Minimal Residual Disease Assessed by WT1 Expression and NPM1 Mutations Specific RQ-PCR Assays Identifies Patients with Distinct Outcomes in the ALFA 0701 Trial and Is Decreased by Treatment with Gemtuzumab Ozogamicin

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 659-659 ◽  
Author(s):  
Juliette Lambert ◽  
Jerome Lambert ◽  
Olivier Nibourel ◽  
Cécile Pautas ◽  
Sandrine Hayette ◽  
...  

Abstract Abstract 659 Background: NPM1 mutations and WT1 expression are frequently identified in acute myeloid leukemia (AML). Both markers have been reported as suitable molecular target for minimal residual disease (MRD) monitoring. Recently, Gemtuzumab Ozogamicin (GO) has been shown to improve event free survival and overall survival in adult AML. Aims: To follow MRD in AML patients with NPM1 mutations (NPM1mut) and/or WT1 expression (WT1+) and treated in the randomized ALFA 0701 GO trial conducted by the Acute Leukemia French Association (Castaigne et al, Lancet 2012), in order to validate their prognostic significance and to evaluate the effect of GO on MRD response. Methods: The ALFA 0701 trial included 278 patients aged 50–70 years with newly-diagnosed de novo AML, randomized to receive or not five doses of 3 mg/m2 GO, on day 1, 4 and 7 of standard induction chemotherapy and on day 1 of two consolidation chemotherapy courses, respectively. A total of 77 NPM1mut (35 in control arm, 42 in GO arm) and 178 WT1+ (87 in control arm, 91 in GO arm) AML patients were studied. WT1 + at diagnosis was defined as a ratio WT1/ABL transcript above 25% in bone marrow (BM) or 5% in peripheral blood (PB) samples. PB and BM samples were collected at diagnosis, after induction and after each consolidation course. MRD levels were assessed using cDNA-based real-time quantitative PCR (RQ-PCR) and reported as the ratio NPM1mut or WT1 transcript/100 ABL transcript. Occurrence of a MRD level below the threshold (NPM1mut <0.1% and WT1 transcript < 0.5% in PB) was defined here as a good MRD response. The prognostic value of MRD was assessed by comparing the outcome of good versus poor MRD responders after induction in terms of cumulative incidence of relapse (CIR) at 18 months. Effect of GO was then evaluated by comparing MRD response in both treatment arms. Results: 178 patients (favorable cytogenetics: 4%, intermediate: 69%, unfavorable: 27%) were WT1+. WT1 transcript levels at diagnosis had no prognostic impact on complete remission (CR) rate or CIR. In the 104 patients who achieved CR, post-induction WT1 MRD level was predictive of relapse: 18-month CIR was 70% (95%CI: [49% - 84%]) in poor responders (n=31) versus 38% [26% - 50%] in good responders (n=73) (p=0.0001). When adjusted on cytogenetics, randomization arm and FLT3-ITD status, WT1 MRD remained independently associated with relapse (HR: 2.43, [1.25 – 4.75], p=0.009). Similarly, NPM1mut transcript levels at diagnosis had no prognostic impact on CR rate or CIR. Among the 67 NPM1mut patients who achieved CR, post-induction NPM1mut MRD level was predictive of relapse: 18-month CIR was 64% [45% – 70%] in poor responders (n=50) versus 25% [10% – 42%] in good responders (n=17) (p=0.0007). When adjusted on randomization arm and FLT3-ITD status, NPM1 MRD remained independently associated with relapse (HR: 4.2, [1.2 – 14.3], p=0.02). We were able to evaluate both MRD markers in 53 patients after induction (NPM1+/WT1+ n=16, NPM1+/WT1- n=24, NPM1-/WT1- n= 13). 18-month CIR was significantly different in the 3 groups (respectively 69% [38% - 87%], 40% [19% - 60%] and 25% [5% - 51%], p=0.008). In patients with poor MRD response assessed by NPM1mut, the presence of a positive WT1 MRD level was associated with a higher CIR. NPM1 MRD good response was more frequently observed in GO arm as compared to control arm (OR: 4.38 [2.12 – 9.03], p<0.0001). The rate of good NPM1 MRD response was 41% versus 7% after induction, 76% versus 35% and 93% versus 62% after first and second consolidation in GO and control arms respectively. WT1 MRD good response was more frequently observed in GO arm, although not reaching statistical significance (OR: 1.84 [0.89 – 3.77] p=0.098). The rate of good WT1 MRD response was 76% versus 65% after induction, 87% versus 66% and 87% versus 80% after first and second consolidation, respectively in GO and in control arms (figure 1). Conclusion: Our study confirms the predictive value of MRD based on NPM1 mutations and WT1 expression in patients treated in the ALFA 0701 randomized trial. In multivariate analysis, WT1 and NPM1 MRD levels remained independent prognostic factors for relapse. We showed that treatment with GO significantly improved MRD response in adult AML. Disclosures: Castaigne: Wyeth: Consultancy.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3532-3532
Author(s):  
Sung-Chao Chu ◽  
Tso-Fu Wang ◽  
Yu-Chieh Su ◽  
Ruey-Ho Kao ◽  
Yi-Fung Wu ◽  
...  

Abstract Abstract 3532 Introduction: The study is to analyze the prognostic impact of post-induction-BM status combining minimal residual disease (MRD), neutrophil/monocyte maturation return to MDS and quantitation of hematogones in adult AML achieving first morphologic complete remission (mCR). The hypothesis is that the detection of aberrant myeloid progenitor cells, aberrant neutrophil/monocyte maturation and hematogones in post-induction BM by flow cytometry may predict the outcome of AML in mCR even without diagnostic specimen. The positive MRD and the aberrant neutrophil/monocyte maturation will predict worse prognosis. In contrast, positive hematogones will predict better prognosis. Methods: Multidimensional flow cytometry was performed on bone marrow specimens from 47 consecutive non-M3 AML patients who had achieved mCR after standard 3+7 induction treatment. The in-remission immunophenotypic evaluation of BM was done before first consolidation treatment. The aberrant myeloid progenitor cells and aberrant neutrophil/monocyte maturation were defined by the flow cytometric scoring system (FCSS)1. Two investigators were blinded to corresponding pathologic, clinical or diagnostic flow cytometric data during initial analysis phase. After reaching agreement of the three parameters: aberrant myeloid progenitor cells (MRD ≥ 0.2% or not), aberrant neutrophil/monocyte maturation (FCSS ≥ 2 points or not) and hematogones (stages I / II B lymphoid progenitor cells ≥ 0.02% or not), those prognostic impacts on leukemia free survival (LFS) and overall survival (OS) were analyzed retrospectively. Results: Table 1 summarizes the clinical characteristics of patients according to the status of MRD and hematogones. Nine (19%) patients who had MRD ≥0.2% had a significantly worse median LFS (9.0 months vs not reached; P =.006) and worse OS (24.0 months vs not reached; P =.059). Fourteen (30%) patients who had hematogone levels ≥ 0.02% had a significantly better median LFS (not reached vs 13.5 months; P =.045) and a trend of better OS (not reached vs 24.0 months; P =.070). Six (13%) patients who had FCSS ≥ 2 points had a worse median LFS (8.0 vs 48.0 months; P =.052) but not significantly worse OS (17.0 vs 28.0 months; P =.804). A Spearman coefficient for MRD ≥0.2% and hematogone levels ≥ 0.02% was -0.317 (P <.029), indicating a mildly negative correlation. However, MRD ≥0.2% and hematogones ≥ 0.02% were almost mutually exclusive. For clinical convenience, a MRD/Hematogone score was proposed that patient with MRD ≥0.2% was defined as 1 point and hematogone ≥ 0.02% was defined as -1 point. All patients could be categorized into three subgroups: 14(30%) patients with -1 point, 24(51%) patients with 0 point and 9(19%) with 1 point. Patients were stratified based on MRD/Hematogone score to assess survival using the Kaplan-Meier method and the Log-rank test. The median LFS were not reached, 48 months and 9 months for patients with -1 point, 0 point and 1 point, respectively (P =.015; Fig. 1). The median OS were not reached (-1 point), not reached (0 point) and 24 months (1 point) (P =.088). Multivariate analyses using Cox proportional hazard model demonstrated MRD/Hematogone score was an independent predictor of LFS (HR = 2.5, 95% CI, 1.2–5.1, P =.016; Table 2) after adjusting for MRC cytogenetic classification, WBC count (above or below 50000/uL) and whether undergoing allogenetic hematopoietic stem cell transplant before 1st relapse or not. Conclusion: Even without a diagnostic specimen, flow cytometry can assess and quantify aberrant myeloid progenitor cells, aberrant neutrophil/monocyte maturation and hematogones in post-induction marrow. MRD ≥0.2% or neutrophil/monocyte abnormalities (FCSS ≥ 2) predicts shorter LFS. Hematogones ≥ 0.02% predicts longer LFS and correlates negatively with MRD ≥0.2%. Proposed MRD/Hematogone score for post-induction adult AML in mCR maybe offer a simple and practical risk assessment. And the score is worthy to be validated by prospective and larger scale study. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4969-4969
Author(s):  
Ihab A. Eldessouki ◽  
Ola Khorshid ◽  
Eman Kandeel ◽  
Nasr Lahloubi

Abstract Background The achievement of complete hematologic remission (CR) is used as predictor for treatment response in patients with myeloid leukemia (AML).However <5% blasts in the bone marrow does not reflect the presence of tumor burden precisely. Minimal residual disease (MRD) in the first complete remission (CR1) may play a critical rule in assessment of treatment response and prediction of subsequent relapse. Patients and Methods Leukemia associated immunophenotyping (LAIP) for 73 patients with denovo AML monitored at diagnosis , day 14 and day28 post-induction by multiparametric flow cytometry (MFC). Results CR achieved in 60(82%) patients and 13(18%) patients did not. Among the 60(80%) patients who achieved CR 9 (15%) were MRD negative and 51(85%) were MRD positive at day14. Significant association between MRD detection and disease free survival (DFS) using 0.01% cut off value (P=.015). Day 28 post induction show highly significant association between MRD and DFS using 0.01% cut off value (P=0.001) as 38(63%) patients were MRD negative and (27%) were positive. Significant association between MRD detection and overall survival (50 month) at day 14 and day 28 (P=0.02, P=0.001) respectively using cut off value 0.01%. MRD was positive in 63(86%) at day 14 and (37%) at day 28. Conclusion MRD detection at day28 and d14 at the end of induction in patients in CR may have a prognostic significance on clinical outcome and may thus be a useful marker for risk stratification. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 543-543
Author(s):  
John Ahman Liu-Yin ◽  
Sarah B. Daly ◽  
Michelle A. Sale ◽  
Stuart Green ◽  
Khalid Tobal ◽  
...  

Abstract The clinical value of serial Minimal Residual Disease (MRD) monitoring in core binding factor (CBF) positive patients was prospectively assessed in the AML-15 Trial which opened in July 2002. The trial compared 3 induction regimens (DA V/S ADE V/S FLAG Ida), followed by randomisation in consolidation (courses 3 and 4) to either MACE or 2 doses of Ara-C (3g/m2 or 1.5g/m2) and to stop or have a 5th course (Ara-C 1.5g/m2). Patients were also randomised to receive Gemtuzumab Ozogamicin (3mg/m2) at induction and/or consolidation. Over 2500 patients have so far been recruited, with 271 CBF patients (155 t(8;21), 116 inv (16)). Complete remission (CR) and relapse rates (RR) at 4 years were 95% and 19% respectively. CBF transcripts (AML1-ETO for t(8;21), CBFB-MYH11 for inv(16)) from bone marrow (BM) and peripheral blood (PB) were measured by real-time quantitative PCR (RQ-PCR) on the 7900 HT ABI machine, at presentation, after each course of chemotherapy and 3 monthly during remission for 2 years. CBF copies were normalised to ABL gene and expressed per 105ABL. The sensitivity of the RQ-PCR assay was 10−5. Data were analysed in 47 relapsed patients and in 92 patients who were in remission for >1 year. In 66 patients, where the reduction of initial CBF transcript level in BM, following induction chemotherapy, was measured, only 1 of 32 patients with >3 log reduction at remission whereas 20/34 patients with <3 log reduction have relapsed, giving relapse rates of 3% and 61% respectively, (2p<0.00001). With respect to BM post induction transcript levels, in the t(8;21) group (n=50), patients with <500 AML1-ETO copies had a 18% RR compared to 62% for patients with >500 copies (2p=0.003) and in the inv (16) patients (n=38), the RR were 8% and 58% respectively for CBFB-MYH11 copies lower or higher than 100 (2p=0.004). After consolidation and during remission, BM and PB transcript levels were also highly predictive of relapse risk. In t(8;21) patients, all 7 with BM AML1-ETO level >500 copies but only 3/45 patients with <500 copies relapsed (RR 100% V/S 9%, 2p<0.0001). Moreover all 12 patients with PB level >50 copies and only 2/52 patients negative for or with <50 AML1-ETO copies relapsed (RR 100% V/S 4%, 2p <0.00001). In inv (16) patients, 13/13 with >100 CBFB-MYH11 copies in BM and 3/25 patients with <100 copies relapsed (RR 100% V/S 9%, 2p<0.00001). In PB, any positive level resulted in relapse in 18/18 patients compared to 2/29 (RR 7%) with a negative MRD for CBFB-MYH11 (2p<0.00001). The interval between molecular and clinical relapse was ≥3 months and there was a significant correlation between BM and PB MRD levels post induction and at first positivity after consolidation (r>0.40, p<0.05). We conclude that MRD monitoring in CBF AML allows risk stratification based on treatment response, and can predict relapse, thus opening the way to risk-directed or pre-emptive therapy. We propose that MRD monitoring by RQ-PCR should be an integral part of the management of CBF positive AML.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 245-245 ◽  
Author(s):  
Ruth Mary de Tute ◽  
Andy C Rawstron ◽  
David A Cairns ◽  
Charlotte Pawlyn ◽  
Faith E Davies ◽  
...  

Abstract Introduction. Minimal residual disease (MRD) is a powerful predictor of outcome in multiple myeloma (MM). We have previously demonstrated, in transplant eligible patients, that the level of MRD as a continuous variable independently predicts both PFS and OS, with approximately a one year median OS benefit per log depletion (J Clin Oncol 2013; 31:2540-7 and Blood 2015; 125:1932-5). The impact of MRD also appears to be independent of therapy received. There is more limited data on the applicability of MRD assessment in transplant ineligible patients, largely as a consequence of low rates of CR historically within this patient cohort. Patients and Methods. In this analysis we have assessed the impact of MRD on PFS amongst patients treated within the non-intensive arm of the NCRI Myeloma XI trial. Patients were randomised between thalidomide (CTDa) and lenalidomide (RCDa) based induction therapies with responding patients being subsequently randomised to maintenance with lenalidomide monotherapy, or no further therapy. Bone marrow aspirates were obtained at the end of induction and this analysis represents a subset of 297 patients (median age 74 years). MRD was assessed using flow cytometry (sensitivity 10-4) with a minimum of 500,000 cells evaluated with six-colour antibody combinations including CD138/CD38/CD45/CD19 with CD56/CD27 in all cases and CD81/CD117 in additional cases as required. Results. Overall MRD-negativity was demonstrated in 41/297 (13.8%). When considered according to induction therapy received 25/154 (16.0%) of patients randomized to RCDa were MRD-negative compared to 16/143 (10.8%) of those randomized to CTDa (p=0.24; Fisher's exact test). MRD-negativity was associated with a significant outcome advantage as the median PFS was 34 months versus 18 months for MRD-positive patients (p<0.0001, HR 0.44 [95% confidence interval (CI 0.29-0.67)]). This effect was noted in both RCDa (median PFS 17m v 32m; p=0.001, HR 0.41 [95%CI 0.23-0.69]) and CTDa (median PFS 19m v 34m; p=0.03, HR 0.49 [95%CI 0.26-0.95]). When the impact of MRD was assessed according to induction regimen the outcome of MRD-negative and MRD-positive patients was similar with both regimens (see figure). The impact of MRD was also assessed as a continuous variable across 5 logs of residual disease. Sequential improvements in outcome with each log reduction were demonstrable. Median PFS for the following disease levels; <0.01%, 0.01 - <0.1%, 0.1% - <1%, 1% - <10% and >/=10% were 34, 26, 16, 14 and 9 months respectively (p<0.0001). This pattern was demonstrable in both RCDa and CTDa treated patients (p<0.0001 for both). Multivariate analysis confirmed the independent predictive value of MRD both as a qualitative and continuous quantitative variable (p<0.0001 for both). In both instances achieving an immunofixation-negative CR was not a significant prognostic variable when included in the model with MRD. Conclusions. We would conclude that MRD is a powerful predictor of outcome in transplant ineligible patients and is a meaningful therapeutic goal in this patient group. In contrast to conventional CR it retains independent prognostic significance both as a quantitative and qualitative variable. This data further supports the role of MRD as a primary endpoint and surrogate marker for survival in future clinical trials. Figure. Figure. Disclosures Rawstron: Janssen: Research Funding; BD Biosciences: Other: Remuneration; Gilead: Consultancy, Honoraria, Research Funding; GlaxoSmithKline: Honoraria; Genzyme: Honoraria; AbbVie: Honoraria; Roche: Honoraria; Celegene: Honoraria. Pawlyn:Celgene: Consultancy, Honoraria, Other: Travel Support; Takeda Oncology: Consultancy. Davies:Janssen: Consultancy, Honoraria; Celgene: Consultancy, Honoraria; Takeda: Consultancy, Honoraria. Kaiser:Amgen: Consultancy, Honoraria; Celgene: Consultancy, Honoraria, Research Funding; Janssen: Honoraria; Takeda: Consultancy; Bristol-Myers Squibb: Consultancy, Other: Travel support; Chugai: Consultancy. Jones:Celgene: Honoraria, Research Funding. Cook:Janssen: Consultancy, Honoraria, Research Funding, Speakers Bureau; Glycomimetics: Consultancy, Honoraria; Takeda: Consultancy, Honoraria, Research Funding, Speakers Bureau; Amgen: Consultancy, Honoraria, Research Funding, Speakers Bureau; Bristol-Myers Squibb: Consultancy, Honoraria; Sanofi: Consultancy, Honoraria, Speakers Bureau; Celgene: Consultancy, Honoraria, Research Funding, Speakers Bureau. Jenner:Janssen: Consultancy, Honoraria, Other: Travel support, Research Funding; Celgene: Consultancy, Honoraria, Research Funding; Novartis: Consultancy, Honoraria; Amgen: Consultancy, Honoraria, Other: Travel support; Takeda: Consultancy, Honoraria, Other: Travel support. Drayson:Abingdon Health: Equity Ownership, Membership on an entity's Board of Directors or advisory committees. Jackson:MSD: Consultancy, Honoraria, Speakers Bureau; Celgene: Consultancy, Honoraria, Other: Travel support, Research Funding, Speakers Bureau; Roche: Consultancy, Honoraria, Speakers Bureau; Amgen: Consultancy, Honoraria, Speakers Bureau; Janssen: Consultancy, Honoraria, Speakers Bureau; Takeda: Consultancy, Honoraria, Other: Travel support, Research Funding, Speakers Bureau. Morgan:Celgene: Consultancy, Honoraria, Research Funding; Takeda: Consultancy, Honoraria; Bristol Meyers: Consultancy, Honoraria; Janssen: Research Funding; Univ of AR for Medical Sciences: Employment. Owen:Celgene: Consultancy, Honoraria, Research Funding; Takeda: Honoraria, Other: Travel support; Janssen: Consultancy, Other: Travel support.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi218-vi218
Author(s):  
Maleeha Qazi ◽  
Sabra Salim ◽  
Kevin R Brown ◽  
Neil Savage ◽  
Nicholas Mickolajewicz ◽  
...  

Abstract Recurrence of solid tumors renders patients vulnerable to a distinctly advanced, highly treatment-refractory disease state that has an increased mutational burden and novel oncogenic drivers not detected at initial diagnosis. Improving outcomes for recurrent cancers requires a better understanding of cancer cell populations that expand from the post-therapy, minimal residual disease (MRD) state. We profiled barcoded tumor cell populations through therapy at tumor initiation/engraftment, MRD and recurrence in our therapy-adapted, patient-derived xenograft models of glioblastoma (GBM). Tumors showed distinct patterns of recurrence in which clonal populations exhibited either a priori, pre-existing fitness, or equipotent fitness acquired through therapy. Characterization of the MRD state by single-cell and bulk RNA sequencing revealed a tumor-intrinsic immunomodulatory signature with strong prognostic significance at the transcriptomic level and in proteomic analysis of cerebrospinal fluid (CSF) collected from GBM patients at all stages of disease. Our results provide insight into the innate and therapy-driven dynamics of human glioblastoma, and the prognostic value of the interrogating of the MRD state in solid cancers.


2014 ◽  
Vol 6 (1) ◽  
pp. e2014062 ◽  
Author(s):  
Orietta Spinelli ◽  
Manuela Tosi ◽  
Barbara Peruta ◽  
Marie Lorena Guinea Montalvo ◽  
Elena Maino ◽  
...  

Acute lymphoblastic leukemia (ALL) is curable in about 40-50% of adult patients, however this is subject to ample variations owing to several host- and disease-related prognostic characteristics. Currently, the study of minimal residual disease (MRD) following induction and early consolidation therapy stands out as the most sensitive individual prognostic marker to define the risk of relapse following the achievement of remission, and ultimately that of treatment failure or success. Because substantial therapeutic advancement is now being achieved using intensified pediatric-type regimens, MRD analysis is especially useful to orientate stem cell transplantation choices. These strategic innovations are progressively leading to greater than 50% cure rates. 


Sign in / Sign up

Export Citation Format

Share Document