1) = 17 (41%), refractory anemia with ringed sideroblasts and thrombocytosis (RARS-T) = 13 (32%) and others = 11 (27%; including 2 pts each with RA, MDS/myeloproliferative disorder-unclassified [MDS/MPD-U] and 5q- syndrome, and 1 pt each with RA with excess blasts [RAEB-1], therapy related MDS [tMDS], refractory cytopenia with multilineage dysplasia [RCMD], MDS-U and atypical chronic myeloid leukemia [Aty CML]). Bone marrow fibrosis was increased in 3 pts with CMML-1 and 1 with RARS-T, and was normal or only focally increased in all other pts. The IPSS risk category was Low in 15 pts, Int-1 in 3 pts and Int-2 in 2 pts. Cytogenetic data was not available in the other pts. Jak-2 mutation analysis was positive in 2 pts with RARS-T, negative in 1 pt each with RARS-T and MDS/MPD-U, and is pending in others. On follow-up, 2 pts with CMML-1 and 1 pt with Aty CML transformed to acute myeloid leukemia (AML) and both pts with 5q- syndrome transformed to CMML-2. Two pts with RARS-T progressed to RAEB-2 and 1 pt with CMML-1 progressed to CMML-2. One pt with CMML-1 developed marked myelofibrosis. Marked thrombocytosis required hydroxyurea treatment in 5 pts. One MDS-U pt received 5-azacytidine. Four of 41 (10%) pts experienced major bleeding events; 3 were receiving aspirin. Five pts required ongoing red cell transfusions. The median survival (MS) was 36 months in RARS-T, 60 months in CMML-1 and 27 months in others; the MS of all 41 pts was 48 months. Causes of death were AML in 3 pts, cytopenias due to MDS in 6 pts and unrelated/unknown in 21 pts. Eleven pts are currently alive. In conclusion, the majority of pts presenting with myelodysplasia and thrombocytosis fall in the MDS/MPD category of the new WHO classification (most commonly CMML or RARS-T), be older, and have low-risk features (IPSS Low or Int-1). The risks of spontaneous bleeding, transformation to AML, progression of disease or myelofibrosis are low, and the overall prognosis is relatively favorable. Platelet counts may reach levels requiring cytoreductive therapy. This study helps better understand the natural history and prognosis of this varied spectrum of MDS and overlap syndromes.


Sign in / Sign up

Export Citation Format

Share Document