Skip to main content

Monitoring of post-transplant MLL-PTD as minimal residual disease can predict relapse after allogeneic HSCT in patients with acute myeloid leukemia and myelodysplastic syndrome

Abstract

Background

The mixed-lineage leukemia (MLL) gene is located on chromosome 11q23. The MLL gene can be rearranged to generate partial tandem duplications (MLL-PTD), which occurs in about 5-10% of acute myeloid leukemia (AML) with a normal karyotype and in 5-6% of myelodysplastic syndrome (MDS) patients. Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is currently one of the curative therapies available for AML and MDS with excess blasts (MDS-EB). However, how the prognosis of patients with high levels of MLL-PTD after allo-HSCT, and whether MLL-PTD could be used as a reliable indicator for minimal residual disease (MRD) monitoring in transplant patients remains unknown. Our study purposed to analyze the dynamic changes of MLL-PTD peri-transplantation and the best threshold for predicting relapse after transplantation.

Methods

We retrospectively collected the clinical data of 48 patients with MLL-PTD AML or MDS-EB who underwent allo-HSCT in Peking University People’s Hospital. The MLL-PTD was examined by real-time quantitative polymerase chain reaction (RQ-PCR) at the diagnosis, before transplantation and the fixed time points after transplantation. Detectable MLL-PTD/ABL > 0.08% was defined as MLL-PTD positive in this study.

Results

The 48 patients included 33 AML patients and 15 MDS-EB patients. The median follow-up time was 26(0.7-56) months after HSCT. In AML patients, 7 patients (21.2%) died of treatment-related mortality (TRM), 6 patients (18.2%) underwent hematological relapse and died ultimately. Of the 15 patients with MDS-EB, 2 patients (13.3%) died of infection. The 3-year cumulative incidence of relapse (CIR), overall survival (OS), disease-free survival (DFS) and TRM were 13.7 ± 5.2, 67.8 ± 6.9, 68.1 ± 6.8 and 20.3% ± 6.1%, respectively. ROC curve showed that post-transplant MLL-PTD ≥ 1.0% was the optimal cut-off value for predicting hematological relapse after allo-HSCT. There was statistical difference between post-transplant MLL-PTD ≥ 1.0% and MLL-PTD < 1.0% groups (3-year CIR: 75% ± 15.3% vs. 0%, P < 0.001; 3-year OS: 25.0 ± 15.3% vs. 80.7% ± 6.6%, P < 0.001; 3-year DFS: 25.0 ± 15.3% vs. 80.7 ± 6.6%, P < 0.001; 3-year TRM: 0 vs. 19.3 ± 6.6%, P = 0.277). However, whether MLL-PTD ≥ 1% or MLL-PTD < 1% before transplantation has no significant difference on the prognosis.

Conclusions

Our study indicated that MLL-PTD had a certain stability and could effectively reflect the change of tumor burden. The expression level of MLL-PTD after transplantation can serve as an effective indicator for predicting relapse.

Peer Review reports

Background

Acute myeloid leukemia (AML) is a highly malignant hematopoietic system disease and myelodysplastic syndrome (MDS) is a type of heterogeneous myeloid malignancies and frequently progress to AML [1,2,3,4]. In previous studies, molecular genetic aberrations have become important approaches for minimal residual disease (MRD) detection for AML and MDS. Especially, the polymerase chain reaction (PCR)-based gene detection has been proven to be an effective MRD monitoring method for AML patients [5,6,7]. However, more than half of AML cases still lack effective specific MRD molecular markers [5].

The mixed-lineage leukemia (MLL) gene, also named lysine (K)-specific methyltransferase 2A (KMT2A), is located on chromosome 11q23. Genetic alterations of the MLL gene are usually associated with the development of acute leukemia [8]. The MLL gene may be rearranged to generate partial tandem duplications (MLL-PTD), which usually spans exons 2 to 6, 2 to 7, and 2 to 8, or exons 3-9, exons 3-10, exons 3-11, or exons 3-10 and exons 3-11 at the molecular level [8,9,10,11]. MLL-PTD has been detected in approximately 5-10% of AML and 5-6% of MDS patients [12,13,14]. Low level of MLL-PTD (< 0.08%) may also be present in the blood and bone marrow of healthy individuals [5]. Previous reports support that polymerase chain reaction (PCR)-based MLL-PTD is a reliable MRD marker and is associated with poor prognosis [5, 12,13,14,15]. For chemotherapy patients, a higher MLL-PTD level at initial diagnosis predicts a lower incidence of chemotherapy complete remission (CR) and a lower survival rate [13]. The dynamic changes of chemotherapy patients also show that MLL-PTD levels within the first 6 months after the start of therapy are useful for early risk assessment of AML patients, and that a reduction of MLL-PTD level ≥ 2 log is a good prognostic factor for overall survival [5]. Furthermore, compared with healthy donors, MLL-PTD level have no difference from that of non-transplanted patients in continuous CR, while was significantly higher than that of transplanted patients in continuous CR [15]. Taken together, these findings support that MLL-PTD is a specific clinical prognostic marker in the initial diagnosis and chemotherapy for AML patients. However, there are few reports on the dynamics of MLL-PTD peri-transplantation, especially after transplantation. Thus, whether MLL-PTD could be used as a stable and reliable MRD marker in the process of transplantation and whether there is an optimal value of MLL-PTD to predict relapse after transplantation will be explored for the first time in our study.

In this study, we investigated a consecutive cohort of 33 AML and 15 MDS patients with MLL-PTD who received allo-HSCT at our institute. Most MLL-PTD MDS cases are classified as MDS with excess blasts (MDS-EB) [16]. Our study purposed to analyze the dynamic changes of MLL-PTD peri-transplantation and the best threshold for predicting relapse after transplantation.

Methods

Patients

The consecutive patients diagnosed with MLL-PTD expression> 0.08% AML or MDS undergoing allo-HSCT between January 2015 and March 2019 at the Peking University People’s Hospital, Institute of Hematology were enrolled in this study. The patients’ data were updated until September 30, 2020. The institutional review board at the hospital approved the protocol, and all patients or their guardians signed consent forms approved by the institutional review board.

Transplantation protocol

All the patients in this study received myeloablative conditioning regimens. Haploidentical HSCT (haplo-HSCT) and matched sibling donor transplantation (MSDT) were performed according to protocols reported previously by our institute [17, 18]. The conditioning regimen for MSDT patients is: Cytarabine (Ara-C) 2 g/m2/d i.v. for 1 day, cyclophosphamide (CTX) 1.8 g/m2/d for 2 days, busulfan (BU) 0.8 mg/kg i.v., q.i.d. for 3 days, and nitrosourea (Simustine, MeCCNU) 250 mg/kg for 1 day. The conditioning regimen for haplo-HSCT patients is: Ara-C 4 g/m2/d i.v. for 2 days, CTX 1.8 g/m2/d for 2 days, BU 0.8 mg/kg i.v., q.i.d. for 3 days, and MeCCNU 250 mg/kg for 1 day, and thymoglobulin (ATG, Sang Stat, Lyon, France) 2.5 mg/kg/d i.v. for 4 days.

Donor Lymphocyte Infusion (DLI)

Prophylactic DLI was administered for patients in relapse or no remission (NR) state before transplantation. The indications for DLI included hematological leukemia relapse, receiving chemotherapy followed by DLI, or positive MRD detection as previously described [19].

Detection of MRD

In this study, MRD was evaluated by Flow Cytometry (FCM) [20], the expression level of WT1 and MLL-PTD determined by RQ-PCR. The pre-transplant FCM, MLL-PTD and WT1 were performed using bone marrow (BM) samples within a month before the transplant as a routine. The post-transplant scheduled time points were + 1, + 2, + 3, + 4.5, + 6, + 9, and + 12 months post-HSCT and every 6 months thereafter.

The patients were analyzed for the presence of MLL-PTD at the MLL gene locus, as described previously [13, 15]. Briefly, MLL primers and hybridization probes were placed in exons 8-10 and 3 of the MLL gene, allowing for detection of MLL-PTD with exon 8/exon 3 fusion, exon 9/exon 3 fusion, or exon 10/exon 3 fusion. The transcript level was calculated as target transcript copies/ABL copies in percentages. Detectable MLL-PTD/ABL > 0.08% was defined as MLL-PTD positive [13]. The WT1 was detected as described previously and a WT1 transcript level less than 0.60% was defined as negative [21].

Definitions and assessments

The day of neutrophil engraftment was defined as the first day of 3 consecutive post-transplantation days on which the absolute neutrophil count (ANC) exceeded 500/μL. Patients who survived at least 28 days were considered to have had successful engraftment. The criteria for grading acute graft versus host disease (aGVHD) have been previously published [22, 23]. CR was defined as hematological CR that is, < 5% BM blasts, the absence of blasts in peripheral blood, the absence of extramedullary disease, an ANC > 1.0 × 109/L, and a platelet count > 100 × 109/L with no red cell transfusions. Hematological relapse was defined by morphologic evidence of disease in the peripheral blood, marrow, or extramedullary sites.

Statistical analysis

The primary study end point was the cumulative incidence of relapse (CIR). The secondary end points were the OS, disease-free survival (DFS) and treatment-related mortality (TRM). CIR, OS, DFS and TRM were defined as previously described [24]. Summary statistics, such as proportions, medians and ranges, were used to describe the patient characteristics and outcomes. The associations between MLL-PTD expression and post-transplantation outcomes were analyzed by the Kaplan-Meier method. Differences in CIR, DFS, OS and TRM between groups were calculated using the log-rank test. A two-sided P value of 0.05 was considered statistically significant. The independence of categorical parameters was calculated using the chi-square test or Fisher exact test, and the distribution of continuous variables was calculated using the Mann-Whitney U-test. All statistical analyses were performed using SPSS 23.0 (Chicago, IL, USA).

Results

Patients characteristics

A total of 33 AML patients included 13 males and 20 females, with a median age of 42 years (10-57 years) and 15 MDS-EB patients included 11 males and 4 females, with a median age of 51 years (4-60 years). The median follow-up time was 26 (0.7-56) months after HSCT. Patient characteristics are shown in Table 1. Of these 33 AML patients, 31 patients had gotten CR after chemotherapy, and 2 patients had gotten NR after 3 courses of chemotherapy. And 5 MDS-EB patients receiving chemotherapy including decitabine had gotten CR pre-transplantation. All patients had neutrophil engraftment, and 39 patients had platelet engraftment. Of the 33 patients with AML, 7 patients (21.2%) died of TRM and 6 patients (18.2%) underwent hematological relapse who died ultimately. The median hematological relapse time was 4.8 months (range 4-9 months) after HSCT in 6 relapsed patients. Of the 15 patients with MDS-EB, 2 patients (13.3%) died of infection. In addition, all enrolled patients had a 3-year CIR of 13.7% ± 5.2%, 3-year OS of 67.8% ± 6.9%, 3-year DFS of 68.1% ± 6.8% and 3-year TRM of 20.3% ± 6.1% (Fig. 1).

Table 1 Characteristics of acute myeloid leukemia and myelodysplastic syndrome patients
Fig. 1
figure 1

Cumulative incidence of relapse, overall survival, disease-free survival and treatment-related mortality of 48 MLL-PTD patients after allo-HSCT

Dynamic changes of MLL-PTD before and after transplantation

Observing the changes in the expression level of MLL-PTD at different time points peri-transplantation is helpful to analyze the stability of MLL-PTD as an MRD indicator in the transplantation system. Our results showed that the MLL-PTD level before transplantation was significantly lower than that at the initial diagnosis, but there were still 37 cases were MLL-PTD positive before transplantation, and 33 of 37 cases became negative within post-transplant 1 month. However, during our follow-up period, 25 cases eventually occurred post-transplant MLL-PTD positive. The median MLL-PTD level in all enrolled patients was decreased by around 35 folds after transplantation compared with that of pre-transplant CR status and was similar to the healthy controls (Table 2). Furthermore, among the 6 relapsed patients after transplantation, 3 of them maintained MLL-PTD at the healthy level (< 0.08%) within a month after transplantation. But before relapse, the MLL-PTD level of these 3 patients gradually increased (> 0.08%) and reached the highest level at the time of relapse. The MLL-PTD level of the other 3 relapsed patients continuously remained > 0.08% after transplantation, and the MLL-PTD levels of these 3 patients suddenly increased by hundreds of times before relapse.

Table 2 Comparison of MLL-PTD and WT1 at the initial diagnosed and peri-transplant patients

The effect of MLL-PTD level before and after transplantation on prognosis

Having analyzed the dynamic changes above which peri-transplant MLL-PTD can stably reflect the disease state we next studied the optimal threshold of post-transplant MLL-PTD for relapse. Our previous study shows that patients with MLL-PTD/ABL ≥ 1% based on initial diagnosis have a poor clinical prognosis [13]. In order to explore whether MLL-PTD could be used as a MRD marker after transplantation, we performed a receiver operating characteristic (ROC) with the highest expression level of post-transplant MLL-PTD before hematological relapse in all patients to determine the optimal cut-off value to predict relapse. The area under the ROC curve value was 0.977 (P < 0.001, Fig. 2A). The optimal cut-off value was MLL-PTD/ABL = 1.0%. And as shown in Fig. 2B, most post-transplant patients with MLL-PTD maintained a low level of expression, only 8 patients had MLL-PTD ≥ 1%, and 6 of the 8 patients eventually relapsed, which also implied the importance of MLL-PTD ≥ 1% in predicting relapse after transplantation. Based on the optimal cut-off value, we divided the post-transplant patients into two groups of MLL-PTD/ABL < 1% and MLL-PTD/ABL ≥ 1% to analyzed the prognostic difference. Our study found that the group of MLL-PTD/ABL ≥ 1.0% had higher 3-year CIR (75 ± 15.3% vs. 0%, P < 0.001, Fig. 3A), and lower 3-year OS (25.0 ± 15.3% vs. 80.7% ± 6.6%, P < 0.001, Fig. 3B) and 3-year DFS (25.0 ± 15.3% vs. 80.7 ± 6.6%, P < 0.001, Fig. 3C) compared with that of group of MLL-PTD/ABL < 1%. However, there was no statistical difference between the two groups in TRM (P > 0.05, Fig. 3D).

Fig. 2
figure 2

The curve of MLL-PTD expression levels post-transplantation. A Receiver operating characteristic (ROC) curve of MLL-PTD expression post-transplantation (AUC = 0.977, P < 0.001). B The level changes of post-transplant MLL-PTD

Fig. 3
figure 3

Kaplan-Meier survival curves analysis of patients between MLL-PTD < 1% and MLL-PTD ≥ 1% after transplantation

Both at the initial diagnosis and post-transplantation, it was analyzed that MLL-PTD = 1% was the optimal cut-off value, which implied that MLL-PTD/ABL = 1% was of important value in predicting prognosis. Therefore, we further analyzed whether MLL-PTD/ABL ≥ 1% before transplantation also indicated a poor prognosis after transplantation. However, our results showed that there was no statistical difference in prognosis between the MLL-PTD/ABL ≥ 1% and MLL-PTD/ABL < 1% group based on the level of MLL-PTD before transplantation (All P > 0.05, Fig. 4A-D), but the group of MLL-PTD/ABL ≥ 1% tended to have lower OS (P = 0.202, Fig. 4B), DFS (P = 0.202, Fig. 4C), and have a higher TRM(P = 0.105, Fig. 4D) compared with that of MLL-PTD/ABL < 1% group .

Fig. 4
figure 4

Kaplan-Meier survival curves analysis of patients between MLL-PTD < 1% and MLL-PTD ≥ 1% before transplantation

Factors affecting the prognosis of transplant patients with MLL-PTD

Factors affecting the prognosis were analyzed, including transplantation age, gender, disease type, donor type, blood type compatibility (Table 3). There was no statistical difference in TRM (P = 0.675), CIR (P = 0.115), DFS (P = 0.151) and OS (P = 0.157) between AML and MDS-EB. Among the 12 patients who received MSDT, 2 (16.7%) patients underwent hematological relapse both at 5 months after HSCT, and 1 patient died of pneumonia at 5.5 months. Among 36 patients who received haplo-HSCT, 4 patients (11.1%) underwent hematological relapse at a median of 4.5 months (range, 4-9 months) after HSCT, and 8 patients (22.2%) died due to TRM at a median of 5.3 months (range, 0.7-17.5 months). Based on the results of the analysis, it seemed that patients who received haplo-HSCT could achieve comparable outcomes compared to those who underwent MSDT (TRM: P = 0.271; CIR: P = 0.653; DFS: P = 0.544; OS: P = 0.560). The factor analysis of MLL-PTD level before and after transplantation showed that there was no statistical difference in pre-transplant MLL-PTD level. And post-transplant group of MLL-PTD/ABL ≥ 1% had a higher CIR, a lower OS and a lower DFS than that of group of MLL-PTD/ABL < 1% (all P < 0.001). In addition, other factors such as age, pre-transplant FCM, WT1 status and prophylactic DLI have no significant impact on prognosis. The ABO blood type and FLT3-ITD mutation at first diagnosis were important risk factors of CIR and OS after transplantation, respectively. Incompatible ABO blood type indicated a higher CIR than that of compatible ABO blood type, and patients with FLT3-ITD mutation had a low OS than that of without FLT3-ITD (Table 3).

Table 3 Univariate analysis of the variables affecting hematological TRM, CIR, DFS and OS in patients with MLL-PTD after allo-HSCT

Comparison of MLL-PTD and other MRD parameters

After transplantation, 8 patients were detected MLL-PTD/ABL ≥ 1.0% at a median of 3 months. Of the 8 patients, 7 patients were simultaneously (5 patients) or subsequently (2 patients) MRD positive detected by FCM at a median of 4.25 months (range,3-12 months), and 6 patients ultimately progressed to hematological relapse at a median of 2 months (range, 0.25–6 months) from the first time MLL-PTD/ABL ≥ 1.0%, half of whom receiving chemotherapy plus DLI. Finally, 2 patients receiving chemotherapy plus DLI became MRD negative gradually.

WT1 has been confirmed in previous studies to be an effective indicator of MRD monitoring and implementing interventions [21]. In order to analyze the specificity and sensitivity of MLL-PTD compared with WT1, we showed in Table 2 the dynamic changes of expression of MLL-PTD and WT1 at the initial diagnosis and different time points before and after transplantation. All 6 relapsed patients were detected MLL-PTD positive prior to relapse, while only 4 patients were detected positive for WT1. As shown in Table 2, the expression levels of MLL-PTD and WT1 both changed with the tumor burden. However, within post-transplant 3 months, MLL-PTD seemed be more sensitive than WT1 for MRD monitoring (P+ 1 month = 0.027; P+ 2 month = 0.009; P+ 3 month = 0.063).

Discussion

MLL-PTD is a special MLL rearrangement gene. No report had focused on the predictive significance of peri-transplant MLL-PTD expression on leukemia relapse after transplantation. In our retrospective study, results showed dynamic changes of MLL-PTD peri-transplantation, and the post-transplant MLL-PTD level is related to the prognosis of patients.

Previous reports have established the best threshold of MLL-PTD at the initial diagnosis for predicting the CR or relapse in AML patients [13, 15]. However, the AML patients with MLL-PTD analyzed in above reports included both non-transplanted patients and transplanted patients. Since different treatments (chemotherapy and transplantation) have a great impact on the prognosis of AML patients, they also have a certain impact on the accuracy of the MLL-PTD threshold for predicting relapse. Allo-HSCT is one of the curative therapies currently available for AML and MDS-EB, so it is very necessary to establish an optimal threshold of post-transplant MLL-PTD for relapse in transplanted AML patients. In the analysis of the post-transplant best cut-off value, we found that MLL-PTD/ABL = 1% can be used as the threshold for predicting relapse. Based on this result, physicians could need to pay more attention to the occurrence of relapse for post-transplant patients with MLL-PTD/ABL ≥ 1%. Under this condition, it is also necessary to shorten the MRD monitoring interval, or give appropriate relapse preventive interventions in combination with the clinical condition.

A stable and reliable MRD marker whose expression level needs to vary with the tumor burden. Our data showed that MLL-PTD levels in relapsed patients were significantly increased before relapse. Importantly, there was no occurrence of MLL-PTD turning negative or losing before relapse, which indicated that MLL-PTD had a certain stability and could effectively reflect the change of tumor burden. As expected, MLL-PTD was available prior to hematological relapse, but the relapse after MLL-PTD positive occurred at different rates. One of the explanations may be due to the patient’s combination of additional mutations such as FLT3-ITD. Previous report confirms that MLL-PTD positive relapses harboring an additional FLT3-ITD mutation to relapse faster than other patients with MLL-PTD alone [15]. In our study, the initial diagnosis of 2 relapsed patients was accompanied by FLT3-ITD mutation. They respectively relapsed at 12 days and 35 days after post-transplant MLL-PTD/ABL ≥ 1%, and the relapse was significantly faster than that of other relapsed patients. These data suggested MLL-PTD patients with other mutations such as FLT3-ITD may need to be shortened intervals of MRD monitoring after transplantation. Of course, a larger sample size or data is needed in the future to further support the above result.

The timely monitoring of MRD in the early stage after transplantation was beneficial to guide early clinical intervention to improve the prognosis of patients. Some studies have confirmed that the WT1 expression level is an independent prognostic indicator that can predict clinical outcome and combined use of WT1 and flow cytometry monitoring can promote sensitivity of predicting relapse after allo-HSCT [21, 25]. For AML and MDS lacking specific markers, we usually need to combine FCM and WT1 to evaluate MRD status. In the study, MLL-PTD became positive before relapse and prior to flow cytometry results. Thus, in contrast to FCM, PCR-based MLL-PTD detection have higher sensitivity. Our data showed that MLL-PTD seemed to be more sensitive than WT1 in early MRD monitoring after transplantation. Furthermore, in contrast to WT1, MLL-PTD is more specific for the type of MLL-PTD positive AML and MDS. However, for post-transplant patients with MLL-PTD, in order to monitor MRD more effectively and accurately, there may not be a better way than monitoring FCM, WT1 and MLL-PTD at the same time.

AML with MLL-PTD is a type of leukemia with a relatively poor prognosis compared with the standard-risk AML [13, 14]. In standard-risk AML, the post-transplant overall CIR and OS are around 15-20% and 60-70% at our institute, respectively [26, 27]. Our present results showed that the overall prognosis of post-transplant MLL-PTD patients (3-year OS: 67.8%; 3-year CIR: 13.7%) was similar to that of standard-risk patients. In addition, the other MLL rearrangement study about the transplant-related prognosis found that allo-HSCT would have a lower relapse risk and a higher survival probability compared to the results obtained from patients with chemotherapy alone [28]. The outcomes of patients with MLL-PTD are similar to the above results. The post-transplant OS in our study was significantly better than that of receiving chemotherapy alone (3-year OS< 40%) in previous study [5]. These data supported that allo-HSCT could achieve good therapeutic effect in patients with MLL-PTD at our institute. Furthermore, haplo-HSCT could achieve the similar therapeutic effect to the MSDT in patients with MLL-PTD. Therefore, our institution’s transplant and relapse prevention system may be effective for MLL-PTD patients.

Conclusions

In conclusion, MLL-PTD expression is a sensitive and specific MRD marker for the MLL-PTD patients received allo-HSCT. MLL-PTD expression level higher than 1.0% suggested a high risk of hematological relapse and tended to have a worse prognosis. Furthermore, allo-HSCT could achieve good therapeutic effect in patients with MLL-PTD AML and MDS-EB. Of course, due to the limited number of patients with MLL-PTD patients, we still need to continue research to accumulate more cases to further confirm the significance of MLL-PTD for MRD monitoring around transplantation.

Availability of data and materials

The datasets used and analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

AML:

Acute myeloid leukemia

MDS:

Myelodysplastic syndrome

Allo-HSCT:

Allogeneic hematopoietic stem cell transplantation

MRD:

Minimal residual disease

RQ-PCR:

Real-time quantitative polymerase chain reaction

TRM:

Treatment-related mortality

CIR:

Cumulative incidence of relapse

OS:

Overall survival

DFS:

Disease-free survival

MLL :

Mixed-lineage leukemia

PTD:

Partial tandem duplications

CR:

Complete remission

MSDT:

Matched sibling donor transplantation

NR:

No remission

DLI:

Donor lymphocyte infusion

BM:

Bone marrow

FCM:

Flow Cytometry

ANC:

Absolute neutrophil count

aGVHD:

Acute graft versus host disease

References

  1. Walter RB, Othus M, Borthakur G, Ravandi F, Cortes JE, Pierce SA, et al. Prediction of early death after induction therapy for newly diagnosed acute myeloid leukemia with pretreatment risk scores: a novel paradigm for treatment assignment. J Clin Oncol. 2011;29:4417–23.

    Article  Google Scholar 

  2. Pellagatti A, Boultwood J. The molecular pathogenesis of the myelodysplastic syndromes. Eur J Haematol. 2015;95:3–15.

    Article  CAS  Google Scholar 

  3. Hosono N. Genetic abnormalities and pathophysiology of MDS. Int J Clin Oncol. 2019;24:885–92.

    Article  CAS  Google Scholar 

  4. Gao MG, Ruan GR, Chang YJ, Liu YR, Qin YZ, Jiang Q, et al. The predictive value of minimal residual disease when facing the inconsistent results detected by real-time quantitative PCR and flow cytometry in NPM1-mutated acute myeloid leukemia. Ann Hematol. 2020;99:73–82.

    Article  CAS  Google Scholar 

  5. Weisser M, Kern W, Schoch C, Hiddemann W, Haferlach T, Schnittger S, et al. Risk assessment by monitoring expression levels of partial tandem duplications in the MLL gene in acute myeloid leukemia during therapy. Haematologica. 2005;90:881–9.

    CAS  PubMed  Google Scholar 

  6. Schnittger S, Weisser M, Schoch C, Hiddemann W, Haferlach T, Kern W. New score predicting for prognosis in PML-RARA+, AML1-ETO+, or CBFBMYH11+ acute myeloid leukemia based on quantification of fusion transcripts. Blood. 2003;102:2746–55.

    Article  CAS  Google Scholar 

  7. Guerrasio A, Pilatrino C, De Micheli D, Cilloni D, Serra A, Gottardi E, et al. Assessment of minimal residual disease (MRD) in CBFbeta/MYH11-positive acute myeloid leukemias by qualitative and quantitative RT-PCR amplification of fusion transcripts. Leukemia. 2002;16:1176–81.

    Article  CAS  Google Scholar 

  8. Poppe B, Vandesompele J, Schoch C, Lindvall C, Mrozek K, Bloomfield CD, et al. Expression analyses identify MLL as a prominent target of 11q23 amplification and support an etiologic role for MLL gain of function in myeloid malignancies. Blood. 2004;103:229–35.

    Article  CAS  Google Scholar 

  9. Schichman SA, Caligiuri MA, Gu Y, Strout MP, Canaani E, Bloomfield CD, et al. ALL-1 partial duplication in acute leukemia. Proc Natl Acad Sci U S A. 1994;91:6236–9.

    Article  CAS  Google Scholar 

  10. Caligiuri MA, Schichman SA, Strout MP, Mrózek K, Baer MR, Frankel SR, et al. Molecular rearrangement of the ALL-1 gene in acute myeloid leukemia without cytogenetic evidence of 11q23 chromosomal translocations. Cancer Res. 1994;54:370–3.

    CAS  PubMed  Google Scholar 

  11. Nilson I, Lochner K, Siegler G, Greil J, Beck JD, Fey GH, et al. Exon/intron structure of the human ALL-1 (MLL) gene involved in translocations to chromosomal region 11q23 and acute leukaemias. Br J Haematol. 1996;93:966–72.

    Article  CAS  Google Scholar 

  12. Meyer C, Hofmann J, Burmeister T, Gröger D, Park TS, Emerenciano M, et al. The MLL recombinome of acute leukemias in 2013. Leukemia. 2013;27:2165–76.

    Article  CAS  Google Scholar 

  13. Kong J, Zhao XS, Qin YZ, Zhu HH, Jia JS, Jiang Q, et al. The initial level of MLL-partial tandem duplication affects the clinical outcomes in patients with acute myeloid leukemia. Leuk Lymphoma. 2018;59:967–72.

    Article  CAS  Google Scholar 

  14. Choi SM, Dewar R, Burke PW, Shao L. Partial tandem duplication of KMT2A (MLL) may predict a subset of myelodysplastic syndrome with unique characteristics and poor outcome. Haematologica. 2018;103:e131–4.

    Article  CAS  Google Scholar 

  15. Ommen HB, Hokland P, Haferlach T, Abildgaard L, Alpermann T, Haferlach C, et al. Relapse kinetics in acute myeloid leukaemias with MLL translocations or partial tandem duplications within the MLL gene. Br J Haematol. 2014;165:618–28.

    Article  CAS  Google Scholar 

  16. Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127:2391–405.

    Article  CAS  Google Scholar 

  17. Huang XJ, Liu DH, Liu KY, Xu LP, Chen H, Han W, et al. Haploidentical hematopoietic stem cell transplantation without in vitro T-cell depletion for the treatment of hematological malignancies. Bone Marrow Transplant. 2006;38:291–7.

    Article  Google Scholar 

  18. Huang X, Liu D, Liu K, Xu L, Chen H, Han W, et al. Haploidentical hematopoietic stem cell transplantation without in vitro T cell depletion for treatment of hematologic malignancies in children. Biol Blood Marrow Transplant. 2009;15(1 Suppl):91–4.

    Article  Google Scholar 

  19. Chinese Society of Hematology, Chinese Medical Association. The consensus of allogeneic hematopoietic transplantation for hematological diseases in China(2016)-- post- transplant leukemia relapse. Zhonghua Xue Ye Xue Za Zhi. 2016;37:846–51.

    Google Scholar 

  20. Liu YR, Zhang LP, Chang Y, Cheng YF, Fu JY, Li LD, et al. Clinical significance for minimal residual disease detection by 4 color flow cytometry in adult and childhood B lineage acute lymphoblastic leukemia. Zhonghua Xue Ye Xue Za Zhi. 2006;27:302–5.

    CAS  PubMed  Google Scholar 

  21. Zhao XS, Jin S, Zhu HH, Xu LP, Liu DH, Chen H, et al. Wilms' tumor gene 1 expression: an independent acute leukemia prognostic indicator following allogeneic hematopoietic SCT. Bone Marrow Transplant. 2012;47:499–507.

    Article  CAS  Google Scholar 

  22. Martin P, Nash R, Sanders J, Leisenring W, Anasetti C, Deeg HJ, et al. Reproducibility in retrospective grading of acute graft-versus-host disease after allogeneic marrow transplantation. Bone Marrow Transplant. 1998;21:273–9.

    Article  CAS  Google Scholar 

  23. Shulman HM, Sullivan KM, Weiden PL, McDonald GB, Striker GE, Sale GE, et al. Chronic graft-versus-host syndrome in man. A long-term clinicopathologic study of 20 Seattle patients. Am J Med. 1980;69:204–17.

    Article  CAS  Google Scholar 

  24. Chang YJ, Xu LP, Wang Y, Zhang XH, Chen H, Chen YH, et al. Controlled, randomized, open-label trial of risk-stratified corticosteroid prevention of acute graft-versus-host disease after Haploidentical transplantation. J Clin Oncol. 2016;34:1855–63.

    Article  CAS  Google Scholar 

  25. Zhao XS, Yan CH, Liu DH, Xu LP, Liu YR, Liu KY, et al. Combined use of WT1 and flow cytometry monitoring can promote sensitivity of predicting relapse after allogeneic HSCT without affecting specificity. Ann Hematol. 2013;92:1111–9.

    Article  CAS  Google Scholar 

  26. Zhao XS, Cao LQ, Qin YZ, Yu W, Zhang XH, Xu LP, et al. Classifying AML patients with inv(16) into high-risk and low-risk relapsed patients based on peritransplantation minimal residual disease determined by CBFβ/MYH11 gene expression. Ann Hematol. 2019;98:73–81.

    Article  Google Scholar 

  27. Zhao X, Wang Z, Ruan G, Liu Y, Wang Y, Zhang X, et al. Impact of pre-transplantation minimal residual disease determined by multiparameter flow cytometry on the outcome of AML patients with FLT3-ITD after allogeneic stem cell transplantation. Ann Hematol. 2018;97:967–75.

    Article  Google Scholar 

  28. Wang Y, Liu QF, Qin YZ, Liu DH, Xu LP, Jiang B, et al. Improved outcome with hematopoietic stem cell transplantation in a poor prognostic subgroup of patients with mixed-lineage-leukemia-rearranged acute leukemia: results from a prospective, multi-center study. Am J Hematol. 2014;89:130–6.

    Article  Google Scholar 

Download references

Acknowledgments

Not applicable.

Funding

The work was supported by the National Key Research and Development Program of China (2017YFA0104500), the National Natural Science Foundation of China (grant no. 81870137) and Innovative Research Groups of the National Natural Science Foundation of China (grant no. 81621001), Beijing Municipal Science and Technology Commission (Z181100009618032).

Author information

Authors and Affiliations

Authors

Contributions

X.-S.Z. designed the study and was responsible for whole project administration. J.K., M.-G.G. and X.-S.Z. analyzed data and wrote the manuscript, J.K. and M.-G.G. contributed equally to this work.; Y.-Z.Q., Y.W., C.-H.Y., Y.-Q.S., Y.-J.C., L.-P.X., X.-H.Z., K.-Y.L. and X.-J.H. contributed to collect samples and validate results. All authors have read and approved the manuscript.

Corresponding author

Correspondence to Xiao-Su Zhao.

Ethics declarations

Ethics approval and consent to participate

The study followed the principles of the Helsinki Declaration and was approved by the Ethics Committee of Peking University People’s Hospital. All subjects obtained informed consent and all patients or their guardians signed consent forms approved by the institutional review board.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kong, J., Gao, MG., Qin, YZ. et al. Monitoring of post-transplant MLL-PTD as minimal residual disease can predict relapse after allogeneic HSCT in patients with acute myeloid leukemia and myelodysplastic syndrome. BMC Cancer 22, 11 (2022). https://doi.org/10.1186/s12885-021-09051-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-021-09051-5

Keywords