scholarly journals Persistence of minimal residual disease assessed by multiparameter flow cytometry is highly prognostic in younger patients with acute myeloid leukemia

Cancer ◽  
2016 ◽  
Vol 123 (3) ◽  
pp. 426-435 ◽  
Author(s):  
Farhad Ravandi ◽  
Jeffrey Jorgensen ◽  
Gautam Borthakur ◽  
Elias Jabbour ◽  
Tapan Kadia ◽  
...  
Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2371-2371
Author(s):  
Wolfgang Kern ◽  
Daniela Voskova ◽  
Claudia Schoch ◽  
Susanne Schnittger ◽  
Torsten Haferlach

Abstract Treatment of acute myeloid leukemia (AML) is adapted according to individual risk profiles. The assessment of levels of minimal residual disease (MRD) is feasible in the vast majority of patients with AML and may significantly improve prognostication. We applied multiparameter flow cytometry to highly sensitively quantify MRD in patients with AML and analyzed its prognostic impact in a multivariate analysis. A total of 356 patients receiving standardized intensive antileukemic treatment was analyzed at different checkpoints: CP1 (up to day 28, n=156), CP2 (day 28–60, n=122), CP3 (day 61–120, n=195), CP4 (day 121–365, n=172), and CP5 (after day 365, n=73). Leukemic cells were identified by their individually defined leukemia-associated aberrant immunophenotypes (LAIPs). MRD levels were calculated as the logarithmic difference (LD) between LAIP-positive cells at diagnosis and LAIP-positive cells at follow-up assessment. The median LD amounted to 2.18 (range, −0.14 to 4.20) at CP1, 2.31 (−0.03 to 4.17) at CP2, 2.49 (0.11 to 4.17) at CP3, 2.58 (−0.28 to 4.28) at CP4, and 2.87 (0.46 to 4.02) at CP5. A higher LD (continuous variable) was related to a better event-free survival (EFS; CP1, p=0.0002; CP2, p=0.00001; CP3, p=0.0002; CP4, p<0.00001; CP5, p=0.00007) and to a better overall survival (OS; CP1, p=0.004; CP2, p=0.001; CP3, p=0.021; CP4, p=0.00006). Other parameters related to EFS and OS were age and cytogenetics in the present series. The prognostic impact of MRD levels on outcome was idependent of cytogenetics and age for EFS (CP2 to CP5) and OS (CP2 and CP4). MRD was the most important prognostic parameter at CP4 and CP5. Separation of patients into two groups, respectively, by the median LD resulted in significant differences in EFS at all CPs and in OS at CP1 to CP4. The largest difference was observed at CP4: median EFS, 57.1 vs. 13.7 months, p<0.00001; 3-year-OS, 95% vs. 65%, p=0.0003. Determination of MRD levels by multiparameter flow cytometry provides a highly powerful and independent prognostic parameter which is applicable to the total of an AML population. It should be evaluated as a stratification parameter in clinical trials.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2579-2579
Author(s):  
Farhad Ravandi ◽  
Jorgensen L Jeffrey ◽  
Elias J Jabbour ◽  
Gautam Borthakur ◽  
Tapan Kadia ◽  
...  

Abstract Background - Minimal Residual Disease (MRD) assessed by Multi-parameter Flow Cytometry (MFC) has not been extensively evaluated as a predictor of outcome in younger patients with acute myeloid leukemia (AML) receiving ara-C plus anthracycline induction Methods - We examined the predictive value of MRD assessment by MFC in mainly younger patients with newly diagnosed AML who were treated with intermediate dose cytarabine (total of ≥ 6 g/m2) and idarubicin based induction chemotherapy. Among 280 patients treated with clofarabine, idarubicin plus ara-C (CIA), fludarabine, idarubicin plus ara-C (FIA), fludarabine, ara-C, GCSF plus idarubicin (FLAG-Ida), cladribine, idarubicin, plus ara-C (CLIA) who achieved complete remission (CR), CR without platelet recovery (CRp), or CR with incomplete count recovery (CRi) 186 had at least one available MRD data and are the subject of this analysis. MRD by MFC was assessed using an 8-color panel containing 19 distinct markers, on bone marrow specimens obtained at the time of achievement of CR/CRi/CRp (approximately 1-2 months post induction), during consolidation (approximately 3-7 months post-induction) and at completion of therapy (≥ 8 months post induction). Residual leukemic blasts were identified based on phenotypic differences from normal myelomonocytic precursors. Sensitivity was estimated at 0.1% in most cases, with maximum achievable sensitivity of 0.01%, depending on the leukemic phenotype. Results - Median age of the patients was 51 years (Range, 17 - 77 years). 6 patients were older than 65 (all with ELN favorable disease). Median WBC at presentation was 4.7 x 109/L (Range, 0.5 - 103 x 109/L). Cytogenetics was favorable risk in 34 (18%), intermediate risk in 115 (62%) and adverse in 27 (15%) and was not available in 10 (5%) Treatment included CIA in 102 (55%), FIA in 34 (18%), FLAG-Ida in 34 (18%) and CLIA in 16 (9%). 166 patients had available samples at 1-2 months post induction and 131 (79%) became MRD negative. Achieving MRD negative status at response was associated with a statistically significant improvement in relapse free survival (RFS) (p= 0.001) and overall survival (OS) (p= 0.003) (Figure 1). 116 patients were evaluated for MRD status during consolidation and 100 (86%) became negative. Achieving a negative MRD status during consolidation was associated with a significant improvement in RFS (p˂0.001) and OS (p˂0.001)(Figure 2). 69 patients were evaluated for MRD status after completion of all therapy and 58 (84%) became negative. Achieving a negative MRD status at completion of therapy was associated with improvement in RFS (P˂0.001) and OS (P˂0.001) (Figure 3). On multivariate analysis including age ˂40 years vs. ≥ 40, WBC at presentation ˂or ≥ 10 x 109/L, cytogenetics (favorable, intermediate, adverse), achieving CR vs. CRp/CRi, and treatment with CIA, FIA, CLIA, or FLAG-Ida, achieving MRD negative status was the only independent predictor of RFS and OS at response (P=0.03 and P=0.005, respectively), during consolidation (p˂0.001 for both), or at completion of therapy (p˂0.001 for both). Conclusion - Achieving MRD negative status by MFC at response, during consolidation therapy and after completion of therapy is associated with a highly significant improvement in the outcome of younger patients with AML receiving ara-C plus idarubicin-based regimens. Figure 1. Figure 1. Figure 2. Figure 2. Figure 3. Figure 3. Disclosures Jabbour: Pfizer: Consultancy, Research Funding. Faderl:Astellas: Research Funding; Celator: Research Funding; JW Pharma: Consultancy; Seattle Genetics, Inc.: Research Funding; Karyopharm: Consultancy, Research Funding; Pfizer: Research Funding; Celgene: Consultancy, Research Funding, Speakers Bureau; Onyx: Speakers Bureau; Ambit: Research Funding; BMS: Research Funding. Pemmaraju:Stemline: Research Funding; Incyte: Consultancy, Honoraria; Novartis: Consultancy, Honoraria, Research Funding; LFB: Consultancy, Honoraria. Konopleva:Novartis: Research Funding; AbbVie: Research Funding; Stemline: Research Funding; Calithera: Research Funding; Threshold: Research Funding. Cortes:BerGenBio AS: Research Funding; Teva: Research Funding; Novartis: Consultancy, Research Funding; BMS: Consultancy, Research Funding; Pfizer: Consultancy, Research Funding; Ariad: Consultancy, Research Funding; Astellas: Consultancy, Research Funding; Ambit: Consultancy, Research Funding; Arog: Research Funding; Celator: Research Funding; Jenssen: Consultancy.


Sign in / Sign up

Export Citation Format

Share Document