scholarly journals Allogeneic Transplant Can Abrogate the Relapse Risk in the Patients with Detectable Measureable Residual Disease By Multicolor Flow-Cytometry at the Time of Assessment of Acute Myeloid Leukemia Patients in First Remission

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 36-37
Author(s):  
Muhned Alhumaid ◽  
Georgina S. Daher-Reyes ◽  
Arjun Law ◽  
Auro Viswabandya ◽  
Armin Gerbitz ◽  
...  

BACKGROUND: Acute myeloid leukemia (AML) is a highly heterogeneous group of diseases with variable response to therapy. Several factors have a prognostic impact for an outcome. Despite intensive chemotherapy and hematopoietic stem cell transplant (HCT), a significant proportion of patients eventually relapse, indicating that morphological assessment is not adequate due to limitations in sensitivity, requiring a better tool for assessment of remission. METHODS: A retrospective analysis was performed in AML patients who achieved first complete remission (CR1) and the outcomes compared according to the performance of HCT, and multi-color flow cytometry (MFC)-based measurable residual disease (MRD) status (defined as negative if patients achieved 0.1% or less) assessed at the time of CR1. In order to take account of the time interval from the MFC-MRD assessment to HCT, we applied a Mantel-Byar test for overall (OS) and relapse-free survival (RFS), considering time-to-HCT as a time-dependent covariate, while Simon and Makuch plot was used. Time-dependent Cox proportional hazard models were applied for multivariate analysis. Cumulative incidence of relapse (CIR) and non-relapse mortality (NRM) where evaluated using Fine-Gray model. RESULTS: A total of 435 patients diagnosed with AML and treated with induction chemotherapy between 2015 and 2018, of whom 380 patients (87%) achieved remission, were included. MFC-MRD was assessed in 336 patients in CR1 (77%), out of 380 patients who achieved CR1, and 200 patients (53%) proceeded to HCT. We evaluated OS, RFS, CIR and NRM according to MFC-MRD status in those patients who had negative MRD (MRDneg; n=218, 65%) vs. those with MRD (MRDpos; n=118, 35%). The OS at 2 years was 67.0% vs.40.7% (p≤0.001), RFS at 2 years was 8.7% vs. 40.6% (p≤0.001), CIR 26.9% vs.21.1% but with borderline significance (p=0.08), and NRM 32.5% vs. 20.2% with borderline significance (p=0.057). In patients who achieved CR, we compared OS, CIR, NRM and RFS between the HCT group (n=200) vs. those who did not undergo HCT (no-HCT; n=235). Between the 2 groups, the OS at 2 years was 55.7% vs. 47.2% (p=0.004); CIR 9.7% vs. 34.6% (p≤0.001); NRM 40.9% vs. 12.6% (p≤0.001). There was no difference in RFS: 49.4% vs. 52.8% (p=0.505). There was no difference in the time interval from the MFC-MRD assessment to HCT between the groups (MRDpos vs MRDneg) with a median of 96 days in overall patients who received HCT (p=0.31). In the overall population, when HCT was accounted as a time-dependent covariate, we failed to observe any difference of OS (HR 1.23; p=0.19) or RFS (HR 1.09; p=0.60) between the HCT vs. no-HCT groups. Then, we compared the OS, RFS, CIR, and NRM between the HCT vs no-HCT groups confined to the subgroups of patients with MFC-MRDneg vs MFC-MRDpos, separately. In the MFC-MRDpos subgroup, patients who underwent HCT did better: OS 54.8% in HCT vs. 25.5% in no-HCT (HR 0.52; p≤0.001) and RFS 48.7% vs. 24.1% (HR 0.45; p≤0.001). However, in the MFC-MRDneg subgroup, similar outcomes were noted between the HCT vs no-HCT groups in terms of OS 60.8% vs. 70.7% (HR 1.27, p=0.32), RFS 51.6% vs. 62.4% (HR 1.25; p=0.46) (Fig 1). With respect to the cause of treatment failure according to treatment modality (HCT vs no-HCT) and MFC-MRD status, Fig 2 revealed different patterns of relapse vs NRM between the HCT and the no-HCT groups. In the MFC-MRDneg subgroup, HCT group showed a higher NRM over the no-HCT group (38.0% vs 8.7%; HR 2.08; p≤0.001), while relapse risk was lower in the HCT group (10.4% vs 29.3%; p≤0.001). In the MFC-MRDpos subgroup, relapse incidence was strikingly different in favor of HCT (9.5% vs 50.0%; HR, p≤0.001). Conclusion: These findings suggest that in AML patients HCT could abrogate the relapse risk in patients who are MFC-MRDpos at the time of remission assessment, while the benefit from HCT was minimal in the subgroup that are MFC-MRDneg. Further study is strongly warranted to reach a clearer conclusion with a larger number of cohorts. Disclosures Lipton: Bristol-Myers Squibb: Honoraria; Takeda: Consultancy, Honoraria, Research Funding; BMS: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; Pfizer: Consultancy, Honoraria, Research Funding; Ariad: Consultancy, Research Funding.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1015-1015
Author(s):  
Pramod Pinnamaneni ◽  
Jeffrey L. Jorgensen ◽  
Hagop M. Kantarjian ◽  
Elias Jabbour ◽  
Sherry R. Pierce ◽  
...  

Abstract Purpose – To determine the value of Minimal Residual Disease (MRD) assessed by Multi-parameter Flow Cytometry (MFC) after achieving initial response to therapy, in predicting outcome in patients with acute myeloid leukemia (AML) Methods – We investigated the predictive value of MRD assessment by MFC in 191 patients with newly diagnosed AML treated between February 2010 and April 2014 at our institution who had available MRD assessment. 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 and at approximately 30 days and 90 days after achieving CR. 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 – Of the 191 patients, 167 (87%) achieved complete remission (CR) or CR without platelet recovery (CRp). Their median age was 58 years (Range, 17-85). 84 (44%) were older than 60 years. Median WBC at presentation was 3.2 x 109/L(Range, 0.5-100.2 x 109/L) and median bone marrow blast percentage was 43% (Range, 11-96%). Cytogenetics was favorable risk in 4 (2%), intermediate risk in130 (68%) and adverse risk in 57 (30%). Treatment included cytarabine plus anthracycline in 170 (89%) and hypomethylating agents-based strategies in 21 (11%). 48 patients had available samples at 30 days post CR and 32 (67%) became MRD negative. Achieving MRD negative status was associated with a statistically significant improvement in CR duration (p=0.02) and overall survival (OS) (p=0.0005). 56 patients were evaluated for MRD status at 90 days and 45 (80%) were negative. Again, achieving MRD negative status was associated with a significant improvement in CR duration (p=0.002) and OS (p=0.0009). Conclusion – Achieving MRD negative status by MFC at 30 and 90 days post CR is associated with an improved outcome in patients with AML Figure 1 Figure 1. Figure 2 Figure 2. Disclosures No relevant conflicts of interest to declare.


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 ◽  
2012 ◽  
Vol 120 (8) ◽  
pp. 1581-1588 ◽  
Author(s):  
Michael R. Loken ◽  
Todd A. Alonzo ◽  
Laura Pardo ◽  
Robert B. Gerbing ◽  
Susana C. Raimondi ◽  
...  

Abstract Early response to induction chemotherapy is a predictor of outcome in acute myeloid leukemia (AML). We determined the prevalence and significance of postinduction residual disease (RD) by multidimensional flow cytometry (MDF) in children treated on Children's Oncology Group AML protocol AAML03P1. Postinduction marrow specimens at the end of induction (EOI) 1 or 2 or at the end of therapy from 249 patients were prospectively evaluated by MDF for RD, and presence of RD was correlated with disease characteristics and clinical outcome. Of the 188 patients in morphologic complete remission at EOI1, 46 (24%) had MDF-detectable disease. Those with and without RD at the EOI1 had a 3-year relapse risk of 60% and 29%, respectively (P < .001); the corresponding relapse-free survival was 30% and 65% (P < .001). Presence of RD at the EOI2 and end of therapy was similarly predictive of poor outcome. RD was detected in 28% of standard-risk patients in complete remission and was highly associated with poor relapse-free survival (P = .008). In a multivariate analysis, including cytogenetic and molecular risk factors, RD was an independent predictor of relapse (P < .001). MDF identifies patients at risk of relapse and poor outcome and can be incorporated into clinical trials for risk-based therapy allocation. This study was registered at www.clinicaltrials.gov as NCT00070174.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 15-15
Author(s):  
Amer M. Zeidan ◽  
Prajwal C Boddu ◽  
Brent L Wood ◽  
Daniel Zelterman ◽  
Richard F. Little ◽  
...  

Study Background: The presence of minimal residual disease (MRD) after IC in AML patients is often a harbinger of relapse, and therapeutic targeting of MRD has emerged as an important strategy to improve long-term outcomes. The importance of the PD-1/PD-L1 pathway in immune evasion of AML has been demonstrated in murine AML models whereby PD-L1 and PD-1 were upregulated in leukemia cells, and PD-1 blockade suppresses invivo leukemia cell proliferation and improved survival in AML bearing mice (Zhang et al., 2009; Zhou et al., 2010). Combining IC with immune checkpoint inhibition (ICPI) has resulted in a significant clinical activity in some advanced solid malignancies including non-small-cell lung cancer. A recent single-arm phase 2 study combining IC with nivolumab showed the combination was feasible and resulted in undetectable MRD in 53% of patients with Complete response (CR)/CR with incomplete count recovery (CRi) (Ravandi et al, 2019). Here we report on the design of the first randomized multi-center clinical trial of IC+ICPI in fit AML patients. Methods: The primary objective of this investigator-initiated Cancer Therapy Evaluation Program (CTEP)-sponsored multi-institutional, randomized phase II study (NCT04214249) is to assess the percentage of patients with MRD negative CR (MRD- CR) as measured by flow cytometry at the end of first cycle of consolidation therapy with IC+ pembrolizumab and compare between the two study arms (Figure 1). Secondary objectives include rates of CR/CRi, complete remission with partial recovery count (CRh) and hematologic improvement (HI) to red blood cells and platelets, MRD negativity at Day 14, MRD-negative CR at end of induction therapy and MRD negative CR after last consolidation cycle, event free survival (EFS), relapse free survival (RFS), duration of response (DOR), and overall survival (OS), and proportion of patients who develop severe toxicity. Important exploratory objectives include MRD assessment by duplex sequencing (DS) and comparing DS and multiparameter flow cytometry for MRD detection. The study will consist of an induction, consolidation, and maintenance phase of therapy. A total of 124 patients will be included (62 patients in the intervention arm and 62 patients in the control arm). Planned stratification factors include 1) age (younger than 65 vs. 65 and older), 2) cytogenetics by FISH or metaphase karyotype (presence vs. absence of core binding factor inversions and translocations), and 3) t-AML or AML arising from prior/antecedent MDS. All patients will receive cytarabine at 100 mg/m2/day continuous infusion on Days 1-7 and either idarubicin 12 mg/m2/day IV on Days 1-3 or daunorubicin 60 mg/m2/day IV on Days 1-3. on Day 8 of the induction chemotherapy, half of the patients will receive pembrolizumab IV (intervention arm), and continue Q3 weeks throughout Induction/Consolidation, and thereafter for up to 2 years from the start of maintenance. The other half will not receive pembrolizumab (control arm). After 31 efficacy-evaluable patients (patients who had a bone marrow biopsy done after the first consolidation cycle) in each of the two study arms have had their primary endpoint results available, a formal futility and toxicity analysis will be conducted. Follow up will continue up to 2 years after the last patient is randomized. Eligible patients are aged ≥18 and ≤75 years with newly diagnosed and pathologically-confirmed AML, including secondary AML arising from prior MDS and therapy-related AML. Patients with FLT3-mutated AML are excluded as well as those with prior allogeneic stem cell transplant, active infection requiring systemic therapy, and requiring use of high dose corticosteroids. Responses in AML patients will be assessed using European Leukemia Net 2017 response criteria. EFS will be calculated as the time from initial treatment to either disease relapse or death. OS will be calculated from time from initial treatment to death. Biomarker analyses will include an extensive array of immunologic and correlative studies that will evaluate PD-1 and PD-L1 expression, immune cell subset analysis, leukemia specific T-cell response, cytokine level, T-cell receptor (TCR) repertoire, as well as RNA-seq, gut microbiome characterization and metabolomics, and whole exome sequencing for tumor and germline DNA. Figure 1 Disclosures Zeidan: Epizyme: Consultancy, Honoraria; Astellas: Consultancy, Honoraria; Daiichi Sankyo: Consultancy, Honoraria; Cardinal Health: Consultancy, Honoraria; Acceleron: Consultancy, Honoraria; Leukemia and Lymphoma Society: Other; Jazz: Consultancy, Honoraria; Celgene / BMS: Consultancy, Honoraria, Research Funding; Abbvie: Consultancy, Honoraria, Research Funding; Otsuka: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria, Research Funding; Seattle Genetics: Consultancy, Honoraria; BeyondSpring: Consultancy, Honoraria; Cardiff Oncology: Consultancy, Honoraria, Other; Takeda: Consultancy, Honoraria, Research Funding; Ionis: Consultancy, Honoraria; MedImmune/Astrazeneca: Research Funding; Trovagene: Consultancy, Honoraria, Research Funding; Aprea: Research Funding; ADC Therapeutics: Research Funding; Astex: Research Funding; Incyte: Consultancy, Honoraria, Research Funding; Taiho: Consultancy, Honoraria; CCITLA: Other; Novartis: Consultancy, Honoraria, Research Funding; Boehringer-Ingelheim: Consultancy, Honoraria, Research Funding; Agios: Consultancy, Honoraria. Radich:Amgen: Consultancy; Bristol-Myers Squibb: Consultancy; Jazz: Consultancy; Novartis Pharmaceuticals Corporation: Consultancy, 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.


Sign in / Sign up

Export Citation Format

Share Document