scholarly journals Superiority of Leukemic Stem Cell-Based Minimal Residual Disease Assay to Traditional Multiparameter Flow Cytometry-Based Method for Relapse Prediction in AML Patients: A Prospective Study with Randomized Training and Validation Sets

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 517-517
Author(s):  
Siqi Li ◽  
Lan-Ping Xu ◽  
Yu Wang ◽  
Xiaohui Zhang ◽  
Huan Chen ◽  
...  

Abstract Background: Minimal/measurable residual disease (MRD) determined by multiparameter flow cytometry (MFC) is an important variable for relapse prediction and treatment approach selection in patients with acute myeloid leukemia (AML). We aimed to investigate whether leukemia-stem cell (LSC)-based assay is superior to traditional MFC methods, including LAIP and D-F-N assays, for MRD evaluation in predicting clinical outcomes. Methods: In this cohort study, a total of 360 AML patients who received allogeneic stem cell transplantation (allo-SCT) between July 2018 and November 2019 were prospectively enrolled. The patients were randomized (1:1) and classified into a training set (n=180) and a validation set (n=180). Posttransplantation MRD were according to LSC based assay, mainly including a cocktail of CD7, CD11b, CD22, CD56, Tim-3, and CLL-1 on CD34 +CD38 - cells, and traditional assay determined by MFC, respectively. Findings: In the training set, patients were classified as LSC positive group (group A) and LSC negative group (group B) according to a cutoff value of CD34 +CD38 -cocktail + LSCs as 0.004%. Subjects in group A had a higher cumulative incidence of relapse (CIR, 42.7% vs. 2.6%, P<0.001) and comparable non-relapse mortality (NRM, 0% vs. 8.1%, P=0.154) compared with cases in group B, leading to inferior leukemia-free survival (LFS, 57.3% vs. 89.3%, P<0.001) and overall survival (OS, 70.8% vs. 90%, P=0.009) of cases in group A to group B. Multivariate analysis showed that positive LSCs after transplantation could independently predict CIR (P<0.001), LFS (P<0.001), and OS (P=0.021). The predictive value of positive LSCs following allo-HSCT for CIR (P<0.001), LFS (P<0.001), and OS (P=0.004) was further confirmed in the validation set. In the total case cohort, multivariate analysis also showed that positive LSCs after transplantation could independently predict CIR (HR=13.999, P<0.001), LFS (HR=5.429, P<0.001), and OS (HR=3.761, P=0.021). The total patients were classified into positive MRD and negative MRD groups according to the traditional MFC method. The results showed that the CIR of patients in the traditional MRD positive group was significantly higher than the CIR of patients in the traditional MRD negative group (44.9% vs. 7.3%, P<0.001), leading to lower LFS (55.1% vs. 85.6%, P<0.001) and OS (55.6% vs. 87.5%, P<0.001). Compared with MRD detected by the traditional MFC method, using LSCs for MRD evaluation has high sensitivity (66.7% vs. 43%), a high C-index (0.76 vs. 0.69) and a high Youden index (0.58 vs. 0.37). The median time from CD34 +CD38cocktail + LSC positivity to relapse was longer than the median time from traditional MRD positivity to relapse by 141.5 days (range, 18-465 days) vs. 64.5 days (range, 13-144 days) (P=0.003). The median level between CD34 +CD38 -cocktail + LSCs and traditional MRD detected by MFC was 0.0072% (range, 0.0007%-3.742%) and 0.16% (range, 0.01%-3.75%) (P<0.001). Interpretation: Our data suggest the superiority of LSC-based MRD assays such as higher sensitivity, low false negativity, and longer time for MRD positivity to relapse to traditional MFC MRD methods for outcome prediction in AML patients received allograft. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2006 ◽  
Vol 107 (6) ◽  
pp. 2271-2278 ◽  
Author(s):  
Christiane Pott ◽  
Carsten Schrader ◽  
Stefan Gesk ◽  
Lana Harder ◽  
Markus Tiemann ◽  
...  

Abstract To evaluate the prognostic impact of minimal residual disease (MRD), quantitative real-time polymerase chain reaction (RQ-PCR) of clonal IGH rearrangements was performed in 29 patients with mantle cell lymphoma (MCL) treated with high-dose radiochemotherapy and autologous stem cell transplantation (ASCT). Fourteen of 27 patients evaluable for MRD after ASCT achieved complete clinical and molecular remission, whereas 13 patients had detectable MRD within the first year after ASCT. Molecular remission after ASCT was strongly predictive for improved outcome, with a median progression-free survival (PFS) of 92 months in the MRD-negative group compared with 21 months in the MRD-positive group (P < .001). Median overall survival (OS) was 44 months in the MRD-positive group and has not been reached in the MRD-negative group (P < .003). In multivariate analysis, molecular remission and bulky disease were independent prognostic factors for PFS (P = .001 and P = .021, respectively). While cyclophosphamide, doxorubicin, vincristine, prednisolone (CHOP)–like cytoreduction had only modest influence, ara-C–containing mobilization and myeloablative radiochemotherapy significantly reduced MRD. Quantitative MRD measured in the stem cell products of 27 patients was not predictive for molecular remission. We conclude that sequential quantitative monitoring of residual disease after ASCT is a powerful indicator for treatment outcome in MCL and defines subgroups of patients with a significantly different prognosis.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1879-1879
Author(s):  
Hiroyuki Takamatsu ◽  
Yoshiyasu Ogawa ◽  
Noriko Kobayashi ◽  
Kazue Obata ◽  
Tadashi Narisawa ◽  
...  

Abstract Abstract 1879 Background: Multiple myeloma (MM) can be cured by allogeneic stem cell transplantation (SCT), which induces PCR-negative molecular remission (MR) in patients with MM. Autologous peripheral blood stem cell transplantation (ASCT) followed by maintenance therapy can also induce PCR-negative MR in MM patients, with a progression-free survival (PFS) rate of 100% at 42 months (JCO 2010). Accordingly, it is essential to assess the depth of remission using clonotype-specific PCR primers in the treatment of MM. To prepare primers for amplification of rearranged regions of the IgH gene specific to individual MM patients, fresh or frozen bone marrow (BM) or peripheral blood cells containing MM cells are required. However, these materials are not always available, particularly when patients are diagnosed and treated at local hospitals. Archival BM slides that had been prepared for diagnosis may serve as a source of DNA for PCR-based minimal residual disease (MRD) detection. Therefore, we examined whether DNA extracted from archival BM slides was useful for designing clonotype-specific IgH PCR primers for MRD detection in autografts as well as in the BM in the post-ASCT setting. Patients and Methods: Twenty-two Japanese patients (9 men and 13 women; median age, 56 years; age range, 48–68) with newly diagnosed MM who received various induction regimens prior to ASCT were retrospectively analyzed. All patients had achieved a very good partial response (VGPR) or complete response (CR) after ASCT. BM slides from 19 MM patients, which had been stored in a steel box at room temperature for 19 to 90 months (median, 60 months), and fresh BM cells from 3 MM patients, obtained at first diagnosis, were subjected to DNA extraction using the QIAamp DNA Micro Kit (QIAGEN). The IGH Gene Clonality Assay Kit (InVivoScribe Technologies) was used to detect IgH gene rearrangements in the extracted DNA. The CDR III region of the IgH gene was sequenced to design clonotype-specific primers for amplification of the rearranged sequence in MM patients. BM slides prepared at 3 to 12 months post-ASCT were also subjected to DNA extraction, followed by MRD detection using clonotype-specific PCR primers. Results: IgH gene clonality was detected in 6 of 8 (75%) unstained slides and 4 of 5 (80%) Giemsa-stained slides that had been preserved for less than 5 years. For slides that had been preserved for more than 5 years, IgH gene clonality was detected in 5 of 11 (45%) unstained slides and 1 of 4 (25%) Giemsa-stained slides. Clonotype-specific IgH PCR primers were prepared in 9 of 19 cases (47%) using BM slides and in 3 of 3 cases (100%) using fresh BM cells. DNA in peripheral blood stem cell autografts of 12 patients who had achieved a VGPR or CR after ASCT was subjected to PCR to amplify clonotype-specific rearranged IgH gene sequences; rearranged sequences were amplified in four patients. Within 2 years post-ASCT, 3 of 4 (75%) patients with MRD-positive autografts (Group A) developed progressive disease, whereas all of the 8 patients with MRD-negative autografts (Group B) remained in remission. Group A showed a higher risk of progression than Group B (P = 0.023) (Figure 1A), although overall survival was comparable between the two groups (P =0.371). When BM slides (10 patients) and fresh BM cells (2 patients) obtained after ASCT were examined for the presence of MRD using clonotype-specific PCR primers, the rearranged IgH fragments were amplified in 5 of 12 patients. PFS of 5 MRD-positive patients tended to be shorter than that of 7 MRD-negative patients (P = 0.147) (Figure 1B). Conclusions: Archival BM slides can be used as a source of DNA to prepare clonotype-specific primers for MRD detection in MM patients whose cyropreserved myeloma cells are not available for DNA preparation. Furthermore, MRD status determined by PCR-based clonality analysis was useful in predicting prognosis of patients with MM. This approach enables us to select appropriate consolidation/maintenance therapy in MM patients based on their MRD status. A large multi-institutional study is currently underway to verify the clinical relevance of MRD detection using this technique. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2592-2592
Author(s):  
Beatriz Bellosillo ◽  
Carles Besses ◽  
Lourdes Florensa ◽  
Raquel Longaron ◽  
Gemma Navarro ◽  
...  

Abstract Introduction. Recently, the JAK2V617F mutation has been reported in the majority of PV patients as well as in a variable percentage of ET and IMF patients. Some authors have reported a high correlation of JAK2 mutation with PRV-1 overexpression and the formation of EECs. In the current study we have analyzed the pattern of positivity of these three biomarkers in a cohort of Ph-negative MPD patients. Patients and methods. 103 Ph-negative MPD patients (58 ET, 37 PV and 8 IMF) from a single institution were studied. Patients were diagnosed according to the PVSG and Barosi criteria. EEC formation was determined at diagnosis. At the time when PRV-1 and JAK2 mutation were analyzed 29/103 patients were receiving platelet-lowering therapy ± ASA, 26/103 patients only received ASA and 48/103 received no specific treatment. PRV-1 expression was quantified by real-time reverse transcriptase (RT)-PCR in RNA from granulocytes. Analysis of JAK2V617F was performed by direct sequencing using granulocyte RNA. Results. JAK2 mutation was observed in 21/58 ET (36.2%), 30/37 PV (81.1%) and 5/8 IMF (62.5%). PRV-1 was overexpressed in 25/58 ET (43.1%), 35/37 PV (94.6%) and 6/8 IMF (75%) and EECs formation was seen in 31/58 ET (53.4%), 34/37 PV (91.9%) and 6/8 IMF (75%). All markers were simultaneously positive (group A) in 43/103 patients (41.7%), concurrently negative (group B) in 25/103 patients (24.3%) and both positive and negative markers (group C) were observed in 35/103 patients (34%). In group A, 65% were PV patients, 26% were ET patients and 9 % were IMF patients. In group B, 8% were PV patients, 84% were ET patients and 8 % were IMF patients. In group C, 20% were PV patients, 74% were ET patients and 6 % were IMF patients. Regarding diagnosis, 76% of PV and 50% of IMF patients belonged to group A, whereas the majority of ET patients (45%) pertained to group C (table). When comparing ET and PV, a significant difference was observed (p&lt;0.001) concerning group distribution. Conclusion. These results show that, although a good correlation has been observed for the simultaneous expression of these three biomarkers, differences in the pattern of positivity, specially in ET, indicate that not all Ph-negative patients share the same pathogenetic mechanisms and point to other coexisting genetic abnormalities. Diagnosis ET PV IMF All markers positive (group A) 11 (19%) 28 (76%) 4 (50%) All markers negative (group B) 21 (36%) 2 (5%) 2 (25%) Positive and negative markers 26 (45%) 7 (19%) 2 (25%) Total 58 37 8


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3275-3275 ◽  
Author(s):  
Christiane Pott ◽  
Carsten Schrader ◽  
Stefan Gesk ◽  
Lana Harder ◽  
Markus Tiemann ◽  
...  

Abstract Mantle cell lymphoma (MCL) is characterized by a poor response to cytostatic therapy and a short survival with a median of less than 3 years. High-dose chemotherapy followed by autologous peripheral stem cell transplantation (ASCT) provides a potential curative option for the treatment of younger patients. The prognostic relevance of minimal residual disease (MRD) detection after intensive conventional treatment and high dose therapy has been proven for follicular lymphoma but is still under debate in MCL. Aim of this study was the quantitative evaluation of MRD and the evaluation of its prognostic impact on progression free survival and long-term remission in patients with MCL after ASCT. Quantitative Real-Time PCR for clonal IGH rearrangements (IGH-RQ-PCR) was performed in 29 patients with MCL treated with CHOP-like induction, stem-cell mobilisation with DexaBEAM. and subsequent ASCT. Quantitative MRD assessment was performed prior and during treatment as well as 3,6 and 12 months after PBSCT by IGH-RQ-PCR. 14/27 patients evaluable for MRD after ASCT achieved a complete clinical and molecular remission, whereas 13 patients had detectable MRD within the first year after ASCT. Molecular remission after ASCT was strongly predictive for improved outcome with a median PFS of 95 months in the MRD negative group compared to 21 months in the MRD positive group (p&lt;0.0001). Overall survival differed significantly with 55.8 months in the MRD positive group, whereas median OS of the MRD negative group has not been reached (p&lt;0.003). In multivariate analysis, molecular remission and bulky disease were independent prognostic factors for PFS (p=0.0007 and p=0.0210 respectively). Quantitative MRD measured in the stem cell products of 28 patients was not predictive for achieving molecular remission. We conclude that sequential quantitative monitoring of residual disease after ASCT is a powerful indicator for treatment outcome in MCL and defines subgroups of patients with a significantly different prognosis.


Author(s):  
U. Thiel ◽  
S. J. Schober ◽  
A. Ranft ◽  
H. Gassmann ◽  
S. Jabar ◽  
...  

AbstractPatients with advanced Ewing sarcoma (AES) carry a poor prognosis. Retrospectively, we analyzed 66 AES patients treated with allogeneic stem cell transplantation (allo-SCT) receiving HLA-mismatched (group A, n = 39) versus HLA-matched grafts (group B, n = 27). Median age at diagnosis was 13 years, and 15 years (range 3–49 years) at allo-SCT. The two groups did not differ statistically in distribution of gender, age, remission status/number of relapses at allo-SCT, or risk stratum. 9/39 (23%) group A versus 2/27 (7%) group B patients developed severe acute graft versus host disease (GvHD). Of patients alive at day 100, 7/34 (21%) group A versus 9/19 (47%) group B patients had developed chronic GvHD. In group A, 33/39 (85%) versus 20/27 (74%) group B patients died of disease and 1/39 (3%) versus 1/27 (4%) patients died of complications, respectively. Altogether 12/66 (18%) patients survived in CR. Median EFS 24 months after allo-SCT was 20% in both groups, median OS was 27% (group A) versus 17% (group B), respectively. There was no difference in EFS and OS in AES patients transplanted with HLA-mismatched versus HLA-matched graft in univariate and multivariate analyses. In this analysis, CR at allo-SCT is a condition for survival (p < 0.02).


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2034-2034
Author(s):  
Christoph M. Bucher ◽  
Dominik Heim ◽  
Andreas Buser ◽  
Jakob R. Passweg ◽  
Alois Gratwohl

Abstract Fetomaternal and maternofetal cell transfer have been described. Their clinical relevance is unknown. We hypothesized that firstborn siblings could tolerize their siblings born thereafter through sequential fetomaternal-maternofetal cell transfer. Hence, stem cell transplants within a family from a firstborn sibling (group A) should result more graft-versus-host disease (GvHD) and worse overall survival than transplants to a firstborn donor (group B). Results of a retrospective single center cohort analysis of 321 HLA-identical sibling donor hemopoietic stem cell transplants showed a survival of 48.3% (+/−10.9%) at 10 years in the group with firstborn donors (group A, 110 patients) as compared to 63.2 (+/−10.6%) in the group with firstborn recipients (group B, 105 patients; p<0.02) and a RR of death after adjustment for other risk factors of 2.6 (CI 1.45–4.66; p<0.001) for the group with firstborn donors. These results support the concept of a clinically relevant tolerizing effect of birth order, possibly mediated by fetomaternal cell transfer in man. Patients characteristics and outcomes Birth Order Donor First Sibling Recipient First Sibling n 110 105 Donor Age median (range) 30.1 (4.9–68) 25.4 (0.4–59.1) p=0.005 Recipient Age median (range) 25.4(3.2–62.1) 30.8 (2.2–63.0) Number of Siblings 2 67 69 n.s. 3 26 22 >3 17 14 Diagnosis n.s. Acute myeloid leukemia n 33 26 Acute lymphoblastic leukemia n 24 24 Chronic myeloid leukemia n 23 23 Lymphoproliferative disorders n 14 13 Severe aplastic anemia n 11 12 Myelodysplastic syndromes n 5 7 Stem Cell Source n.s. Bone Marrow n 71 73 PBSCT n 39 32 Conditioning n.s. With totoal body irradiation n 18 22 Without total body irradiation n 92 83 Outcomes Survival at 10 yrs % 48.3 63.2 p<0.02 Relapse at 3 yrs. % 26 20 n.s. Acute GvHD p=0.017 < Grade II n 46 61 >= Grade II n 64 44 Chronic GvHD n.s. none n 28 30 n.a. n 29 17 Limited n 32 30 Extensive n 21 23 Figure 1: Kaplan Meyer estimate of cumulative survival of groups A(firstborn donor: gray line) and B (firstborn recipient: black line). + indicates censored patient. Figure 1:. Kaplan Meyer estimate of cumulative survival of groups A(firstborn donor: gray line) and B (firstborn recipient: black line). + indicates censored patient.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2739-2739
Author(s):  
In Hae Park ◽  
June-Won Cheong ◽  
Jee Young Lee ◽  
Jin Seok Kim ◽  
Hyun Ju Chun ◽  
...  

Abstract Alemtuzumab is an anti-CD52 antibody, and has been used for lymphoid malignancies or as a member of non-myeloablative conditioning regimen in allogeneic transplantation. Especially, in non-myeloablative stem cell transplantation (NST), it has been reported that alemtuzumab is effective for graft-versus-host disease (GVHD), but the immune reconstruction after transplantation is delayed. In this study, we comparatively evaluated the efficacy of alemtuzumab for non-myeloablative conditioning, GVHD prophylaxis, and immune recovery in NST for hematologic diseases. We have compared the results in 28 recipients of a sibling or unrelated NST enrolled. The recipients were divided into 2 groups according to the use of alemtuzumab. In group A (n=21), the conditioning regimen was a combination of fludarabine, cyclophosphamide (or busulfan) and antithymocyte globulin (ATG), and group B (n=7) received fludarabine, cyclophosphamide (or busulfan), and alemtuzumab instead of ATG. GVHD prophylaxis was by cyclosporin A or FK506 plus methotrexate. There were no significant differences in the graft engraftment and period of granulocyte colony-stimulating factor infusion. Patients receiving alemtuzumab had a significantly lower incidence of acute GVHD (stage 2 or more) (14.3% versus 38.1%, P=0.03) and chronic GVHD (14.3% versus 52.4%, P=0.005). The relapse rate after transplantation was 28.6% (6 patients) in group A and 14.3 (1 patients) in group B (P=0.04). Flow cytometric analysis of peripheral mononuclear cells for evaluation of immune recovery showed that T-cell and NK-cell recovery were delayed in both groups. However, T-cell and NK-cell recovery after transplantation occurred earlier in patients received alemtuzumab. No significant differences were observed in disease-free or overall survival between two groups. In conclusion, alemtuzumab can be recommended for immune suppression in NST, with successful control over acute/chronic GVHD and inducing relatively earlier immune recovery after transplantation.


Sign in / Sign up

Export Citation Format

Share Document