Prognostic Significance of β-2 Microglobulin Levels in Acute Myeloid Leukemia: Analysis of 1293 Patients.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 802-802
Author(s):  
Apostolia-Maria Tsimberidou ◽  
Hagop M. Kantarjian ◽  
Michael J. Keating ◽  
Susan O’Brien ◽  
Sijin Wen ◽  
...  

Abstract Introduction: β-2 Microglobulin (β2M) is a single polypeptide chain that is linked non-covalently to the major histocompatibility complex class I cell surface antigen. Its specific function is unknown, but serum β2M levels reflect membrane turnover (tumor mass and growth rate) and renal function. Elevated serum β2M levels are associated with poor survival in several hematologic malignancies, but its prognostic significance in acute myeloid leukemia (AML) is unknown. The purpose of this study was to determine the association between β2M levels and pretreatment characteristics and clinical outcomes in newly diagnosed AML. Patients and Methods: From 1990 to 2005, β2M levels were prospectively measured in 1293 patients with AML. Serum β2M was quantified by radioimmunoassay (normal range, 0.7–2.0 mg/L). Results: The median patient age was 61 yrs (range, 16–89 yrs); 54% were >60 yrs. Cytogenetics were favorable in 7% of patients, intermediate in 60%, poor-risk in 29%, and 4% of patients had insufficient metaphases. Zubrod performance status (PS) was 0–1 in 73% of patients, 2 in 20%, and 3–4 in 7%. Eighty-five percent had de novo AML, and 91% received Ara-C-based therapy. High β2M levels were more common in patients who were older (cor=0.22); had high early risk of mortality (ERM) score (cor =0.33); high levels of creatinine (cor=0.63), and uric acid (cor=0.29), high white blood cell counts (cor=0.26), or circulating monocytes (cor = 0.23); prolonged prothrombin time (PT) (cor=0.26); worse PS (cor=0.27); and low albumin levels (cor=−0.22)(p<0.001 for all variables). High β2M levels were correlated with worse-risk cytogenetics (p=0.03), RAS mutation (p=0.003), baseline infection (p=0.04), and secondary AML (p=0.01). The median follow-up of surviving patients was 3.8 yrs. In multivariate analysis, independent factors predicting response were younger age (p<0.0001), better-risk cytogenetics (p<0.0001), Ara-C-based therapy (p<0.0001), lower levels of β2M (p=0.0001), shorter PT (p=0.006), de novo AML (p=0.007), lower LDH levels (p=0.02), and lower bilirubin levels (p=0.03). Factors independently prognostic of longer survival were younger age (p<0.0001), better-risk cytogenetics (p<0.0001), better PS (p<0.0001), de novo AML (p<0.0001), lower serum uric acid levels (p=0.0001), lower LDH levels (p=0.0007), shorter PT (p=0.0009), lower β2M levels (p=0.003), higher hemoglobin levels (p=0.005), Ara-C-based therapy (p=0.007), and lower bilirubin levels (p=0.02). Factors independently prognostic of longer event-free survival (EFS) were younger age (p<0.0001), better-risk cytogenetics (p<0.0001), Ara-C-based therapy (p<0.0001), de novo AML (p=0.0001), better PS (p=0.0003), lower uric acid levels (p=0.0004), lower LDH levels (p=0.001), lower β2M levels (p=0.002), higher hemoglobin levels (p=0.003), shorter PT (p=0.02), and lower bilirubin levels (p=0.045). Conclusions: Elevated serum β2M levels are an independent adverse prognostic factor for response, survival, and EFS in the context of established prognostic factors for AML. Outcomes by β2M levels β2M (mg/L) <2.0 2.0–2.9 3.0–3.9 ≥4.0 p-value No. pts. 251 448 260 334 CR, % 67 66 54 39 <0.0001 Median EFS (yrs) 0.69 0.58 0.31 0.15 <0.0001 Median Survival (yrs) 1.33 1.19 0.63 0.42 <0.0001

1997 ◽  
Vol 15 (6) ◽  
pp. 2262-2268 ◽  
Author(s):  
M Wetzler ◽  
M R Baer ◽  
S H Bernstein ◽  
L Blumenson ◽  
C Stewart ◽  
...  

PURPOSE c-mpl, the human homolog of v-mpl, is the receptor for thrombopoietin. Given that c-mpl expression carries an adverse prognosis in myelodysplastic syndrome and given the prognostic significance of expression of other growth factor receptors in other diseases, we attempted to determine whether c-mp/mRNA expression is a prognostic factor in acute myeloid leukemia (AML). PATIENTS AND METHODS We analyzed bone marrow samples from 45 newly diagnosed AML patients by reverse-transcription polymerase chain reaction. RESULTS Samples from 27 patients (60%) expressed c-mpl mRNA (c-mpl+); their clinical and laboratory features were compared with those of the 18 patients without detectable levels of c-mpl(c-mpl-). No significant differences in age, sex, leukocyte count, French-American-British subtype, or karyotype group were found. c-mpl+ patients more commonly had secondary AML (41% v 11%; P = .046) and more commonly expressed CD34 (67% v 12%; P = .0004). There was no significant difference in complete remission (CR) rate. However, c-mpl+ patients had shorter CR durations (P = .008; median, 6.0 v > 17.0 months). This was true when only de novo AML patients were considered and when controlling for age, cytogenetics, or CD34 expression. There was a trend toward shorter survival in c-mpl+ patients (P = .058; median, 7.8 v 9.0 months). CONCLUSION These data suggest that c-mpl expression is an adverse prognostic factor for treatment outcome in adult AML that must be considered in the analysis of clinical studies using thrombopoietin in AML.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 7-8
Author(s):  
Hassan Awada ◽  
Cassandra M Kerr ◽  
Heesun J. Rogers ◽  
Jaroslaw P. Maciejewski ◽  
Valeria Visconte

Inversion or translocation of the chromosome 3, specifically inv(3)(q21q26.2/ t(3;3)(q21;q26.2) are present in 1-2% of acute myeloid leukemia (AML) cases and are classified as a distinct entity in the 2016 WHO classification. Hallmark genetic alterations in this entity include mutations in GATA2 and MECOM. In fact, these rearrangements result in over activation of MECOM due to juxtaposition with a distal GATA2 enhancer. Cytomorphologic phenotypes include anemia, normal to elevated platelets and multilineage dysplasia in a hyperplastic bone marrow (BM). Studies have shown the frequent occurrence of NF1, NRAS and RUNX1 mutations. While proceeding towards our molecularly informed AML subtyping (Awada, Blood 2019;1406), we observed a high occurrence of somatic mutations in the splicing factor SF3B1 in inv(3)/t(3;3) AML. We were particularly intrigued by this observation considering several key aspects of SF3B1 mutations in the context of MDS. For instance, SF3B1 mutations are highly associated with clear phenotypic and morphologic features and carry favorable prognosis in MDS. These mutations are often found in patients carrying less deleterious abnormalities [e.g., del(5q)] and their founder clonal nature has been uncovered through experimental studies. Recent studies unveiled the occurrence of SF3B1 mutations in de novo AML and low complete remission rate when combined with other mutations (e.g., DNMT3A). To investigate whether SF3B1 mutations were unequivocally frequent in inv(3)/t(3;3) AML compared to other splicing factor mutations, we moved forward in dissecting the clinical, morphologic and molecular profiles of these cases. We analyzed results from whole exome sequencing and targeted deep sequencing from the Cleveland Clinic and publicly available data of AML with inv(3)/t(3;3) (de novo AML, n=32; secondary AML from antecedent myeloid neoplasms, n=11; t-AML, n=1). In our cohort, mutations in the most common components of the RNA splicing machinery (SF3B1, SRSF2, U2AF1, ZRSR2) were observed in 27% (n=12) of the patients. Among splicing factor mutations, SF3B1 was the most mutated gene (77%; 10/13 total mutations); 7 cases had inv(3) and 3 had t(3;3). Mutations were observed at canonical sites: K700E (70%) and K666N (30%) with no difference compared to the hotspots observed in MDS. Sixty% of patients were female. Median age was 61 years (range, 36-73). Anemia was present in 50%, leukopenia in 10% and thrombocytopenia in 50% of the patients. For 40% of the cases, BM smears for iron staining was available and showed absence of ringed sideroblasts. Complex karyotype (CK) was present in 20% of the patients; -7/del(7q) was present as the only cytogenetic abnormality in 30% or with CK in 10% pf the cases. Variant allele frequency (VAF) of SF3B1mutations in inv(3)/t(3;3) was not different than the those without inv(3)/t(3;3) (42% vs 40%). Survival analysis was performed in 3 subgroups: SF3B1MT AML (n=70), SF3B1MT AML + inv(3)/t(3;3) (n=10), AML + inv(3)/t(3;3) (n=34). SF3B1MT AML + inv(3)/t(3;3) and AML + inv(3)/t(3;3) had similar OS (11.7 vs 9.7 months) which was shorter than the that of SF3B1MT AML without any inv(3)/t(3;3) (19.4 months, P=0.002) suggesting that SF3B1MT in the context of inv(3)/t(3;3) might hold a different prognostic significance strongly due to the presence of inv(3)/t(3;3). Given this observation, we delved into the clonal diversity of SF3B1 mutations and its co-occurrence with other molecular mutations. Comparison of VAFs showed that SF3B1 mutations in relation to other mutations were dominant/founder in 30%, secondary/subclonal in 20% while co-dominant to another gene (VAF differences <5%) in 50% of the cases. The most common co-dominant gene mutation was GATA2 (60%, 3/5). Top mutations by frequency were in GATA2 (30%), ASXL1 (20%) and NRAS (20%). Hemizygous GATA2 mutations were detected in 15% of SF3B1MT AML + inv(3)/t(3;3). In our cohort, other RAS gene mutations were detected in 10% of the patients each, including CBL, NF1 and PTPN11). One SF3B1MT AML + inv(3)/t(3;3) case also harbored a SRSF2 mutation with a parallel median VAF of 40% and 41%, respectively. The results of our study are summarized in Fig. 1. In sum, we describe that SF3B1 mutations occur in combination with inv(3)/t(3;3) in AML and might represent a subclass of this entity in which lesions in SF3B1 gene could potentially hide a cryptic association between splicing abnormalities and disease phenotypes. Figure 1 Disclosures Maciejewski: Alexion, BMS: Speakers Bureau; Novartis, Roche: Consultancy, Honoraria.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1385-1385
Author(s):  
Wade L Schulz ◽  
Thomas JS Durant ◽  
Henry Rinder ◽  
Christopher A Tormey ◽  
Richard Torres ◽  
...  

Abstract Background: Tumor heterogeneity has been documented in several malignancies and is generally associated with more aggressive disease and poorer prognosis. While heterogeneity has been documented in acute myeloid leukemia (AML), risk prognosis in AML is currently assessed by cytogenetics and the presence of recurrent mutations in genes such as FLT3 and NPM1, which are typically tested on a single gene basis. Prior approaches to identify molecular clonality have been laborious and therefore of limited clinical utility. The advent of next generation sequencing (NGS), however, allows for the rapid assessment of subclones within tumor populations. Currently, the prognostic significance of tumor heterogeneity in AML is not well defined. Therefore, this pilot study assessed the clinical significance of molecular clonality in AML using data generated by an NGS platform. Methods: We selected patients with de novo AML who had blood or bone marrow submitted for sequencing at initial diagnosis. Patients who subsequently elected for comfort measures only without receiving AML-directed therapy were excluded. Study patients were sequenced with a 25 gene NGS panel on the Ion Torrent platform. This panel included genes that have been shown to be frequently mutated in AML and which lead to increased cellular proliferation or impaired differentiation, with particular emphasis on therapeutic and prognostic mutations. Germline mutations, identified by allelic fraction and/or minor allele frequency, were excluded. Remaining somatic mutations were analyzed with software employing a variational Bayesian algorithm to predict tumor heterogeneity such that patients were assessed for the presence of tumor heterogeneity (≥2 predicted clones) at the time of diagnosis. Heterogeneity was recorded and compared to several clinical parameters, including post-chemotherapy survival at 120 days, patient age, gender, karyotype, and association with the recurrent genetic mutations NPM1, FLT3-ITD, and CEBPA. Results: Twenty patients met the study criteria of de novo AML and elected to receive therapy. The mean age at diagnosis was 59 years (38-77 years); overall survival was 75% at 120 days. Half of the patients (n=10) were determined to have ≥2 clones. Patients with either a complex karyotype or the FLT3-ITD mutation were classified as high risk (n=11). While survival at 120 days was lower in the high risk group (64%) compared to normal risk subjects (89%), this difference did not reach statistical significance (p=0.19). By contrast, patients with tumor heterogeneity (≥2 clones) had significantly lower 120 day survival (50%) when compared to AML patients with a single clone (100%) (p=0.01). There was no correlation between patients with multiple clones and high risk classification (p=0.18). Conclusions: These pilot data present some of the first prospective evidence that heterogeneity detected by NGS is an adverse prognostic indicator for early survival in de novo AML post-chemotherapy. This finding may not be surprising given that the presence of tumor heterogeneity has been shown to confer adverse prognostic risk in several solid malignancies. Still, this early impact of molecular clonality in AML may have important consequences on choice of therapy. Further prospective studies are needed to confirm our findings and to determine the long-term outcomes in AML patient subsets, as well as examining whether a distinct approach to therapy in AML patients with clonal heterogeneity leads to improved early survival. Figure 1. Figure 1. Figure 2. Figure 2. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2001 ◽  
Vol 98 (7) ◽  
pp. 2200-2209 ◽  
Author(s):  
Wolfgang R. Sperr ◽  
John-Hendrik Jordan ◽  
Mehrdad Baghestanian ◽  
Hans-Peter Kiener ◽  
Puchit Samorapoompichit ◽  
...  

α- and β-tryptase genes encode serine proteases that are abundantly expressed by mast cells. Under physiologic conditions other myeloid cells are virtually tryptase negative. However, tryptases are also expressed in several myeloid leukemia cell lines. In this study, serum total tryptase levels were determined in 150 patients with acute leukemias (de novo acute myeloid leukemia [AML], n = 108; secondary AML, n = 25; acute lymphoid leukemia [ALL], n = 17) by fluoroenzyme immunoassay. In healthy subjects (n = 30), tryptase levels ranged between 2.0 and 12.6 ng/mL. Elevated tryptase levels (> 15) were detected in 42 (39%) of 108 patients with de novo AML and in 11 (44%) of 25 patients with secondary AML. No elevated tryptase levels were found in patients with ALL. In de novo AML, elevated tryptase levels were frequently detected in patients with French-American-British classification M0 (6 of 9), M2 (9 of 14), M3 (4 of 6), and M4eo (7 of 7), and less frequently in M1 (7 of 20), M4 (6 of 26), M5 (2 of 18), M6 (0 of 5), or M7 (1 of 3). The highest tryptase levels were found in M4eo. Immunohistochemical staining of bone marrow sections with anti-tryptase antibody as well as immunoelectron microscopy revealed tryptase expression in the cytoplasm of myeloblasts. As assessed by Northern blotting and reverse transcriptase–polymerase chain reaction, AML cells expressed α-tryptase messenger RNA (mRNA) but little or no β-tryptase mRNA. In AML patients with elevated serum tryptase before chemotherapy, who entered complete remission, tryptase levels returned to normal or near normal values. Blast cell persistence or regrowth was associated with a persistently elevated level or recurrent increase of tryptase. Together, tryptase is expressed in myeloblasts in a group of AML and may serve as a useful disease-related marker.


Author(s):  
Yu-Hung Wang ◽  
Chien-Chin Lin ◽  
Chia-Lang Hsu ◽  
Sheng-Yu Hung ◽  
Chi-Yuan Yao ◽  
...  

AbstractExpression of long non-coding RNA KIAA0125 has been incorporated in various gene expression signatures for prognostic prediction in acute myeloid leukemia (AML) patients, yet its functions and clinical significance remain unclear. This study aimed to investigate the clinical and biological characteristics of AML bearing different levels of KIAA0125. We profiled KIAA0125 expression levels in bone marrow cells from 347 de novo AML patients and found higher KIAA0125 expression was closely associated with RUNX1 mutation, but inversely correlated with t(8;21) and t(15;17) karyotypes. Among the 227 patients who received standard chemotherapy, those with higher KIAA0125 expression had a lower complete remission rate, shorter overall survival (OS) and disease-free survival (DFS) than those with lower expression. The prognostic significance was validated in both TCGA and GSE12417 cohorts. Subgroup analyses showed that higher KIAA0125 expression also predicted shorter DFS and OS in patients with normal karyotype or non-M3 AML. In multivariable analysis, higher KIAA0125 expression remained an adverse risk factor independent of age, WBC counts, karyotypes, and mutation patterns. Bioinformatics analyses revealed that higher KIAA0125 expression was associated with hematopoietic and leukemic stem cell signatures and ATP-binding cassette transporters, two predisposing factors for chemoresistance.


Blood ◽  
2005 ◽  
Vol 105 (6) ◽  
pp. 2527-2534 ◽  
Author(s):  
Christian Récher ◽  
Odile Beyne-Rauzy ◽  
Cécile Demur ◽  
Gaëtan Chicanne ◽  
Cédric Dos Santos ◽  
...  

AbstractThe mammalian target of rapamycin (mTOR) is a key regulator of growth and survival in many cell types. Its constitutive activation has been involved in the pathogenesis of various cancers. In this study, we show that mTOR inhibition by rapamycin strongly inhibits the growth of the most immature acute myeloid leukemia (AML) cell lines through blockade in G0/G1 phase of the cell cycle. Accordingly, 2 downstream effectors of mTOR, 4E-BP1 and p70S6K, are phosphorylated in a rapamycin-sensitive manner in a series of 23 AML cases. Interestingly, the mTOR inhibitor markedly impairs the clonogenic properties of fresh AML cells while sparing normal hematopoietic progenitors. Moreover, rapamycin induces significant clinical responses in 4 of 9 patients with either refractory/relapsed de novo AML or secondary AML. Overall, our data strongly suggest that mTOR is aberrantly regulated in most AML cells and that rapamycin and analogs, by targeting the clonogenic compartment of the leukemic clone, may be used as new compounds in AML therapy.


Blood ◽  
2011 ◽  
Vol 118 (15) ◽  
pp. 4188-4198 ◽  
Author(s):  
Sebastian Schwind ◽  
Guido Marcucci ◽  
Jessica Kohlschmidt ◽  
Michael D. Radmacher ◽  
Krzysztof Mrózek ◽  
...  

AbstractLow MN1 expression bestows favorable prognosis in younger adults with cytogenetically normal acute myeloid leukemia (CN-AML), but its prognostic significance in older patients is unknown. We analyzed pretherapy MN1 expression in 140 older (≥ 60 years) de novo CN-AML patients treated on cytarabine/daunorubicin-based protocols. Low MN1 expressers had higher complete remission (CR) rates (P = .001), and longer overall survival (P = .03) and event-free survival (EFS; P = .004). In multivariable models, low MN1 expression was associated with better CR rates and EFS. The impact of MN1 expression on overall survival and EFS was predominantly in patients 70 years of age or older, with low MN1 expressers with mutated NPM1 having the best outcome. The impact of MN1 expression was also observed in the Intermediate-I, but not the Favorable group of the European LeukemiaNet classification, where low MN1 expressers had CR rates and EFS similar to those of Favorable group patients. MN1 expresser-status-associated gene- and microRNA-expression signatures revealed underexpression of drug resistance and adverse outcome predictors, and overexpression of HOX genes and HOX-gene–embedded microRNAs in low MN1 expressers. We conclude that low MN1 expression confers better prognosis in older CN-AML patients and may refine the European LeukemiaNet classification. Biologic features associated with MN1 expression may help identify new treatment targets.


2020 ◽  
Vol 38 (30) ◽  
pp. 3506-3517 ◽  
Author(s):  
Chong Chyn Chua ◽  
Andrew W. Roberts ◽  
John Reynolds ◽  
Chun Yew Fong ◽  
Stephen B. Ting ◽  
...  

PURPOSE The B-cell lymphoma 2 (BCL-2) inhibitor venetoclax has an emerging role in acute myeloid leukemia (AML), with promising response rates in combination with hypomethylating agents or low-dose cytarabine in older patients. The tolerability and efficacy of venetoclax in combination with intensive chemotherapy in AML is unknown. PATIENTS AND METHODS Patients with AML who were ≥ 65 years (≥ 60 years if monosomal karyotype) and fit for intensive chemotherapy were allocated to venetoclax dose-escalation cohorts (range, 50-600 mg). Venetoclax was administered orally for 14 days each cycle. During induction, a 7-day prephase/dose ramp-up (days −6 to 0) was followed by an additional 7 days of venetoclax combined with infusional cytarabine 100 mg/m2 on days 1-5 and idarubicin 12 mg/m2 intravenously on days 2-3 (ie, 5 + 2). Consolidation (4 cycles) included 14 days of venetoclax (days −6 to 7) combined with cytarabine (days 1-2) and idarubicin (day 1). Maintenance venetoclax was permitted (7 cycles). The primary objective was to assess the optimal dose schedule of venetoclax with 5 + 2. RESULTS Fifty-one patients with a median age of 72 years (range, 63-80 years) were included. The maximum tolerated dose was not reached with venetoclax 600 mg/day. The main grade ≥ 3 nonhematologic toxicities during induction were febrile neutropenia (55%) and sepsis (35%). In contrast to induction, platelet recovery was notably delayed during consolidation cycles. The overall response rate (complete remission [CR]/CR with incomplete count recovery) was 72%; it was 97% in de novo AML and was 43% in secondary AML. During the venetoclax prephase, marrow blast reductions (≥ 50%) were noted in NPM1-, IDH2-, and SRSF2-mutant AML. CONCLUSION Venetoclax combined with 5 + 2 induction chemotherapy was safe and tolerable in fit older patients with AML. Although the optimal postremission therapy remains to be determined, the high remission rate in de novo AML warrants additional investigation (ANZ Clinical Trial Registry No. ACTRN12616000445471).


Sign in / Sign up

Export Citation Format

Share Document