scholarly journals Survival Analysis According to Minimal Residual Disease By Flow Cytometry in Acute Myeloid Leukemia after First Induction

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1433-1433
Author(s):  
Claudia Nunez-Torron ◽  
Juan Marquet Palomanes ◽  
Fernando Martín Moro ◽  
Adolfo Saez-Martin ◽  
Alejandro Luna ◽  
...  

Introduction: Cytogenetic and molecular landscape at diagnosis and the depth of response to induction therapy are the most powerful prognostic tools available in patients with Acute Myeloid Leukemia (AML). Among the tests to measure Minimal Residual Disease (MRD), one of the most commonly used is flow cytometry. Nowadays, there is no consensus about optimal time for measurement and the threshold above which has greater prognostic value in AML, as well as its involvement in therapeutic management. Material and methods: We performed a single-center retrospective analysis of 62 patients diagnosed with AML between 2015 and 2019 that reached complete remission after first induction. Patients were stratified according to the European Leukemia Net (ELN) risk classification. MRD measurement was made in bone marrow samples with an 8-color flow cytometer (sensitivity 10-5), cut-off 0.1%. We divided our cohort in two groups according to MRD after induction: negative MRD (MRD-) and positive MRD (MRD+). The baseline characteristics of each group were compared using the Chi2 test. The survival analysis was performed through Kaplan-Meier method and the risk was calculated with Cox regression. The Overall Survival (OS) was defined as the period of time from diagnosis to death and the leukemia-free state (LFS) as the period of time from CR to either relapse or death. Statistical analysis were carried out using SPSS version 19.0. P<0.05 was defined as statistically significant difference. Results: Baseline characteristics of the sample are represented in Table 1. The median follow-up was 15 months (1-45). The 3-year LFS for MRD- and MRD+ was 50% and 29% respectively (figure 1A) showing a hazard ratio (HR) of 2.02 (CI 95% 0.98-4.6, p=0.05). The 3-year OS was 50% and 36% for MRD- and MRD+ respectively (figure 1B) and HR was 1.3 (CI 95% 0.6-2.8, p=0.4). In MRD+ group according to ELN risk classification, the 3-year LFS was 100% vs 21% vs 19% for favorable, intermediate and adverse risk, respectively (p=0.01) with a HR of 2.1 (CI 95% 1.1-3.9, p=0.01) in the univariate analysis. The 3-year OS was 100% vs 24% vs 18% for each subgroup (p=0.03) with a HR of 2.1 (CI 95% 1.06-4.4, p= 0.03). Thirty-eight patients received consolidation with hematopoietic stem cell transplantation (HSCT), and MRD prior to HSCT was measured in 32 of 38, being positive in 10 patients. All cases with MRD+ before HSCT belonged to MRD+ after induction group (10/20) (p=0.03). In MRD+ group, the 3-year LFS was 36% among those who received HSCT vs 18% in those who did not (log rank p=0.02, HR 3.1 CI 95% 1.3-7.8 p=0.01). Conclusions: The persistence of MRD > 0.1% after first induction by flow cytometry has shown in our population the identification of a AML subgroup of high risk, specially relevant in the intermediate risk group of ELN classification. MRD+ leads to higher risk of relapse, and these patients benefit from more aggressive therapeutic strategies, including allogeneic HSCT. However, MRD+ group has more risk of MRD persistence prior to HSCT, the last being a knowing factor of relapse after allogeneic HSCT, what would justify more aggressive strategies after HSCT in these patients. Disclosures Piris-Villaespesa: Novartis: Honoraria, Other: Advisory Boards. García Gutiérrez:BMS: Honoraria, Research Funding; Novartis: Honoraria, Research Funding; Pfizer: Honoraria, Research Funding; Incyte: Honoraria, Research Funding.

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.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 15-16
Author(s):  
Claudia Nunez-Torron ◽  
Fernando Martin Moro ◽  
Juan Marquet Palomanes ◽  
Miguel Piris-Villaespesa ◽  
Ernesto Roldan ◽  
...  

Introduction: Patients with Acute Myeloid Leukemia (AML) and positive Minimal Residual Disease (MRD) prior to allogeneic transplant are currently considered to be a group at high risk of relapse. Multiparameter flow cytometry is a standard technique to measure MRD, and generally we use a 0.1% threshold for positivity. The clinical significance of those patients with an MRD levels >0% but <0.1% is uncertain and it is recommended to define the prognosis of this subgroup. Material and methods: We performed a single-center retrospective analysis of 88 patients transplanted between 2012 and 2020. All patients achieved complete remission (CR) with or without hemoperipheral recovery prior to allogeneic transplant. We have divided our cohort into three groups according to MRD state by flow cytometry: Group 1 patients with negative MRD, Group 2 patients with MRD level >0% but <0.1% and Group 3 patients with MRD ≥ 0.1%. The baseline characteristics of each group were compared using the Chi2 test. The survival analysis was performed through Kaplan-Meier method and the risk was calculated with Cox regression. The Overall Survival (OS) was defined as the time from transplantation to death and the Relapse-Free Survival (RFS) as the time from transplantation to either relapse or death. P<0.05 was defined as statistically significant difference. Results: The baseline characteristics of our cohort are reflected in Table 1. We did not find statistical significant differences except for the response to induction. The median follow-up of the entire cohort was 13.5 months (range 6-43.5). The 4-year RFS (4y-RFS) was 47% and the 4-year OS (4y-OS) 50%. The 4y-RFS was 52.5% in Group 1 vs 59% in Group 2 vs 30% in Group 3. The 4y-OS was 60% in Group 1 vs 60% in Group 2 vs 31% in Group 3 (Image 1). The Hazard Ratio (HR) for RFS and OS comparing Group 1 vs Group 2 was 0.9 [95% CI ((0.3-2.5)] and 1.1 [95% CI (0.4-3)] respectively. The HR for the RFS and OS comparing Group 1 vs 3 was 1.2 [95% CI (0.9-1.7)] and 1.2 [95% CI (0.8-1.6)]. We have stratified patients according to the European LeukemiaNet risk classification. In Group 1, the 4y-RFS was 79% in patients with Favorable Risk (FR) vs 55% in those with Intermediate Risk (IR) and 53% in patients with Adverse Risk (AR) [HR 1.2, 95% CI (0.6-2.3)] and the 4y-OS was 79% vs 54% vs 53% respectively [HR 1.3, 95% CI (0.6-2.5)]. In Group 2, the 4y-RFS was 100% in those with FR vs 83% in IR vs 33% in AR [HR 3.9, 95% CI (0.4-30)] and the 4y-OS was 100% vs 82% vs 36% respectively [HR 4, 95% CI (0.5-32%)]. In Group 3, the 4y-RFS in patients with FR was 82% vs 0% in IR vs 0% in AR [HR 2.1, 95% CI (1.1-4.1)] and the 4y-OS was 82% vs 0% vs 0% respectively [HR 1.6, 95% CI (0.8-3.3)] (Image 2). Conclusions: In our cohort, positive MRD >0.1% prior to transplant identified a group with worse RFS and OS compared to those with negative MRD or positive MRD level >0% but <0.1%. Positive MRD >0.1% is especially relevant in the IR and AR groups of the European LeukemiaNet risk classification. In the AR subgroup even any detectable level of positive MRD could identify patients with unfavorable post-transplant OS and RFS outcomes. We must establish post-transplant strategies in these patients to improve survival. Disclosures Garcia-Gutiérrez: Pfizer: Consultancy, Other: Travel, Accommodation, Expenses, Research Funding; Incyte: Consultancy, Other: Travel, Accommodation, Expenses, Research Funding; Bristol-Myers Squibb: Consultancy, Other: Travel, Accommodation, Expenses, Research Funding; Novartis: Consultancy, Other: Travel, Accommodation, Expenses, Research Funding.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1470-1470
Author(s):  
Lorena Lobo Figueiredo Pontes ◽  
Leandro F. Dalmazzo ◽  
Luciana Correa Oliveira de Oliveira ◽  
Bárbara A Santana-Lemos ◽  
Felipe M Furtado ◽  
...  

Abstract Abstract 1470 Investigation of minimal residual disease (MRD) after remission induction (RI) therapy provides important information for risk assessment in patients with acute myeloid leukemia (AML). The presence of immature and chemotherapy-resistant leukemic stem cells (LSC) within the bulk of AML blasts at diagnosis and in post-induction bone marrow (BM) may lead to relapses. Nevertheless, whether the frequency of cells with LSC characteristics and their clearance after induction is correlated with prognosis has not been established. Using four-color multiparametric flow cytometry (MFC), BM quantification of leukemic associated phenotypes (LAPs) and LSCs was performed at diagnosis and after the first RI from 37 AML patients, excluding acute promyelocytic leukemia, with a median age of 48 years and a male/female ratio of 0.95. All patients received the conventional daunorrubicin and cytarabin (3 + 7) RI chemotherapy. Complete remission (CR) was defined as BM blast count inferior to 5%. Thirty-three patients were classified according to the European Leukemia Net recommendation and 9/32 (28.1%) patients were allocated in favorable, 14/32 (43.8%) in intermediate and 9/32 (28.1%) in poor cytogenetic/molecular risk group. FLT3-ITD mutation was detected in 9/33 (27.3%) and 5/28 (17.9%) carried NPM1 mutation. MRD identification was performed on 20 erythrocyte-lysed whole BM samples after staining with a panel of directly conjugated monoclonal antibodies. Five patients were excluded from this analysis because a LAP could not be identified. Blasts gating was performed considering the low expression of CD45 and sideward scatter (SSC) and CD34 expression (these latter, when more than 20% of blasts were detected at diagnosis). Within this population, the following LAPs were investigated: CD15/CD117, HLA-DR/CD13, HLA-DR/CD33, CD2/CD56, CD19/CD11b, CD42a/CD33, CD64/CD11c or CD14/CD11c. LSCs were selected by the same CD45dim × SSC gating strategy and defined as CD34+/CD38−/CD123+. After staining procedures, at least one hundred thousand events were acquired in a FACScalibur flow cytometer and analysis was performed using the Cell Quest software. LAP and LSC quantification, at diagnosis and at days 21 to 30 of RI, was analyzed as a categorical variable defined as lower or higher than the median and was compared to the following variables: age (< or > 60 years old); WBC count (< or > 30 × 103); cytogenetic/molecular risk; and morphological CR. The comparison between LAP and LSC quantification at both time points was also assessed. Comparison of categorical variables was performed using Fisher's exact test or Yates' corrected chi-square for two or more variables, respectively. Statistical analyses were performed using SPSS 13.0 software and P < 0.05 was considered to be significant. LSC quantification at diagnosis was found at varying frequencies across different cytogenetic/molecular risk groups, being higher at the poor risk group (P = 0.041). Of note, 100% of the poor risk patients had high levels of LSC at diagnosis. In addition, the presence of FLT3-ITD mutation was associated with higher amounts of the LSC population at diagnosis (P = 0.043). The most frequent detected LAP was CD45dim/CD34+/HLA-DR+/CD13+. Low or high expression of the LAP was not correlated with the prognostic variables at diagnosis. CR rate was 83.33% and was not different in the groups with high or low levels of LSC at diagnosis. However, LAP and LSC quantification after RI were found to be correlated (P = 0.018), suggesting that the LSC subpopulation can be useful for MRD monitoring at this early treatment time-point. Therefore, LSC quantification by MFC at diagnosis can identify patients at high risk of relapse and offers the opportunity to study the stem cell compartment after chemotherapy. These findings are particularly important for the intermediate normal karyotype risk group patients, who frequently do not have specific molecular targets for MRD monitoring. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 934-934 ◽  
Author(s):  
Thomas Köhnke ◽  
Daniela Sauter ◽  
Katharina Ringel ◽  
Jan Braess ◽  
Wolfgang Hiddemann ◽  
...  

Abstract Abstract 934 Background: Induction chemotherapy in acute myeloid leukemia (AML) has been shown to successfully induce complete remission in over 70% of patients. However, a majority of patients experience subsequent relapse. Assessment of minimal residual disease (MRD) by flow cytometry at time of aplasia, after induction and after consolidation therapy has been shown to be of prognostic relevance for relapse free survival (RFS) and overall survival (OS). However, studies utilizing MRD diagnostics to guide therapeutic decisions in adult AML (excluding APL) are yet to be performed. Methods: From the database at the Laboratory of Leukemia Diagnostics at our clinic datasets of 583 patients with newly diagnosed AML treated between 2000 and 2011 were analyzed. Patients with biphenotypic acute leukemia, M3 according to FAB classification, as well as those not treated with intensive induction chemotherapy were excluded. To be eligible for further analysis, at least two samples of bone marrow blood (at primary diagnosis and at one further timepoint during or after treatment) had to be available for MRD assessment by 3-color-flow cytometry at our laboratory. Cytogenetic and molecular risk stratification was performed at our clinic and assigned in accordance to the European LeukemiaNet (ELN) guidelines. We used Cox Proportional Hazards Regression to determine prognostic factors for OS and RFS and Kaplan-Meier estimator to determine OS and RFS of the proposed score. Results: Data of 217 Patients fulfilled the inclusion criteria and were therefore eligible for further analysis. 171 (78,8%) patients achieved CR after induction therapy. Of these patients, 120 had flow cytometry data available at time of aplasia and were included in further analysis. The median age was 54,5 y and the median OS 1007 days. Here, only “favorable” ELN risk stratification was associated with significantly longer OS (favorable vs. intermediate-I, Intermediate-II & adverse, Hazard Ratio, HR 0,36, 95% CI 0,19–0,69, p=0,0019), whereas RFS did not yield a significant difference (HR 0,64, 0,37-1,13, p=0,125). Age > 60y was associated with significantly shorter OS (HR 2,07, 1,23-3,47, p=0,0058) and RFS (HR 1,83, 1,11-3,01, p=0,018). And though leukemia-associated phenotypes (LAIP) ≥0,15% at time of aplasia were not predictive of OS (HR 1,32, 0,79–2,23, p=0,293) they were highly predictive of shorter RFS (HR 2,15, 1,30–3,55, p=0,003). Combining these three factors in a simple prognostic score (ELN risk group “favorable” = 0 points, “intermediate-I”, “intermediate-II” or “adverse” = 1 point; age > 60y = 1 point; LAIP at time of aplasia ≥0,15% = 1 point, see table I) identified three distinct groups (0 points: good, 1 point: intermediate, 2–3 points: poor, see table II) which were predictive of both OS and RFS (see figures 1 and 2). Interestingly, this score was capable of identifying a small group of patients with a very good prognosis (n=18, median OS and RFS not reached after >6 years) while at the same time equally dividing up the remaining patients within the intermediate and poor prognosis group (n=52 vs. 50, median OS 1182 vs. 677 days, median RFS 1180 vs. 334 days). Conclusion: MRD based therapeutic decisions and risk-adapted therapy have long been suggested in management of adult AML. Here, we propose a prognostic score for patients with AML achieving CR under intensive induction chemotherapy. The addition of MRD Flow to established genetic prognostic markers as well as age improves the prediction of relapse free and overall survival. Application of this score in therapeutic decisions could assist the treating physician and avoid over-treatment. To further evaluate our proposed prognostic score, it has to be applied in a prospective study for further evaluation and determination of its clinical significance. These data will be the basis for therapeutic trials guided by MRD assessment. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3695-3695 ◽  
Author(s):  
Michele Malagola ◽  
Crisitina Skert ◽  
Enrico Morello ◽  
Francesca Antoniazzi ◽  
Erika Borlenghi ◽  
...  

Abstract Background: Although a complete remission (CR) can be achieved in 70-80% of newly diagnosed acute myeloid leukemia (AML) patients, relapses occur in up to the 50% of cases. Thus, minimal residual disease (MRD) monitoring is a major issue for early detection of patients at high-risk of treatment failure and relapse. Aim: to dynamically evaluate WT1 pan-leukemic molecular marker of MRD in patients with AML. Matherial and methods: 107 newly diagnosed AML patients consecutively treated between 2010 and 2013 were monitored with quantitative WT-1 from bone marrow (BM) and peripheral blood (PB) at baseline, after induction, after the first consolidation course, before allogeneic stem cell transplantation (allo-SCT), at the 3rd and the 6th month after transplantation Results: At diagnosis, 104/107 (97%) had increased PB and BM WT1 levels assessed according to the ELN assay. Eighty-eight out of 107 patients (82%) achieved a complete remission (CR) after induction, 30/88 (34%) relapsed during follow up and 24/107 (22%) were addressed to allogeneic stem cell transplantation (allo-SCT). By univariate analysis, PB-WT > 50x10^4/ABL and BM-WT1 > 250x10^4/ABL after induction (PB: p=0.02; BM: p=0.04), after consolidation (PB: p=0.003), at the end of treatment (PB and BM: p=0.001), at 3rd month of follow up (PB and BM: p=0.005) and at 6th month of follow up (PB: p=0.005) were associated with a reduced overall survival (OS). By multivariate analysis, a BM-WT1 > 250 x 10^4/ABL at the end of treatment was significantly associated with a reduced OS. In order to adapt the cut-off of WT1 in our series of patients, we considered WT1 levels as continuous variables and categorized them at approximately the 25th, 50th, and 75th percentile. A cut-off of PB-WT1 > 25x10^4/ABL and BM-WT1 > 125x10^4/ABL at the end of the treatment program was identified as correlated with reduced leukemia-free survival (LFS) and OS (p=0.001). Similarly, and restricting the analysis on the 24 patients allo-transplanted in CR, 8/11 (73%) with pre-transplant PB-WT1 ≥ 5 and 4/13 (31%) with PB-WT1 < 5 relapsed, respectively (p=0.04). The incidence of relapse was higher in AML patients with PB-WT1 ≥ 5 measured at 3rd (56% vs 38%; p=0.43) and 6th month (71% vs 20%; p=0.03) after allo-SCT. Interestingly, 5/5 (100%) patients with pre-transplant PB-WT1 ≥ 5 who never reduced this level at 3rd or 6th month after allo-SCT experienced a disease recurrence. Conclusions: our data, although retrospectively collected, show that WT1 monitoring may be useful to predict the relapse in AML patients. Acknowledgments: This work was supported in part by Banca di Credito Cooperativo di Pompiano e Franciacorta and Lions Club Bassa Bresciana Association. Disclosures Russo: Celgene: Research Funding; Gilead: Research Funding; Novartis: Consultancy.


Sign in / Sign up

Export Citation Format

Share Document