Flow Cytometric Minimal Residual Disease Levels After First Inducton Can Define T-Acute Lymphoblastic Leukemia Patients with High Risk of Relapse

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4817-4817 ◽  
Author(s):  
Veselka Nikolova ◽  
Velizar Shivarov ◽  
Ricardo Morilla

Abstract Abstract 4817 T-cell acute lymphoblastic leukemia (T-ALL) patients have increased risk for treatment resistance and early relapse. The precise bone marrow evaluation for the presence of minimal residual disease (MRD) is essential for guiding treatment options. This requires techniques more sensitive than the level of sensitivity of light microscopic technique such as multicolour flow cytometry (FCM). Immunophenotypic alterations called leukemia associated immunophenotypic patterns (LAIP) (i.e.aberrant myeloid markers) and ectopic phenotypic expression (i.e. appearance of immature phenotypes such as TdT, CD1a and CD3 outside their normal site in the thymus) are of benefit to track the residual leukemic cells in T-ALL. A retrospective data analysis of MRD was done comprising T-ALL patients diagnosed and followed-up at the Institute of Cancer Research/Royal Marsden Hospital by means of 3-colour flow cytometry (3C FCM).The aim was to answer a question whether the 3C FCM can reliably split patients into two groups (positive, MRD+ and negative, MRD-) and predict a subsequent relapse and to define a right time point for performing MRD tests. Eight patients were enrolled in the study following the inclusion criteria: (i) complete remission after 1st induction phase of chemotherapy, (ii) presence of LAIP or an ectopic phenotypic expression, and (iii) monitored at defined time points after initial treatment. MRD was measured during the first year of treatment as follows: at the end of phase 1 induction (day 29–35, MRD1), before the start of consolidation (3 months, MRD2), after consolidation (MRD3), during the maintenance therapy (12 months, MRD4). Immunophenotyping was performed on lysed-washed bone marrow samples using CD45 gating strategy and originally defined blast gates at diagnosis. The phenotypes to be followed-up included: TdT+/CytCD3+, CD34+/CYTCD3+, TdT+/CD2+, CD8+/CD10+, CD2+/CD10+, CD7+/CD10+, CD7+/CD33+, CD7+CD34+. Patients were divided into 2 groups in relation to subsequent relapse. Group 1 included 6 patients without relapse. Patient characteristics of the group were: male:female 5:1, mean age 17.7 years, overall survival (OS) 59 months, relapse free survival (RFS) 85 months. Group 2, relapsed patients, included 2 men, mean age 56 years, OS 13 months, RFS 8.5 months. According to the EGIL classification system the 2 men in Group 2 were with an early T-precursor phenotype, whilst Group 1 was heterogenous but cortical-T-ALL predominated. Cytogenetics/FISH and RQ-PCR studies were performed at diagnosis and showed normal karyotype in only one of the Group 2 patients. MRD results showed a difference between the two groups as regards MRD1 and MRD2 time points. Group 1 patients had negative or low MRD levels (below 0.18%) in their MRD1 bone marrow - MRD-, n=4 and MRD+,n=2 (0.18% and 0.12% respectively, sensitivity 0.04%). Those of them who were tested at MRD2 and MRD3 were negative. Both patients in Group 2 showed higher levels of MRD positivity at MRD1 (1% of total bone marrow cells), the first one of them also being positive at MRD2 and the second one becoming MRD+ at MRD4 time point. Although turning to MRD- at MRD3 time point both Group 2 patients relapsed 2.5 and 4.5 months, respectively, after the end of consolidation treatment. Additionally, Group 1 patients had a significantly longer RFS than Group 2 (median 58 months RFS vs. 8.5 months; P <0.001). Conclusions: Reliable detection of MRD in T-ALL is possible by 3C FCM using a combination of TdT and a T cell marker (cytCD3 or mCD3) as such a combination is normally found exclusively in the thymus. The higher MRD-positive levels in Group 2 reflect the more resistant disease in this group and higher probability of early relapse and shortened overall survival. Early T-cell precursor phenotype in these patients appeared to be a subtype at very high risk for treatment failure irrespective of the lack or the presence of genetic lesions. Based on MRD positivity above 0.18% at time points MRD1 or both MRD1 and MRD2 these patients need reassessment of treatment options and more intensive therapy has to be considered for relapse prevention. Finally, the results of our retrospective study suggest the usefulness of implementation of MRD testing by FCM for taking clinical decisions in the prospective clinical trials for novel therapies for T-ALL. Acknowledgments: The study was supported by the Union for International Cancer Control, Geneva, Switzerland (Grant ICRETT-080–2011) Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2494-2494
Author(s):  
Myriam Ruth Guitter ◽  
Jorge Gabriel Rossi ◽  
Elisa Sajaroff ◽  
Carolina Carrara ◽  
Pizzi Silvia ◽  
...  

Abstract Introduction: Despite the advances observed in the outcome of pediatric acute lymphoblastic leukemia (ALL) treatment during the last 20 years, relapse remains the most common cause of treatment failure in childhood ALL. Several factors have been associated to the prognosis of these patients; however, minimal residual disease (MRD) emerges as a relevant predictor of outcome. Objectives: The aims of this study were to assess MRD by flow-cytometry in relapsed ALL and to evaluate its prognostic impact as a predictor factor of outcome at the end of the induction therapy and prior to hematopoietic stem cell transplantation (HSCT). Patients and Methods: From Aug'10 to Jun'15, 123 ALL patients were treated at our center. MRD determination at least at two time-points during relapse treatment was a requirement for considering a patient eligible for the present study. Sixty-six cases were excluded due to the following causes: 10 patients died during induction, 2 died early in complete remission (CR), 29 did not respond to chemotherapy, in 13 patients MRD determination was not performed: 4 did not have clinical data available, 4 patients were Down Syndrome and 4 children received treatment for relapse in other centers. Thus, fifty-seven patients achieved CR and were evaluated for MRD at two time points. Of them, 56 patients belonged to S4 and S3 and 1 patient to S1 group as defined by the Berlin-Frankfurt-Münster stratification for relapsed ALL. MRD was analyzed by multiparametric flow-cytometry following ALL-IC 2009 guidelines. Negative MRD was defined as disclosing less than 0.1% of blasts. For this analysis, patients were stratified based on MRD levels at two different time points: after end of induction, before HSCT or at any other time point during the follow-up for patients who did not undergo HSCT. Three groups were defined: Group-1: negative at both time points (n= 23), Group-2: positive at 1 time point (n= 13) and Group-3: positive at both time points (n= 21). Patients who relapsed before receiving HSCT were considered Group-3. Twenty-five patients underwent HSCT: 13 of them from Group-1, 9 from Group-2 (2 had positive MRD previous to receive HSCT) and 3 patients from Group-3. HSCT was performed with matched familiar donor in 16 cases and matched unrelated donor in 9 cases. Results: The distribution of events according to receiving or not HSCT was: 5 died due to transplant related mortality (TRM), 9 relapsed after receiving HSCT and 16 during treatment with chemotherapy. With a median follow-up of 16 (range: 6-67) months, overall 3-year EFS probability (EFSp) (SE) was 32 (8)%. The 3-year EFSp was 75 (11)% for Group-1, 24 (14)% for Group-2 and 0% for Group-3 (p-value <0.00001). Comparing patients who did not receive HSCT vs. patients who did, EFSp (SE) was 32 (12)% and 29 (11)% respectively (p-value: non-significant). The EFSp (SE) according to MRD groups in patients who underwent HSCT was: Group-1: 53 (19)%, Group-2: 14 (13)% and 0% for Group-3 (p-value: 0.06). Conclusions: MRD quantification by flow-cytometry demonstrated to be a significant prognostic factor for relapsed ALL. Both, TRM and death in CR rates, were high and should be decreased by improving supportive measures. MRD determination by flow-cytometry in patients who underwent HSCT showed a trend to achieve a better EFSp, thus representing a relevant tool for stratifying relapsed ALL patients in order to achieve a better selection of patients to receive HSCT. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 322-322
Author(s):  
Jean-Michel Cayuela ◽  
Paola Ballerini ◽  
Marina Romeo ◽  
Vahid Asnafi ◽  
Marie-Francoise Auclerc ◽  
...  

Abstract TEL-AML1 fusion transcripts are found in 25% of children with B-cell precursor ALL (BCP-ALL). From June 1993 to December 1999, 1195 children with BCP-ALL were included in the FRALLE 93 protocol. Out of these, 792 were evaluated for TEL-AML1 transcript expression. There is no difference in terms of initial features, DFS, EFS, survival between evaluated (792) and non evaluated (403) patients. Out of the 792 pts, 191 (24%) expressed TEL-AML1 transcripts at diagnosis. To assess the potential prognostic value of TEL-AML1 transcripts quantification, we have retrospectively analysed follow up marrow samples using Europe Against Cancer procedures for real time quantitative RT-PCR assay, on ABI PRISM 7700 (2 reference labs) and Light Cycler apparatus (1 reference lab). Out of the 191 TEL-AML1+ve pts, 83 were evaluated for MRD at different time points after induction therapy (median = D41 (34–55) (53 evaluable pts), at D111 (62–158) (62 pts), at D216 (159–325) (33 pts) and at D838 (365–1287) (49 pts). According to normalized Ct values, samples were attributed to 4 MRD level ranging from 0 to 3 and defined as follows: 0: Ct>40 ; 1 : 36<Ct≤40 ; 2 : 33<Ct≤36 ; 3 : Ct≤33, corresponding respectively to undetectable MRD ; MRD<10-4 ; 10-4≤MRD<10-3 ; MRD≥ 10-3, with respect to dilution of REH cDNA. Distribution of pts according to MRD level at different time points after induction treatment are summarized in the following table. Seventeen relapses have occurred at a median time of 41 months (17–73)(bone marrow: 7, BM + other: 5, testis: 3, CNS: 2). A level 2 positivity at the end of induction was associated with an increased risk of relapse of 3.31(95%CI:1.02 – 10.76, p =.047) while level 3 positivity was associated with a relative risk of 9.52 (95%CI: 2.91 – 31.08, p =.0002). Positivity at D111 was associated with an increased risk of relapse of 8.6 (2.0 – 38.5, p = 0.0042), whatever the level. Combination of data obtained at D41 and D111 allows to distinguish 3 subsets of pts with decreasing relapse-free survival: from 97.5% (95%CI: 85–100%) in pts with no positivity at D111 whatever the D41 result, to 75% (95%CI: 58–92%) in pts with MRD +ve at D111 with low level at D41 and 42% (95%CI: 14–69%) in pts with MRD +ve at D111 with level 2 or 3 at D41 (p<.0001). No other prognostic factor was found (age, sex, WBC, D8 steroid response, D21 bone marrow response) which renders the MRD profile unique in this matter. Conclusion: RQ-PCR-based MRD detection is a powerful prognostic tool in TEL-AML1+ve leukemia. Combination of two time points allows a relevant stratification of pts according to the risk of relapse, compatible with clinical decision making towards intensification or deescalation in the setting of controlled trials FU time point Number of pts in MRD classes (number of relapses) 0 1 2 3 Not evaluated D41 27 (3) 11 (2) 10 (4) 5 (4) 30 (4) D111 40 (2) 14 (6) 7 (2) 1 (1) 21 (6) D216 29 (2) 2 (1) 1 (0) 1 (1) 50 (13) D838 47 (8) 1 (0) 1 (1) 0 34 (8)


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 701-701
Author(s):  
Yousif Matloub ◽  
Paul S. Gaynon ◽  
Somasundaram Jayabose ◽  
Bruce C. Bostrom ◽  
Stephen P. Hunger ◽  
...  

Abstract Three thousand fifty four children with NCI SR ALL were enrolled on CCG-1991; 2075 eligible patients were randomized and began treatment with intrathecal cytarabine, vincristine (V), dexamethasone (DX), and pegylated asparaginase (ASP). Bone marrow status was assessed at Day 7 and 14, and 28 of Induction. Slow early responders (SER’s) (Day 7/14 M3-M3, or M3-M2; and M2 at Day 28 Induction) received rescue daunorubicin and were assigned to augmented BFM therapy (N Engl J Med1998; 338:1663). Other patients were designated rapid early responders (RER’s) and randomly allocated to V/ intravenous methotrexate versus oral 6MP/oral methotrexate, and DX pulses in months 3–4 and 7–8 of therapy and single or double delayed intensification. The 5-year EFS for RER’s and SER’s was 90.5% (SE ± 1.0%) and 84.7% (SE = ±3.7%). Eight hundred three patients elected to participate in a companion study of minimal residual disease (MRD), which was successfully performed on BM samples of 750 patients (93.4%). Out of 1362 BM submitted samples, 1360 were successfully tested. MRD was assessed by real-time quantitative PCR of clone-specific immunoglobulin heavy chain, immuno-globulin kappa deleting element, and T-cell receptor gene rearrangements on Day 14 Time Point (TP #1) for patients not achieving M1 status at Day 7, end Induction (TP #2) and Day 84 (RER’s) or Day 119 (SER’s) (TP #3), i.e., Day 28 of Interim Maintenance). Various levels of MRD positivity were explored for prognostic significance (see Table). At TP’s 1, 2, and 3, 14%, 57%, and 78% had undetectable MRD with sensitivity of 0.01% or better. At the three time points patients with detectable MRD were 2.7 to 4.3 times more likely to fail than patients with undetectable MRD. TP #1, however, unlike TP #2 and TP #3 was not predictive of EFS in our study. Patients who had MRD > 0.01% at TP#1 had a much lower EFS at 4 years if at TP #3 MRD persisted at > 0.01% vs ≤ 0.01% (78 ± 0.1% vs 94 ± 0.05%, p = 0.01). We assessed MRD by PCR at three TP’s in a homogeneous population of children with SR ALL receiving V/ DX/ASP. At end of induction, 57% of patients were MRD negative but still accrued 1/3 of adverse events. Time Point 1 Time Point 2 Time Point 3 MRD Day 14 (Induction Day 14) Day 28 (RER) or 35 (SER) (End Induction) Day 84 (RER) or 119 (SER) (Interim Maintenance Day 28) *Sensitivity < 0.01% “absolute” negative* 1/44 13/340 15/350 low positive < 0.01% 1/27 8/89 5/50 positive 0.01 %–0.1% 6/87 6/104 3/37 positive 0.1%–1% 6/91 4/43 4/9 positive > 1% 6/67 7/21 1/1 total 20/316 45/597 30/448


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2611-2611
Author(s):  
Yuko Sekiya ◽  
Yusuke Okuno ◽  
Hideki Muramatsu ◽  
Atsushi Narita ◽  
Kyogo Suzuki ◽  
...  

Abstract Purpose Next-generation sequencing (NGS)-based monitoring of minimal residual disease (MRD) was developed to increase the sensitivity and specificity of standard MRD detection methods. However, few published studies have tested the clinical utility of this novel technique. We assessed the clinical utility of NGS-MRD in a uniformly treated cohort of patients with pediatric B-cell precursor acute lymphoblastic leukemia (BCP-ALL). PATIENTS AND METHODS We enrolled 79 unselected patients with pediatric BCP-ALL. Bone marrow samples were collected at the time of diagnosis, on days 33 and 80, at pre- and post-maintenance therapy time points (4-5 and 24 months, respectively), and upon relapse. Genomic DNA was extracted from frozen bone marrow mononuclear cells at each time point. We used diagnostic samples to define the immunoglobulin heavy chain (IGH), complementarity-determining region 3 (CDR3), and T-cell receptor gamma chain (TCRG) loci. From these samples, we detected leukemia-specific CDR3 sequences in >5.0% of all sequence reads. In addition, we performed a multiplex polymerase chain reaction (PCR) to determine the IGH, CDR3, and TCRG loci and subsequently assessed MRD using NGS. The result was considered positive for NGS-MRD if the leukemia-specific CDR3 sequence was detected. The resulting positive MRD values were categorized as "low positive" (<10−4) or "high positive" (≥10−4). RESULTS We detected leukemia-specific CDR3 sequences in 72 of 79 patients (91%). MRD was measured in 232 samples and we obtained positive results in 59 samples. MRD was detected in 51% (28/55) samples on day 33, and the frequencies of positive MRD decreased to 25% (16/65), 19% (11/58), and 7.4% (4/54) samples at day 80, 4-5 months, and 24 months, respectively. Each of the four patients with a positive MRD at 24 months relapsed shortly after detection. In a univariate analysis, the MRD values at day 80 {risk ratio [RR; 95% confidence interval (CI)] = 7.438 (2.561-21.6), p < 0.001}, 4-5 months [RR (95% CI) = 10.24 (3.374-31.06), p < 0.001], and 24 months [RR (95% CI) = 19.26 (4.974-74.59), p < 0.001] showed a statistically significant association with inferior leukemia-free survival (LFS). The classification of patients as either low or high positive for NGS-MRD at day 80 was a significant risk factor for poor LFS [low positive, RR (95% CI) = 6.63 (2.01-21.82), p = 0.002; high positive, RR (95% CI) = 9.40 (2.32-38.17), p = 0.002]. Furthermore, both low and high positivity for MRD at 4-5 months was also a significant risk factor for poor LFS [low positive, RR (95% CI) = 10.32 (3.07-34.70), p < 0.001; high positive, RR (95% CI) = 10.04 (2.00-50.34), p = 0.005]. In an assessment of three multivariate Cox proportional hazard models, we found that both low and high positive NGS-MRD values at day 80 [low positive, RR (95% CI) = 6.05 (1.80-20.39), p = 0.0037; high positive, RR (95% CI) = 8.20 (1.92-35.07), p = 0.002] and at 4-5 months [low positive, RR (95% CI) = 12.98 (3.49-48.28), p < 0.001; high positive, RR (95% CI) = 23.16 (3.28-163.7), p < 0.001] were independent covariates predictive of poor LFS. CONCLUSION We detected leukemia-specific CDR3 rearrangements in 91% of our cohorts, which was comparable with the frequencies detected using sensitive real-time quantitative (RQ)-PCR methods. In both univariate and multivariate analyses, low and high positive NGS-MRD results were significantly associated with poor LFS. In addition, we found that MRD positivity at later time points (4-5 and 24 months) was predictive of a high incidence of relapse and poor LFS. Therefore, NGS-MRD can identify a greater number of patients who are at a high risk of relapse and candidates for intensified chemotherapy or allogeneic HSCT. Our study demonstrates the potential superiority of NGS over the current standard method of MRD monitoring. However, standardization, quality control, and validation of this new technology are warranted prior to its use in routine practice. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5358-5358
Author(s):  
Marion Eveillard ◽  
Nelly Robillard ◽  
Richard Garand ◽  
Fanny Rialland ◽  
Nicolas Blin ◽  
...  

Abstract The efficacy of induction chemotherapy in childhood acute lymphoblastic leukemia (ALL) is usually evaluated on day 35. However, at this stage, many patients have already begun to recover and present with a regenerative bone marrow (BM) where hematogones may make the identification of residual blast cells problematic both in morphology and in flow cytometry (FCM). In the FRALLE (French Acute Lymphoblastic Leukemia) trials, evaluation is proposed on days 8, 21 and 35. Here we evaluated whether FCM performed on day 21 (D21), when hematogones are still absent, would prove informative. The cohort reported here was constituted of 45 children aged between 1 and 20 years old (median 6) treated for ALL according to the FRALLE recommendations since 2006. There were 81% B-ALL, 17% T-ALL and 2% of mixed phenotype acute leukemia (MPAL, T/My). At diagnosis, the mean percentage of BM blasts was 50%. Classification according to the European Group for Immunophenotyping of Leukemia (EGIL) was 3 B-I, 21 B-II, 11 B-III, 2 B-IV and 1 T-I, 2 T-II and 4 T-III. Extensive immunophenotyping at diagnosis identified a median of 3 leukemia associated immunophenotypes (LAIP, range 1-5), defined as discriminant from hematogones. Corticosensitivity was defined on a complete blood count (CBC) as less than 1 G/L of blast cells on day 8. Chemosensitivity was assessed on a bone marrow aspiration at day 21, both morphologically (< 5% blasts) and in FCM (MRD0). Molecular biology (according to Biomed2) was performed on BM samples collected on days 35 (MRD1) and 70 (MRD2). Follow-up median time was 59 months (3-276). Corticosensitivity was observed for 39/43 patients (one had received corticosteroids for a tonsillitis before being referred and diagnosed with ALL and another one had less than 1 G/L of blasts at diagnosis). Five/44 patients were identified as chemoresistant by morphology on D21 (one aplastic sample). Enough cells were available for minimal residual disease (MRD) by FCM in 43 patients, on bone marrow collected on EDTA. As a mean, 586 328 total nucleated cells were acquired in FCM (range 9 616 - 1 751 000) thereby providing good sensitivity. Multiparameter FCM in 6 to 8 colors was performed on a single tube, customized according to each patient’s LAIP. Five MRD thresholds were defined as follows : level 1, >10-2 detected blasts; level 2, 10-3- <10-2detected blasts; level 3, 10-4- <10-3detected blasts; level 4, 10-5- <10-4 detected blasts or no event detected; level 5, <10-5detected blasts or no event detected. The table below indicates the partition of patients according to these MRD levels on D21. Table 1LevelPatient numbersDetectable MRDAbsence of MRD166028803660415695818 Event-free survival (EFS; Kaplan Meier Log rank test) was statistically significant (p=0.023) when comparing patients with level 4 or 5 MRD to the others. Level 3 patients had an intermediate EFS and OS. In an independent cohort of 79 patients with evaluable FMC MRD on day 21 from a different center, the same good prognosis value of MRD lower than 10-4at that time point was confirmed for EFS (p=0.03). Childhood ALL has become of relatively good prognosis with the progress in therapies. However, it is noteworthy that half the patients had detectable MRD at levels 1, 2 or 3, which probably prompted the clinicians to intensify treatment. However, below level 3, i.e.<10-4, FCM MRD on D21 is of excellent prognosis, even if detectable. In summary, this work demonstrates the feasibility of FCM evaluation on D21, with the advantage of not being complicated by the presence of hematogones. Moreover, interpretation remains delicate in morphology at this time point where BM smears may be very poor. FCM thereby provides precious early information on chemosensitivity with a single patient-adapted antibody combination. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Vol 17 (1) ◽  
Author(s):  
Andrea Rzepiel ◽  
Nóra Kutszegi ◽  
András Gézsi ◽  
Judit C. Sági ◽  
Bálint Egyed ◽  
...  

Abstract Background Treatment stratification based on bone marrow minimal residual disease (MRD) at set time points has resulted in considerably improved survival in pediatric acute lymphoblastic leukemia (ALL). Treatment response is assessed using bone marrow samples. MicroRNAs (miRs) easily traffic among fluid spaces and are more stable than most other RNA classes. We examined the role of circulating miRs as putative less invasive MRD biomarkers. Methods In an exploratory experiment, expression of 46 preselected miRs was studied in platelet-free blood plasma samples of 15 de novo, 5 relapsed ALL patients and 10 controls by Custom TaqMan Array Advanced MicroRNA Card. Based on their high expression in ALL compared to controls, and on the reduction observed along the induction therapy, four miRs were selected for further analyses: miR-128-3p, -181a-5p, -181b-5p and 222-3p. Their expression was measured by qPCR at 4 time points in 27 de novo ALL patients treated in the ALL IC-BFM 2009 study. Results The expression of all 4 miRs significantly decreased over the first week of therapy (miR-128-3p: log2 fold change − 2.86; adjusted p 3.6 × 10−7; miR-181b-5p: log2 fold change − 1.75; adjusted p 1.48 × 10−2; miR-181a-5p: log2 fold change -1.33; adjusted p 3.12 × 10−2; miR-222-3p: log2 fold change − 1.25; adjusted p 1.66 × 10−2). However, no significant further reduction in miR expression was found after the 8th day of therapy. Measured drop in expression of 2 miRs at day 8 strongly correlated with day 15 bone marrow flow cytometry MRD results (miR-128-3p: Pearson’s r = 0.88, adjusted p = 2.71 × 10−4; miR-222-3p: r = 0.81, adjusted p = 2.99 × 10−3). Conclusion In conclusion, these circulating miRs might act as biomarkers of residual leukemia. MiR-128-3p and miR-222-3p in blood predict day 15 flow cytometry MRD results 7 days earlier. Although, their sensitivity falls behind that of bone marrow flow cytometry MRD at day 15.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. e22000-e22000
Author(s):  
O. Bulent Zulfikar ◽  
Basak Koc

e22000 Background: The prognosis of children and adolescents with acute lymphoblastic leukemia (ALL) has dramatically improved. This success is associated with both by multiagent chemotherapy regimens and by definition of clinical, biological and treatment response that allow the administration of risk-adapted therapy, including allogeneic hematopoietic stem cell transplantation (HSCT). In treatment of relapsed and resistance ALL, allogeneic HSCT continues to play a major curative role. In the present study, we desciribed the patients who underwent HSCT in our clinic in the last 21 years. Methods: From 1999 to 2020, 147 patients who diagnosed with ALL and treated with the COG protocols at the Istanbul University Oncology Institute were retrospectively reviewed and 17 of them relapsed. HSCT was applied 7 of the relapsed cases and also 3 resistant cases who had suitable matched donors. The demographic features, laboratory findings and treatment responses of 10 patients were recorded from the patients’ medical records. Results: All 10 patients were B-ALL with median diagnosis age of 79.5 months (range: 32-195) and 5 were male. Characteristics of patients given in Table 1. HSCT was performed due to late relaps in 7 patients. Three of the 7 relaps were only bone marrow and other 4 had combined. Patient #2 had both breast, conjunctiva and bone marrow for the 1st relaps and only conjunctiva for the 2nd one and this patient had also t(9;22) in the 1st relaps. Patient #3 had bone marrow+central nervous system relaps and patient #7 had bone marrow for the 1st relaps and testis and bone marrow for the 2nd one. Other 3 had poor response to treatment and Minimal Residual Disease (MRD) was high in End of Introduction (EoI) and End of Consolidation (EoC)). All patients had allogeneic HSCT and 8 are alive. Conclusions: HSCT remains the standard-of-care treatment for ALL patients who carry high-risk features predicting leukemia recurrence and for those experiencing high-risk first relapse or multiple relapses. Additionally, defining the indications of HSCT are dynamic and it could change according to treatment options as well as new molecular and biological findings. It is important to identify the patients who have high relapse risk and HSCT should have priority in patients whom MRD is high in EoI and EoC.[Table: see text]


Blood ◽  
2021 ◽  
Author(s):  
Swantje Buchmann ◽  
Martin Schrappe ◽  
Andre Baruchel ◽  
Andrea Biondi ◽  
Michael J. Borowitz ◽  
...  

Comparison of treatment strategies in de novo pediatric acute lymphoblastic leukemia (ALL) requires standardized measures of efficacy. Key parameters that define disease-related events, including 'complete remission' (CR), 'treatment failure' (TF; not achieving CR), and 'relapse' (loss of CR) require an updated consensus incorporating modern diagnostics. We collected the definitions of CR, TF and relapse from recent and current pediatric clinical trials for the treatment of ALL, including the key components of response evaluation (timing, anatomic sites, detection methods, and thresholds), and found significant heterogeneity, most notably in the definition of TF. Representatives of the major international ALL clinical trial groups convened to establish consensus definitions. CR should be defined at a time point no earlier than at the end of induction (EOI), and should include the reduction of blasts below a specific threshold in bone marrow and extramedullary sites, incorporating minimal residual disease (MRD) techniques for marrow evaluations. TF should be defined as failure to achieve CR by a pre-specified time point in therapy. Relapse can only be defined in patients who have achieved CR, and must include a specific threshold of leukemic cells in the bone marrow confirmed by MRD, the detection of central nervous system leukemia, or documentation of extramedullary disease. Definitions of TF and relapse should harmonize with eligibility criteria for clinical trials in relapsed/refractory ALL. These consensus definitions will enhance the ability to compare outcomes across pediatric ALL trials, and facilitate development of future international collaborative trials.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 4618-4618
Author(s):  
Larisa Shelikhova ◽  
Maria Ilushina ◽  
Alexander Popov ◽  
Zhanna Shekhovtsova ◽  
Dmitriy Balashov ◽  
...  

Introduction Allogeneic hematopoietic stem cell transplantation (HSCT) is indicated for patients with relapsed or refractory acute lymphoblastic leukemia. Patients with persistence of minimal residual disease (MRD) before HCT are at increased risk of disease relapse. Multiparameter flow cytometry (MFC) is the most commonly used method of MRD detection in clinical practice. This study aimed to evaluate MRD status before HCT on outcome of ALL patients receiving allogeneic HSCT from haploidentical donors with TCRαβ+/CD19+ depletion of the graft. Materials and methods A total of 120 pts with ALL (T-lineage ALL (T-ALL)- 37, B cell precursor (BCP)-ALL-83, 45 female, 75 male, median age 8.7 years (0.5-20) underwent allogeneic HSCT between June 2013 and June 2019. All pts received Haplo graft and were in morphologic remission. Disease status at transplant was CR1 in 35 pts, CR2 in 68 pts and CR>2 in 17 pts. Transplantation in CR1 was performed according to risk stratification scheme in the current institutional ALL protocol (Moscow-Berlin 2008, 2015). MRD detection in the bone marrow prior to НSСТ was performed in all pts by MFC according the AIEOP BFM FLOW Network SOP. MRD negativity was defined as <0.001% of all bone marrow nucleated cells. Seventy-nine patients were MRD negative before HSCT, 41 were MRD-positive. The median MRD level (among MRD-positive patients) prior HCT was 0.025%. Thirty (25%) pts received treosulfan-based myeloablative preparative regimen, while TBI-based regimen was used in 90 (75%) pts. Two regimens of GvHD prophylaxis were used. Regimen 1 (n=27): thymoglobulin 5mg/kg, rituximab 200 mg/m2 and bortezomib on day +2, +5; regimen 2 (n=93): tocilizumab at 8 mg/kg on day -1 and post-transplant bortezomib, 89 pts receive additional abatacept at 10 mg/kg on day +2, +7, +14, +28. TCR αβ+/CD19+ depletion of HSCT with CliniMACS technology was implemented in all cases. The median dose of CD34+ cells was 9.3 x106/kg (range 4.3-19.8), αβ T cells - 30x103/kg (range 1-361). Median time of follow-up for survivors was 1.6 years (range: 0.13 - 4.8). Results Primary engraftment was achieved in 116 of 120 pts (3 pts died before engraftment due to septic events, one relapsed early), the median time to neutrophil and platelet recovery was 13 and 14 days, respectively. All engrafted pts had verified morphologic remission and achieved sustained complete donor chimerism by day +30, seven of them had detectable MRD (5 of them were MRD-positive before HCT). Transplant-related mortality was 5 % (95% CI: 2-11). The cumulative incidence (CI) of relapse at 1.5 years was 25%(95%CI:18-35) for the whole cohort. Among patients, who had MRD-negative remission prior to HSCT, CI of relapse was 14 % (95%CI:8-26) with median time of relapse of 0.54 months, as compared to MRD-positive cohort, with CI of relapse of 44 % (95%CI:31-63) with median time to relapse 0.29 months, p=0.0004. pEFS (event=death or relapse) was 70% (95%CI: 61-78) for the whole cohort, in MRD (-) group pEFS was 79% (95%CI: 68-88), as compared to 56%(95%CI:40-72) in the MRD(+) group, p=0.025. CI of relapse in BCP-ALL pts, who had MRD-negative remission prior to HCT was 12 %(95%CI:6-25), in MRD(+) group 49% (95%CI:33-72), p=0.0002, in T-ALL pts in MRD (-) and MRD (+) groups CI of relapse was 17 % (95%CI:6-51) and 38 % (95%CI:20-76), respectively, p=0.14. Conclusion These results suggest that MRD detection by multiparameter flow cytometry prior to HSCT is a highly significant prognostic factor in the setting of haploidentical HSCT on the platform of ab T cell depletion. We expect that further improvement of the outcome can be achieved based on the combination of current safe haplo HSCT platform and novel targeted immunotherapy approaches. Figure Disclosures Maschan: Miltenyi Biotec: Other: lecture fee.


Sign in / Sign up

Export Citation Format

Share Document