Next Article in Journal
Retroperitoneal Lymph Node Dissection in Colorectal Cancer with Lymph Node Metastasis: A Systematic Review
Previous Article in Journal
Tissue-Specific microRNA Expression Profiling to Derive Novel Biomarkers for the Diagnosis and Subtyping of Small B-Cell Lymphomas
Previous Article in Special Issue
NEO212, a Perillyl Alcohol-Temozolomide Conjugate, Triggers Macrophage Differentiation of Acute Myeloid Leukemia Cells and Blocks Their Tumorigenicity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Haploidentical versus Double-Cord Blood Stem Cells as a Second Transplantation for Relapsed Acute Myeloid Leukemia

1
Department of Hematology, Catholic Hematology Hospital, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
2
Leukemia Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 24 November 2022 / Revised: 23 December 2022 / Accepted: 5 January 2023 / Published: 10 January 2023
(This article belongs to the Special Issue New Targets and Therapies of Acute Myeloid Leukemia)

Abstract

:

Simple Summary

Second stem cell transplantation (SCT2) may provide long-term remission for patients with relapsed acute myeloid leukemia (AML) after the first transplantation (SCT1). There are increasing demands for alternative donors, namely haploidential and umbilical cord blood, even in SCT2. In this single-center retrospective analysis for AML patients who relapsed after SCT1, we compared SCT2 outcomes with haploidentical donors (HIT) or double-cord blood (dCBT). In our study, HIT had superior transplant outcomes to dCBT as SCT2 in AML, due to the high non-relapse mortality in the dCBT group, which resulted in poorer survival. In a subgroup analysis, pre-transplant WT1-MRD positivity was associated with higher relapse rates and worse outcomes.

Abstract

There are limited data on second stem cell transplantation (SCT2) outcomes with alternative donors for relapsed AML after the first stem cell transplantation (SCT1). We analyzed the outcomes of 52 adult AML patients who received SCT2 from haploidentical donors (HIT, N = 32) and double-cord blood (dCBT, N = 20) between 2008 and 2021. The HIT group received T-cell-replete peripheral blood stem cells after reduced-toxicity conditioning with anti-thymocyte globulin (ATG), while the dCBT group received myeloablative conditioning. For a median follow-up of 64.9 months, the HIT group, compared to the dCBT group, had earlier engraftment, superior 2-year overall survival (OS), disease-free survival (DFS), and non-relapse mortality (NRM) with similar relapse. Multivariate analysis demonstrated that HIT was significantly associated with better OS, DFS, and lower NRM than dCBT. Both longer remission duration after SCT1 and complete remission at SCT2 were significantly associated with a lower relapse rate. In addition, bone marrow WT1 measurable residual disease (MRD) positivity was significantly associated with inferior OS and higher relapse. This study suggests that T-cell-replete HIT with ATG-based GVHD prophylaxis may be preferred over dCBT as SCT2 for relapsed AML and that WT1-MRD negativity may be warranted for better SCT2 outcomes.

1. Introduction

Relapse after stem cell transplantation (SCT) is a major cause of treatment failure in patients with acute myeloid leukemia (AML) and generally leads to poor survival outcomes [1,2]. Conventional chemotherapy and novel agents for post-SCT relapse in AML patients exhibit limited efficacy, especially in terms of achieving a durable response [1,3,4,5]. A recent observational study from the European Society for Blood and Marrow Transplantation (EBMT) compared a second allogeneic SCT with donor lymphocyte infusion (DLI) in relapsed AML after allograft. While both treatment modalities demonstrated similar overall survival (OS), DLI recipients had a significantly lower rate of remission compared to those who underwent a second SCT (SCT2) [3]. Most studies on SCT2 for hematologic malignancies repeatedly identified short remission duration after first SCT (SCT1) and active disease status before SCT2 as prognostic predictors of inferior outcomes, while there were contradictory results on the prognostic role of age, conditioning intensity at SCT1, prior graft-versus-host disease (GVHD) after SCT1, and transplant from a human leukocyte antigen (HLA)-mismatch donor [1,2,3,6,7,8,9,10,11,12,13,14,15,16,17,18].
Transplantation with HLA-mismatched alternative donors, such as haploidentical (HIT) and umbilical cord blood (CBT), has been increasingly used for patients without a suitable HLA-matched donor. Historically, HLA-mismatched transplantations were associated with higher incidences of GVHD, non-relapse mortality (NRM), and inferior survival. However, T-cell-replete strategies using anti-thymocyte globulin (ATG) and/or post-transplant cyclophosphamide (PTCy) as GVHD prophylaxis in HIT seem to overcome these disadvantages, leading to non-inferior outcomes as in the setting of SCT1 [19,20]. Umbilical cord blood is readily available and used across HLA barriers, with one of the main reasons being the limited number of T-cells. Double-cord blood unit infusions are frequently applied to overcome the small number of stem cells in a single unit [21,22,23,24,25]. However, there are limited data on the outcomes of SCT2 using these alternative donors in relapsed AML. Several retrospective studies for T-cell-replete HIT using various conditioning regimens with PTCy-based GVHD prophylaxis [14,16,26] and no data for CBT have been reported in this setting. There have yet to be any reports comparing HIT with CBT as SCT2. We developed T-cell-replete HIT using a unique conditioning regimen that is based on fludarabine, busulfan, and an intermediate dose of total body irradiation (TBI) [27,28]. The reduction in the TBI dose from a classical TBI-based myeloablative conditioning (MAC) can reduce nonhematologic toxicity; many authors frequently describe such conditioning intensity as reduced toxicity conditioning (RTC) [29,30,31,32,33,34,35]. Using the RTC regimen, along with ATG-based GVHD prophylaxis, we reported comparable transplant outcomes to matched unrelated donors in a prospective, biologically randomized study [19]. In this study, we compared the outcomes of T-cell-replete HIT using ATG-based GVHD prophylaxis with CBT using double units (dCBT) in relapsed AML patients after SCT1.

2. Results

2.1. Patient Characteristics

A total of 52 patients were included in this study. Patient-, disease-, and treatment-related characteristics at the time of diagnosis, SCT1, and SCT2 are described in Table 1. The median age at SCT2 was 47 years (range 23–67 years) and 53.8% of the patients were male (N = 28). The HIT (N = 32) and dCBT (N = 20) groups had no difference in the median age of patients (46.5 years vs. 49 years; p = 0.785). The patients in the HIT group were more likely to have received autologous transplant and myeloablative conditioning at SCT1 compared to patients in the dCBT group (41% vs. 5%, p < 0.001; 78% vs. 25%, p < 0.001, respectively). However, there was no difference between the groups with respect to cytogenetic risk, FLT3-ITD mutations, duration of remission after SCT1, intensity of salvage therapy, disease remission status before SCT2, and time from relapse to SCT2. The total infused nucleated cells and CD34+ cells were significantly greater in the HIT group compared to the dCBT group at SCT2 (p < 0.001 for both types).

2.2. Engraftment, GVHD, and Other Complications

The dCBT group exhibited a significantly delayed recovery of neutrophil and platelet counts compared to the HIT group (Figure 1A,B). The median (range) time to neutrophil engraftment were 12 days (6–23) for the HIT group and 28.5 days (16–75) for the dCBT group (p < 0.001). The median (range) days to platelet engraftment were 13 days (8–39 days) for the HIT group and 52 days (18–167 days) for the dCBT group (p < 0.001). The cumulative incidences of acute GVHD and grade 2–4 acute GVHD at 6 months were similar in each group (HIT vs. dCBT; acute GVHD, 41% vs. 45%, p =.866, Figure 1C; grade 2–4 acute GVHD, 38% vs. 33%, p = 0.426). The HIT group exhibited more frequent all grade chronic GVHD and moderate or severe chronic GVHD at 2 years compared to the dCBT group but without statistical significance (chronic GVHD, 46% vs. 15%, p = 0.057, Figure 1D; moderate or severe chronic GVHD, 29% vs. 6%, p = 0.123). There was no significant difference in CMV-related events (DNAemia, disease, or receiving anti-CMV treatment), thrombotic microangiopathy, sinusoidal obstruction syndrome, or herpes zoster infection between the two groups (Table S1). However, hemorrhagic cystitis occurred more frequently in the HIT group compared to the dCBT group (31% vs. 5%, p = 0.035).

2.3. Survival Outcomes between HIT and dCBT

The median (range) follow-up period from the time of SCT2 for all the patients was 64.9 months (12.6–151.6 months) for survivors. The 2-year cumulative incidence of NRM was lower in the HIT group compared to the dCBT group (22% vs. 50%, p = 0.014; Figure 1E), while there was no difference in the 2-year CIR (34% vs. 31%, p = 0.747, Figure 1F). The 2-year OS and 2-year DFS were significantly better for the HIT group than the dCBT group (43% vs. 30%, p = 0.018, Figure 1G; 44% vs. 19%, p = 0.023, Figure 1H, respectively). The early NRM (<100 days after transplant) was higher without statistical significance in dCBT than HIT (25% vs. 9%, p = 0.235). Infection was the most common cause of NRM in both the HIT and dCBT groups (16% vs. 35%, p = 0.175) with a higher tendency in dCBT, but no significant differences in NRM causes between the two groups were observed (Table S2). All the patients who relapsed died (N = 11, 34.3% of the HIT group; N = 6, 30.0% of the dCBT group), except for one patient in the HIT group, who is currently alive at more than 2 years after the third allogeneic transplant with double-cord blood.

2.4. Factors Affecting Survival Outcomes

Forest plots of the univariate analysis to identify risk factors associated with SCT2 outcomes (OS, DFS, NRM, and CIR) were presented in Figure S2. HIT, compared to dCBT, was associated with lower NRM (hazard ratio [HR] 0.34, 95% confidence interval [CI] 0.14–0.84, p = 0.019), resulting in superior OS (HR 0.46, 95% CI 0.23–0.89, p = 0.021) and DFS (HR 0.47, 95% CI 0.24–0.92, p = 0.027). Older age at SCT2 was associated with worse NRM (HR 1.04, 95% CI 1.01–1.07, p = 0.010) while disease remission at SCT2 was associated with less CIR (HR 0.25, 95% CI 0.10–0.63, p = 0.003). For patients in remission before SCT2 with available WT1-MRD data, WT1 greater than 250 copies/104 ABL at SCT2 resulted in worse OS, DFS, and CIR (HR 2.96, 95% CI 1.23–7.12, p = 0.016; HR 3.47, 95% CI 1.44–8.38, p = 0.006; HR 4.87, 95% CI 1.50–15.8, p = 0.008, respectively). Prior acute or chronic GVHD at SCT1 was not associated with NRM (acute GVHD grade ≥ 2: HR 0.76, 95% CI 0.17–3.48, p = 0.720; moderate or severe chronic GVHD: HR 0.91, 95% CI 0.29–2.88, p = 0.871).
On multivariate analysis (Figure 2), HIT was significantly associated with lower NRM (HR 0.29, 95% CI 0.12–0.74, p = 0.009) and superior OS (HR 0.43, 95% CI 0.22–0.84, p = 0.014) and DFS (HR 0.49, 95% CI 0.25–0.95, p = 0.035), compared to dCBT. Older age was associated with worse NRM and OS (in decades, NRM, HR 1.73, 95% CI 1.22–2.46, p = 0.002; OS, HR 1.01, 95% CI 1.01–1.80, p = 0.044). Disease in remission at SCT2 (HR 0.23, 95% CI 0.10–0.56, p = 0.001) as well as longer than 9 months of remission after SCT1 (HR 0.41, 95% CI 0.17–0.99, p = 0.047) resulted in less CIR. On a separate univariate (Figure S3) and multivariate analysis (Figure 3) with 38 patients in remission with available WT1-MRD status before SCT2, the WT1-MRD positivity (>250 copies/104 ABL) was significantly associated with worse OS (HR 4.56, 95% CI 1.78–11.7, p = 0.002), DFS (HR 6.92, 95% CI 2.48–19.3, p < 0.001), and CIR (HR 5.82, 95% CI 1.88–18.1, p = 0.002).

2.5. Subgroup Analysis with Patients Who Underwent Allogeneic SCT1

Given that the HIT group received more autologous transplantation for SCT1 compared to the dCBT group, we performed another subgroup analysis with patients who underwent allogeneic SCT1 (HIT group, N = 19; dCBT group, N = 19, Table S4). Among patients with allogeneic SCT1, no significant difference was observed in GVHD occurrence after SCT1 (HIT vs. dCBT; acute GVHD grade ≥ 2: 16% vs. 21%, p = 1.000; moderate or severe chronic GVHD: 16% vs. 32%, p = 0.447). One patient in HIT group experienced a recurrence of acute skin GVHD after salvage therapy and eventually died due to disease relapse after SCT2. The subgroup analysis (Figure S4) showed similar trends of superior 2-year OS (41% vs. 26%, p = 0.074) and DFS (42% vs. 14%, p = 0.111) in the HIT group compared to the dCBT group as in the whole group but without statistical significance. We observed lower rates of NRM (21% vs. 52%, p = 0.068) in the HIT group than in the dCBT group with similar CIR (37% vs. 33%, p = 0.745). There were no significant differences in acute GVHD and chronic GVHD in both groups. Prior GVHD did not affect the aforementioned survival outcomes (Figure S2).
In addition, a subgroup analysis (Figure S5) determining whether autologous or allogeneic SCT1 would have an effect on outcomes after HIT as SCT2 showed no significant difference in 2-year OS (auto vs. allo, 46% vs. 41%, p = 0.480), DFS (46% vs. 42%, p = 0.293), NRM (23% vs. 21%, p = 0.737), and CIR (31% vs. 37%, p = 0.577), or in acute and chronic GVHD.

3. Discussion

This retrospective single-center cohort study investigated the clinical outcomes of SCT2 with HIT and dCBT for AML patients who relapsed after SCT1. OS, the primary endpoint, exhibited better results in HIT compared to dCBT, primarily due to the higher NRM in dCBT. Neutrophil/platelet engraftment and NRM favored the HIT group, while CIR and acute/chronic GVHD did not differ between the two groups. Advanced disease status at SCT2 and a shorter time to relapse after SCT1 (less than 9 months) were significantly associated with a higher CIR. In addition, subgroup analysis revealed that a WT1-MRD status was predictive for post-transplant relapse and subsequent survival outcomes in this setting.
This is the first report, to the best of our knowledge, on T-cell-replete HIT as SCT2 using ATG-based GVHD prophylaxis in AML. Our HIT protocol was also characterized using a unique reduced-toxicity regimen including fractionated TBI 800 cGy with T-cell-replete PBSC and validated in a prospective study demonstrating the equivalence in survival outcomes with matched unrelated SCT for AML in the setting of SCT1 [19]. The advantages of our HIT protocol in SCT1, such as perfect engraftment with rapid neutrophil and platelet recovery, were again observed in this study of SCT2 for relapsed AML after SCT1. Several retrospective studies have described outcomes of HIT as SCT2 in relapsed hematologic malignancies (Table S3) [14,16,26]. They commonly used PTCy-based GVHD prophylaxis but with differences in conditioning intensity and stem cell sources, contrasting our study of relapsed AML using a uniform protocol consisting of ATG-based GVHD prophylaxis with T-cell-replete PBSC for HIT. A direct comparison may be limited due to the wide range of clinical factors, such as baseline patient and disease characteristics, the proportion of AML patients in the study population, time from the first transplant to relapse, salvage therapies before SCT2, remission status before SCT2, conditioning intensity, and proportion of PBSC as the stem cell source. Nonetheless, estimated OS after at least one year ranged from 29% to 45% in those studies, which is comparable to our 2 year-OS of 43%. CIR, NRM, and acute GVHD were similar as well, while the incidence of chronic GVHD in our cohort was higher than in previous studies. The current study first demonstrates that HIT using T-cell-replete PBSC and ATG-based GVHD prophylaxis has comparable outcomes for SCT2 in AML patients compared to those with HIT using PTCy-based GVHD prophylaxis. Given that the EBMT registry data reported better outcomes in leukemia-free survival, GVHD-free and relapse-free survival, GVHD, and NRM for PTCy-based regimens compared to ATG-based GVHD prophylaxis in AML for SCT1 [38], the superiority of PTCy over ATG is not yet conclusive. Prospective randomized trials comparing PTCy- and ATG-based GVHD prophylaxis for T-cell-replete HIT are warranted in the setting of SCT2 as well as SCT1.
This is also the first study to describe the outcomes of dCBT as SCT2 in AML, which was compared with HIT as SCT2. Our dCBT protocol has been utilized for acute lymphoblastic leukemia (ALL) in first CR with comparable survival outcomes with transplants from HLA-matched siblings or unrelated donors [25,39]. Lower incidences of chronic GVHD and CIR, but higher NRM, were characterized in dCBT as SCT1 for ALL in CR1 compared to other donor types. Older age over 40 was associated with higher early death (less than 100 days from transplant) and NRM. Many studies have been conducted to investigate the role of reduced-intensity conditioning (RIC) in cord-blood transplantation to reduce NRM and improve survival [40,41,42]. However, large registry data in AML showed that RIC did not significantly reduce NRM [37,38] but increased the risk of relapse [38] compared to MAC. Several comparative data sets for HIT and CBT in SCT1 for hematologic malignancies have been published [39,43,44,45,46,47,48,49]. Japanese [46] and European [49] registry data reported that CBT and HIT exhibited similar survival outcomes in adult AML patients. On the other hand, a recent BMT CTN 1101 trial, a randomized multicenter phase 3 trial of acute leukemia and lymphoma in remission comparing dCBT and HIT from BM using different RIC regimens in both donor types, showed that NRM and OS favored HIT over dCBT with statistical significance [44]. Indeed, CBT has concerns about a higher NRM compared to other donor types, primarily due to delayed immune reconstitution and increased risk for graft failure [43,44,45,47]. In line with the randomized phase 3 trial in SCT1 [44], our study demonstrates that delayed engraftment and higher NRM in dCBT lead to inferior OS compared to HIT in the setting of SCT2, even after age adjustment in multivariate analysis. The dCBT group had higher early NRM than the HIT group. However, previously observed stronger graft-versus-leukemia effects of dCBT versus other donor types in SCT1 for ALL [39] were not observed in AML when compared to HIT for SCT2. These findings suggest that T-cell-replete HIT with ATG-based GVHD prophylaxis would be the preferred approach in the setting of SCT2. Additional randomized controlled trials must be conducted to verify our results. Moreover, HIT has a benefit over dCBT in providing platforms for prophylactic or pre-emptive interventions using donor-originated cells, such as DLI [50], NK cells [51], or antigen-specific T cells [52]. On the other hand, given the higher CIR than NRM in HIT as SCT2 in this study, antibody or cell-based immunotherapy [50,51,52] and post-transplant maintenance therapy [53] under clinical trials must be considered to prevent relapse in SCT2.
Our data revealed that the disease status before SCT2 and time from SCT1 to relapse were significant prognostic factors in the setting of SCT2. Our group previously reported that later relapse (greater than 9 months) from the first transplant, younger age, and remission at SCT2 were associated with superior survival in AML patients who relapsed after SCT1 [11]. In line with these findings, the current study using alternative donors showed that longer times to relapse after SCT1 and disease in remission at SCT2 were significantly associated with lower CIR. Given the conflicting results about the impact of time from the first transplant to relapse on outcomes of HIT for SCT2 [14,16,17,26], our data support the impact of time from SCT1 to relapse. Notably, subgroup analysis with patients in remission at SCT2 revealed that pre-transplant WT1-MRD status was a significant factor for post-transplant relapse and poorer survival. Our group reported the prognostic role of WT1-MRD assessment in SCT1 for AML [19,54,55], which was consistent in this study with SCT2. These findings suggest the importance of quality in response to salvage treatment at relapse after SCT1 for the success of SCT2. Various emerging treatments, such as targeted therapy and immunotherapy [56], would be applicable to induce a deeper remission through various combinations with existing treatments. In addition, these emerging treatments would be applicable after SCT2 as a pre-emptive therapy based on MRD monitoring or prophylactic maintenance therapy. In this regard, relapsed AML patients after SCT1 should be enrolled in clinical trials with novel therapeutic strategies if possible.
This study has several limitations, including its retrospective nature, small sample size, imbalance in conditioning intensity, long treatment period, and limited genomic profiling. The donor selection process may have introduced bias into the results, as cord-blood transplants were provided when neither an HLA-matched donor nor a haploidentical donor was available. Given the limited reports of SCT2 using alternative donors and the lack of prospective data, our findings showed that T-cell-replete HIT with ATG-based GVHD prophylaxis had better outcomes than dCBT in the setting of SCT2. Both the HIT and dCBT groups were well balanced except for transplant type at SCT1 and conditioning intensity at SCT2. Autologous transplantation was more frequently performed for SCT1 in the HIT group compared to the dCBT group. However, there were no significant differences in the survival outcomes of SCT2 between patients with autologous and allogeneic SCT1 in the HIT group. Furthermore, a subgroup analysis including patients who underwent allogeneic SCT1 in both groups revealed a similar trend of superiority of the HIT group over the dCBT group in survival outcomes. Based on the differences in transplant protocols, including conditioning intensity, the outcomes between HIT and dCBT should be interpreted as results not from different donor types but from the transplant strategy to utilize stem cells from different donors. Given the aforementioned limitations, the findings in this study must be confirmed in a large prospective study.

4. Conclusions

Our findings show that T-cell-replete HIT with ATG-based GVHD prophylaxis would be the preferred approach in SCT2 compared to dCBT. Given the importance of a higher quality of response to salvage treatment at relapse for the success of SCT2 in this study, various emerging treatments, such as novel targeted therapy and immunotherapy, must be actively incorporated into the standard treatment for relapsed AML patients after SCT1. In addition, adjuvant treatment with prophylactic or pre-emptive strategies after transplant to prevent relapse may improve outcomes after SCT2.

5. Materials and Methods

5.1. Patients

We retrospectively analyzed data from consecutive patients with AML who underwent SCT2 from either haploidentical donors or double-cord bloods for relapse after SCT1 between 1 January 2008 and 31 December 2021. The patients were first assigned to HIT in the absence of HLA-matched sibling or unrelated donors and secondarily to dCBT in the absence of available haploidentical donors. Cord-blood units were selected as previously described [39]. The patients have received either autologous or allogeneic SCT1 and a different donor from SCT1 was selected for SCT2 according to a previously described strategy from our institution [11]. This study was approved by the Institutional Review Board of Seoul St. Mary’s Hospital in accordance with the principles of the Declaration of Helsinki. The survival data had been updated as of 30 June 2022.

5.2. Transplantation Procedures

The conditioning regimens and GVHD prophylaxis for HIT and dCBT were described in Figure S1. The HIT group received RTC with fractionated TBI (8.0 Gy), fludarabine (150 mg/m2), intravenous busulfan (6.4 mg/kg), and ATG (5.0 mg/kg; Sanofi-Genzyme, Lyon, France) for in vivo T-cell depletion and tacrolimus with methotrexate for GVHD prophylaxis. The dCBT group received myeloablative conditioning (MAC) with fractionated TBI (12.0 Gy), fludarabine (150 mg/m2), and cytarabine (9.0 g/m2) and tacrolimus and mycophenolate mofetil for GVHD prophylaxis. None of the patients received ex vivo T-cell depletion or PTCy. For the HIT group, we used peripheral blood as the stem cell source (PBSC). For all the patients undergoing dCBT, double-cord blood units were infused. The details for cord-blood selection, transplant procedures, and WT1 quantification in bone marrow (BM) for measurable residual disease (MRD) assessments were previously described [11,19,25,39,54].

5.3. Definitions

Remission, representing complete hematologic remission (CR), was defined by the International Working Group response criteria for AML [57]. Relapse was defined as the reappearance of leukemic blasts in the peripheral blood or ≥5% infiltration of a representative BM smear. Neutrophil engraftment was defined as an absolute neutrophil count >0.5 × 109/L in the first three consecutive days and platelet engraftment was defined as a platelet count >20 × 109/L in the first of five consecutive days without transfusions. OS was defined as the time from the second transplantation to death from any cause. Disease-free survival (DFS) was defined as survival without evidence of relapse or progression. Relapse was defined as the recurrence of leukemia, regardless of site. NRM was defined as death without evidence of relapse.

5.4. Statistical Analysis

Patient-, disease-, and treatment-related variables of the groups (HIT vs. dCBT) were compared using χ2 or Fisher exact test for categorical variables and the Mann–Whitney test for continuous variables. Baseline characteristics were summarized using the median and range for continuous variables and numbers and frequencies for categorical variables. The primary end point was OS. Secondary end points included DFS, cumulative incidence of relapse (CIR), NRM, acute GVHD, chronic GVHD, and neutrophil/platelet engraftment. Death for CIR, relapse for NRM, and both death and relapse for GVHD/engraftment were treated as competing events for cumulative incidence estimation. The survival outcomes were calculated using the Kaplan–Meier method, while the cumulative incidence function was used to estimate the CIR, NRM, acute GVHD, and chronic GVHD. Univariable analysis was performed using the logrank test for OS and DFS and the Gray test for CIR and NRM. The variables with a p-value less than 0.1 were included in multivariable analysis. The multivariable analyses were performed using the Cox proportional hazard regression model for OS and DFS, while the Fine-Gray method was used for relapse and NRM. All the statistical analyses were performed using R 4.1.2 (R Foundation for Statistical Computing, Vienna, Austria; http://www.r-project.org/, accessed on 30 July 2022) and EZR 1.55 [58]. The statistical significance was set at a p value less than 0.05.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/cancers15020454/s1, Figure S1: Conditioning scheme of haploidentical and cord blood transplantation; Figure S2: Univariate analysis for risk factors associated with SCT2 outcomes; Figure S3: Univariate analysis for risk factors associated with SCT2 outcomes for patients in complete remission before SCT2 with available WT1-PCR (N = 38); Figure S4: Transplant outcomes of subgroup of patients who received first transplant from allogeneic donor; Figure S5: Transplant outcomes of subgroup of patients who received second stem cell transplant from haploidentical donor; Table S1: Complications after second transplantation; Table S2: Causes of non-relapse mortality (NRM); Table S3: Comparison of outcomes after second haploidentical transplantation for relapsed AML; Table S4: Characteristics of patients who received first transplant from allogeneic donor.

Author Contributions

Conceptualization, J.-H.L., B.-S.C. and H.-J.K.; methodology, J.-H.L. and B.-S.C.; formal analysis, J.-H.L.; data curation, J.-H.L., B.-S.C., D.K., G.-J.M., S.-S.P., S.P., J.-H.Y., S.-E.L., K.-S.E., Y.-J.K., S.L., C.-K.M., S.-G.C., J.-W.L. and H.-J.K.; writing—original draft preparation, J.-H.L. and B.-S.C.; writing—review and editing, B.-S.C. and H.-J.K.; supervision, H.-J.K.; project administration, H.-J.K.; funding acquisition, B.-S.C. and H.-J.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (2022R1A2C2004517) and a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health and Welfare, Republic of Korea (HI22C1314). The content is solely the responsibility of the authors and does not reflect the views or the official policy or position of the Ministry of Health and Welfare or the Korean government.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Institutional Review Board of Seoul St. Mary’s Hospital (KC22RISI0836, 16 November 2022).

Informed Consent Statement

Patient consent was waived because formal consent is not required for this type of study.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request. The data are not publicly available due to ethical considerations.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sauer, T.; Silling, G.; Groth, C.; Rosenow, F.; Krug, U.; Gorlich, D.; Evers, G.; Albring, J.; Besoke, R.; Mesters, R.M.; et al. Treatment strategies in patients with AML or high-risk myelodysplastic syndrome relapsed after Allo-SCT. Bone Marrow Transplant. 2015, 50, 485–492. [Google Scholar] [CrossRef] [PubMed]
  2. Schmid, C.; Labopin, M.; Nagler, A.; Niederwieser, D.; Castagna, L.; Tabrizi, R.; Stadler, M.; Kuball, J.; Cornelissen, J.; Vorlicek, J.; et al. Treatment, risk factors, and outcome of adults with relapsed AML after reduced intensity conditioning for allogeneic stem cell transplantation. Blood 2012, 119, 1599–1606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Kharfan-Dabaja, M.A.; Labopin, M.; Polge, E.; Nishihori, T.; Bazarbachi, A.; Finke, J.; Stadler, M.; Ehninger, G.; Lioure, B.; Schaap, N.; et al. Association of Second Allogeneic Hematopoietic Cell Transplant vs Donor Lymphocyte Infusion with Overall Survival in Patients with Acute Myeloid Leukemia Relapse. JAMA Oncol. 2018, 4, 1245–1253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Federico, V.; Dargenio, M.; Matera, R.; Canaris, A.; Bozzoli, V.; Caretto, V.; Carlino, D.; De Paolis, M.R.; Fina, M.P.; Loglisci, G.; et al. Venetoclax Combined with Hypomethylating Agent (HMA) is Safe and Effective May Be a Good Bridge to Transplant in High-risk Acute Myeloid Leukemia. Bone Marrow Transplant. 2021, 56, 28–29. [Google Scholar]
  5. Perl, A.E.; Larson, R.A.; Podoltsev, N.A.; Strickland, S.; Wang, E.S.; Atallah, E.; Schiller, G.J.; Martinelli, G.; Neubauer, A.; Sierra, J.; et al. Follow-up of patients with R/R FLT3-mutation-positive AML treated with gilteritinib in the phase 3 ADMIRAL trial. Blood 2022, 139, 3366–3375. [Google Scholar] [CrossRef] [PubMed]
  6. Hosing, C.; Saliba, R.M.; Shahjahan, M.; Estey, E.H.; Couriel, D.; Giralt, S.; Andersson, B.; Champlin, R.E.; De Lima, M. Disease burden may identify patients more likely to benefit from second allogeneic hematopoietic stem cell transplantation to treat relapsed acute myelogenous leukemia. Bone Marrow Transplant. 2005, 36, 157–162. [Google Scholar] [CrossRef]
  7. Christopeit, M.; Kuss, O.; Finke, J.; Bacher, U.; Beelen, D.W.; Bornhäuser, M.; Schwerdtfeger, R.; Bethge, W.A.; Basara, N.; Gramatzki, M.; et al. Second Allograft for Hematologic Relapse of Acute Leukemia after First Allogeneic Stem-Cell Transplantation from Related and Unrelated Donors: The Role of Donor Change. J. Clin. Oncol. 2013, 31, 3259–3271. [Google Scholar] [CrossRef]
  8. Andreola, G.; Labopin, M.; Beelen, D.; Chevallier, P.; Tabrizi, R.; Bosi, A.; Michallet, M.; Santarone, S.; Ehninger, G.; Polge, E.; et al. Long-term outcome and prognostic factors of second allogeneic hematopoietic stem cell transplant for acute leukemia in patients with a median follow-up of 10 years. Bone Marrow Transplant. 2015, 50, 1508–1512. [Google Scholar] [CrossRef]
  9. Ruutu, T.; de Wreede, L.C.; van Biezen, A.; Brand, R.; Mohty, M.; Dreger, P.; Duarte, R.; Peters, C.; Garderet, L.; Schonland, S.; et al. Second allogeneic transplantation for relapse of malignant disease: Retrospective analysis of outcome and predictive factors by the EBMT. Bone Marrow Transplant. 2015, 50, 1542–1550. [Google Scholar] [CrossRef] [Green Version]
  10. Orti, G.; Sanz, J.; Bermudez, A.; Caballero, D.; Martinez, C.; Sierra, J.; Cabrera Marin, J.R.; Espigado, I.; Solano, C.; Ferra, C.; et al. Outcome of Second Allogeneic Hematopoietic Cell Transplantation after Relapse of Myeloid Malignancies following Allogeneic Hematopoietic Cell Transplantation: A Retrospective Cohort on Behalf of the Grupo Espanol de Trasplante Hematopoyetico. Biol. Blood Marrow Transplant. 2016, 22, 584–588. [Google Scholar] [CrossRef] [Green Version]
  11. Park, S.S.; Kim, H.J.; Min, K.I.; Min, G.J.; Jeon, Y.W.; Yoon, J.H.; Yahng, S.A.; Shin, S.H.; Lee, S.E.; Cho, B.S.; et al. Prognostic Prediction Model for Second Allogeneic Stem-Cell Transplantation in Patients with Relapsed Acute Myeloid Leukemia: Single-Center Report. Clin. Lymphoma Myeloma Leuk. 2018, 18, e167–e182. [Google Scholar] [CrossRef]
  12. Schneidawind, C.; Hagmaier, V.; Faul, C.; Kanz, L.; Bethge, W.; Schneidawind, D. Second allogeneic hematopoietic cell transplantation enables long-term disease-free survival in relapsed acute leukemia. Ann. Hematol. 2018, 97, 2491–2500. [Google Scholar] [CrossRef]
  13. Gyurkocza, B.; Storb, R.; Chauncey, T.R.; Maloney, D.G.; Storer, B.E.; Sandmaier, B.M. Second allogeneic hematopoietic cell transplantation for relapse after first allografts. Leuk. Lymphoma 2019, 60, 1758–1766. [Google Scholar] [CrossRef]
  14. Kharfan-Dabaja, M.A.; Labopin, M.; Brissot, E.; Kroger, N.; Finke, J.; Ciceri, F.; Deconinck, E.; Blaise, D.; Chevallier, P.; Gramatzki, M.; et al. Second allogeneic haematopoietic cell transplantation using HLA-matched unrelated versus T-cell-replete haploidentical donor and survival in relapsed acute myeloid leukaemia. Br. J. Haematol. 2021, 193, 592–601. [Google Scholar] [CrossRef]
  15. Yalniz, F.F.; Saliba, R.M.; Greenbaum, U.; Ramdial, J.; Popat, U.; Oran, B.; Alousi, A.; Olson, A.; Alatrash, G.; Marin, D.; et al. Outcomes of Second Allogeneic Hematopoietic Cell Transplantation for Patients with Acute Myeloid Leukemia. Transplant. Cell Ther. 2021, 27, 689–695. [Google Scholar] [CrossRef]
  16. Tischer, J.; Engel, N.; Fritsch, S.; Prevalsek, D.; Hubmann, M.; Schulz, C.; Zoellner, A.K.; Bucklein, V.; Lippl, S.; Reibke, R.; et al. Second haematopoietic SCT using HLA-haploidentical donors in patients with relapse of acute leukaemia after a first allogeneic transplantation. Bone Marrow Transplant. 2014, 49, 895–901. [Google Scholar] [CrossRef] [Green Version]
  17. Shimoni, A.; Labopin, M.; Finke, J.; Ciceri, F.; Deconinck, E.; Kroger, N.; Gramatzki, M.; Stelljes, M.; Blaise, D.; Stoelzel, F.; et al. Donor selection for a second allogeneic stem cell transplantation in AML patients relapsing after a first transplant: A study of the Acute Leukemia Working Party of EBMT. Blood Cancer J. 2019, 9, 88. [Google Scholar] [CrossRef] [Green Version]
  18. Vrhovac, R.; Labopin, M.; Ciceri, F.; Finke, J.; Holler, E.; Tischer, J.; Lioure, B.; Gribben, J.; Kanz, L.; Blaise, D.; et al. Second reduced intensity conditioning allogeneic transplant as a rescue strategy for acute leukaemia patients who relapse after an initial RIC allogeneic transplantation: Analysis of risk factors and treatment outcomes. Bone Marrow Transplant. 2016, 51, 186–193. [Google Scholar] [CrossRef] [Green Version]
  19. Cho, B.S.; Min, G.J.; Park, S.; Park, S.S.; Shin, S.H.; Yahng, S.A.; Jeon, Y.W.; Yoon, J.H.; Lee, S.E.; Eom, K.S.; et al. Haploidentical vs matched unrelated donor transplantation for acute myeloid leukemia in remission: A prospective comparative study. Am. J. Hematol. 2021, 96, 98–109. [Google Scholar] [CrossRef]
  20. Chang, Y.J.; Zhao, X.Y.; Huang, X.J. Haploidentical Stem Cell Transplantation for Acute Myeloid Leukemia: Current Therapies, Challenges and Future Prospective. Front. Oncol. 2021, 11, 758512. [Google Scholar] [CrossRef]
  21. Barker, J.N.; Weisdorf, D.J.; Wagner, J.E. Creation of a double chimera after the transplantation of umbilical-cord blood from two partially matched unrelated donors. N. Engl. J. Med. 2001, 344, 1870–1871. [Google Scholar] [CrossRef] [PubMed]
  22. De Lima, M.; St John, L.S.; Wieder, E.D.; Lee, M.S.; McMannis, J.; Karandish, S.; Giralt, S.; Beran, M.; Couriel, D.; Korbling, M.; et al. Double-chimaerism after transplantation of two human leucocyte antigen mismatched, unrelated cord blood units. Br. J. Haematol. 2002, 119, 773–776. [Google Scholar] [CrossRef] [PubMed]
  23. Lister, J.; Gryn, J.F.; McQueen, K.L.; Harris, D.T.; Rossetti, J.M.; Shadduck, R.K. Multiple unit HLA-unmatched sex-mismatched umbilical cord blood transplantation for advanced hematological malignancy. Stem Cells Dev. 2007, 16, 177–186. [Google Scholar] [CrossRef] [PubMed]
  24. Sideri, A.; Neokleous, N.; Brunet De La Grange, P.; Guerton, B.; Le Bousse Kerdilles, M.C.; Uzan, G.; Peste-Tsilimidos, C.; Gluckman, E. An overview of the progress on double umbilical cord blood transplantation. Haematologica 2011, 96, 1213–1220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Yoon, J.H.; Min, G.J.; Park, S.S.; Park, S.; Lee, S.E.; Cho, B.S.; Eom, K.S.; Kim, Y.J.; Kim, H.J.; Min, C.K.; et al. Durable outcomes of double-cord blood transplantation in adults with acute lymphoblastic leukemia: High-risk features for early and long-term mortality. Ther. Adv. Hematol. 2022, 13, 20406207221076762. [Google Scholar] [CrossRef]
  26. Srour, S.A.; Kongtim, P.; Rondon, G.; Chen, J.; Petropoulos, D.; Ramdial, J.; Popat, U.; Kebriaei, P.; Qazilbash, M.; Shpall, E.J.; et al. Haploidentical transplants for patients with relapse after the first allograft. Am. J. Hematol. 2020, 77, 45–49. [Google Scholar] [CrossRef]
  27. Cho, B.S.; Yoon, J.H.; Shin, S.H.; Yahng, S.A.; Lee, S.E.; Eom, K.S.; Kim, Y.J.; Lee, S.; Min, C.K.; Cho, S.G.; et al. Comparison of allogeneic stem cell transplantation from familial-mismatched/haploidentical donors and from unrelated donors in adults with high-risk acute myelogenous leukemia. Biol. Blood Marrow Transplant. 2012, 18, 1552–1563. [Google Scholar] [CrossRef] [Green Version]
  28. Yahng, S.A.; Kim, J.H.; Jeon, Y.W.; Yoon, J.H.; Shin, S.H.; Lee, S.E.; Cho, B.S.; Eom, K.S.; Kim, Y.J.; Lee, S.; et al. A well-tolerated regimen of 800 cGy TBI-fludarabine-busulfan-ATG for reliable engraftment after unmanipulated haploidentical peripheral blood stem cell transplantation in adult patients with acute myeloid leukemia. Biol. Blood Marrow Transplant. 2015, 21, 119–129. [Google Scholar] [CrossRef] [Green Version]
  29. Bacigalupo, A.; Ballen, K.; Rizzo, D.; Giralt, S.; Lazarus, H.; Ho, V.; Apperley, J.; Slavin, S.; Pasquini, M.; Sandmaier, B.M.; et al. Defining the intensity of conditioning regimens: Working definitions. Biol. Blood Marrow Transplant. 2009, 15, 1628–1633. [Google Scholar] [CrossRef] [Green Version]
  30. Giralt, S.; Ballen, K.; Rizzo, D.; Bacigalupo, A.; Horowitz, M.; Pasquini, M.; Sandmaier, B. Reduced-intensity conditioning regimen workshop: Defining the dose spectrum. Report of a workshop convened by the center for international blood and marrow transplant research. Biol. Blood Marrow Transplant. 2009, 15, 367–369. [Google Scholar] [CrossRef] [Green Version]
  31. Gyurkocza, B.; Sandmaier, B.M. Conditioning regimens for hematopoietic cell transplantation: One size does not fit all. Blood 2014, 124, 344–353. [Google Scholar] [CrossRef]
  32. Schmid, C.; Schleuning, M.; Ledderose, G.; Tischer, J.; Kolb, H.J. Sequential regimen of chemotherapy, reduced-intensity conditioning for allogeneic stem-cell transplantation, and prophylactic donor lymphocyte transfusion in high-risk acute myeloid leukemia and myelodysplastic syndrome. J. Clin. Oncol. 2005, 23, 5675–5687. [Google Scholar] [CrossRef]
  33. Marks, R.; Potthoff, K.; Hahn, J.; Ihorst, G.; Bertz, H.; Spyridonidis, A.; Holler, E.; Finke, J.M. Reduced-toxicity conditioning with fludarabine, BCNU, and melphalan in allogeneic hematopoietic cell transplantation: Particular activity against advanced hematologic malignancies. Blood 2008, 112, 415–425. [Google Scholar] [CrossRef]
  34. Wais, V.; Kundgen, L.; Bohl, S.R.; von Harsdorf, S.; Schlenk, R.F.; Dohner, K.; Teleanu, V.; Bullinger, L.; Nguyen, T.M.; Drognitz, K.; et al. Reduced-toxicity conditioning for allogeneic hematopoietic cell transplantation in elderly or comorbid patients with AML using fludarabine, BCNU and melphalan: Disease stage at transplant determines outcome. Bone Marrow Transplant. 2018, 53, 94–96. [Google Scholar] [CrossRef] [Green Version]
  35. Spyridonidis, A.; Labopin, M.; Savani, B.N.; Niittyvuopio, R.; Blaise, D.; Craddock, C.; Socie, G.; Platzbecker, U.; Beelen, D.; Milpied, N.; et al. Redefining and measuring transplant conditioning intensity in current era: A study in acute myeloid leukemia patients. Bone Marrow Transplant. 2020, 55, 1114–1125. [Google Scholar] [CrossRef]
  36. Grimwade, D.; Hills, R.K.; Moorman, A.V.; Walker, H.; Chatters, S.; Goldstone, A.H.; Wheatley, K.; Harrison, C.J.; Burnett, A.K.; National Cancer Research Institute Adult Leukaemia Working Group. Refinement of cytogenetic classification in acute myeloid leukemia: Determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. Blood 2010, 116, 354–365. [Google Scholar] [CrossRef] [Green Version]
  37. Eom, K.S.; Min, W.S.; Kim, H.J.; Cho, B.S.; Choi, S.M.; Lee, D.G.; Lee, S.; Min, C.K.; Kim, Y.J.; Cho, S.G.; et al. FLANG salvage chemotherapy is an effective regimen that offers a safe bridge to transplantation for patients with relapsed or refractory acute myeloid leukemia. Med. Oncol. 2011, 28 (Suppl. S1), S462–S470. [Google Scholar] [CrossRef]
  38. Ruggeri, A.; Sun, Y.; Labopin, M.; Bacigalupo, A.; Lorentino, F.; Arcese, W.; Santarone, S.; Gulbas, Z.; Blaise, D.; Messina, G.; et al. Post-transplant cyclophosphamide versus anti-thymocyte globulin as graft- versus-host disease prophylaxis in haploidentical transplant. Haematologica 2017, 102, 401–410. [Google Scholar] [CrossRef] [Green Version]
  39. Yoon, J.H.; Min, G.J.; Park, S.S.; Park, S.; Lee, S.E.; Cho, B.S.; Eom, K.S.; Kim, Y.J.; Kim, H.J.; Min, C.K.; et al. Impact of donor type on long-term graft-versus-host disease-free/relapse-free survival for adult acute lymphoblastic leukemia in first remission. Bone Marrow Transplant. 2021, 56, 828–840. [Google Scholar] [CrossRef]
  40. Rocha, V.; Mohty, M.; Gluckman, E.; Rio, B.; Eurocord; Reduced-Intensity Conditioning Subcommittee of the Acute Leukaemia Working Party; French Society of Bone Marrow Transplantation and Cellular Therapy. Reduced-intensity conditioning regimens before unrelated cord blood transplantation in adults with acute leukaemia and other haematological malignancies. Curr. Opin. Oncol. 2009, 21 (Suppl. S1), S31–S34. [Google Scholar] [CrossRef]
  41. Sheth, V.; Volt, F.; Sanz, J.; Clement, L.; Cornelissen, J.; Blaise, D.; Sierra, J.; Michallet, M.; Saccardi, R.; Rocha, V.; et al. Reduced-Intensity versus Myeloablative Conditioning in Cord Blood Transplantation for Acute Myeloid Leukemia (40–60 years) across Highly Mismatched HLA Barriers-On Behalf of Eurocord and the Cellular Therapy & Immunobiology Working Party (CTIWP) of EBMT. Biol. Blood Marrow Transplant. 2020, 26, 2098–2104. [Google Scholar] [CrossRef] [PubMed]
  42. Baron, F.; Ruggeri, A.; Beohou, E.; Labopin, M.; Sanz, G.; Milpied, N.; Michallet, M.; Bacigalupo, A.; Blaise, D.; Sierra, J.; et al. RIC versus MAC UCBT in adults with AML: A report from Eurocord, the ALWP and the CTIWP of the EBMT. Oncotarget 2016, 7, 43027–43038. [Google Scholar] [CrossRef] [Green Version]
  43. Brunstein, C.G.; Fuchs, E.J.; Carter, S.L.; Karanes, C.; Costa, L.J.; Wu, J.; Devine, S.M.; Wingard, J.R.; Aljitawi, O.S.; Cutler, C.S.; et al. Alternative donor transplantation after reduced intensity conditioning: Results of parallel phase 2 trials using partially HLA-mismatched related bone marrow or unrelated double umbilical cord blood grafts. Blood 2011, 118, 282–288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Fuchs, E.J.; O’Donnell, P.V.; Eapen, M.; Logan, B.; Antin, J.H.; Dawson, P.; Devine, S.; Horowitz, M.M.; Horwitz, M.E.; Karanes, C.; et al. Double unrelated umbilical cord blood vs HLA-haploidentical bone marrow transplantation: The BMT CTN 1101 trial. Blood 2021, 137, 420–428. [Google Scholar] [CrossRef] [PubMed]
  45. Kekre, N.; Antin, J.H. Cord blood versus haploidentical stem cell transplantation for hematological malignancies. Semin. Hematol. 2016, 53, 98–102. [Google Scholar] [CrossRef]
  46. Konuma, T.; Kanda, J.; Yamasaki, S.; Harada, K.; Shimomura, Y.; Terakura, S.; Mizuno, S.; Uchida, N.; Tanaka, M.; Doki, N.; et al. Single Cord Blood Transplantation Versus Unmanipulated Haploidentical Transplantation for Adults with Acute Myeloid Leukemia in Complete Remission. Transplant. Cell Ther. 2021, 27, 334.e1–334.e11. [Google Scholar] [CrossRef]
  47. Wu, R.; Ma, L. Haploidentical Hematopoietic Stem Cell Transplantation Versus Umbilical Cord Blood Transplantation in Hematologic Malignancies: A Systematic Review and Meta-Analysis. Cell Transplant. 2020, 29, 963689720964771. [Google Scholar] [CrossRef]
  48. Ruggeri, A.; Labopin, M.; Sanz, G.; Piemontese, S.; Arcese, W.; Bacigalupo, A.; Blaise, D.; Bosi, A.; Huang, H.; Karakasis, D.; et al. Comparison of outcomes after unrelated cord blood and unmanipulated haploidentical stem cell transplantation in adults with acute leukemia. Leukemia 2015, 29, 1891–1900. [Google Scholar] [CrossRef]
  49. Ruggeri, A.; Galimard, J.E.; Labopin, M.; Rafii, H.; Blaise, D.; Ciceri, F.; Diez-Martin, J.L.; Cornelissen, J.; Chevallier, P.; Sanchez-Guijo, F.; et al. Comparison of Outcomes after Unrelated Double-Unit Cord Blood and Haploidentical Peripheral Blood Stem Cell Transplantation in Adults with Acute Myelogenous Leukemia: A Study on Behalf of Eurocord and the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation. Transplant. Cell Ther. 2022, 28, 710.e1–710.e10. [Google Scholar] [CrossRef]
  50. Yan, C.H.; Liu, D.H.; Liu, K.Y.; Xu, L.P.; Liu, Y.R.; Chen, H.; Han, W.; Wang, Y.; Qin, Y.Z.; Huang, X.J. Risk stratification-directed donor lymphocyte infusion could reduce relapse of standard-risk acute leukemia patients after allogeneic hematopoietic stem cell transplantation. Blood 2012, 119, 3256–3262. [Google Scholar] [CrossRef]
  51. Choi, I.; Yoon, S.R.; Park, S.Y.; Kim, H.; Jung, S.J.; Jang, Y.J.; Kang, M.; Yeom, Y.I.; Lee, J.L.; Kim, D.Y.; et al. Donor-derived natural killer cells infused after human leukocyte antigen-haploidentical hematopoietic cell transplantation: A dose-escalation study. Biol. Blood Marrow Transplant. 2014, 20, 696–704. [Google Scholar] [CrossRef]
  52. Kinoshita, H.; Cooke, K.R.; Grant, M.; Stanojevic, M.; Cruz, C.R.; Keller, M.; Fortiz, M.F.; Hoq, F.; Lang, H.; Barrett, A.J.; et al. Outcome of donor-derived TAA-T cell therapy in patients with high-risk or relapsed acute leukemia post allogeneic BMT. Blood Adv. 2022, 6, 2520–2534. [Google Scholar] [CrossRef]
  53. Xuan, L.; Liu, Q. Maintenance therapy in acute myeloid leukemia after allogeneic hematopoietic stem cell transplantation. J. Hematol. Oncol. 2021, 14, 4. [Google Scholar] [CrossRef]
  54. Cho, B.S.; Min, G.J.; Park, S.S.; Shin, S.H.; Yahng, S.A.; Jeon, Y.W.; Yoon, J.H.; Lee, S.E.; Eom, K.S.; Kim, Y.J.; et al. WT1 Measurable Residual Disease Assay in Patients with Acute Myeloid Leukemia Who Underwent Allogeneic Hematopoietic Stem Cell Transplantation: Optimal Time Points, Thresholds, and Candidates. Biol. Blood Marrow Transplant. 2019, 25, 1925–1932. [Google Scholar] [CrossRef]
  55. Cho, B.S.; Yahng, S.A.; Min, G.J.; Park, S.; Park, S.S.; Shin, S.H.; Jeon, Y.W.; Yoon, J.H.; Lee, S.E.; Eom, K.S.; et al. Comparable Outcomes After Alternative and Matched Sibling Donor Hematopoietic Stem Cell Transplantation and the Role of Molecular Measurable Residual Disease for Acute Myeloid Leukemia in Elderly Patients. Transplant. Cell Ther. 2021, 27, 774.e–774.e12. [Google Scholar] [CrossRef]
  56. Kayser, S.; Levis, M.J. Updates on targeted therapies for acute myeloid leukaemia. Br. J. Haematol. 2022, 196, 316–328. [Google Scholar] [CrossRef]
  57. Cheson, B.D.; Bennett, J.M.; Kopecky, K.J.; Buchner, T.; Willman, C.L.; Estey, E.H.; Schiffer, C.A.; Doehner, H.; Tallman, M.S.; Lister, T.A.; et al. Revised recommendations of the International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia. J. Clin. Oncol. 2003, 21, 4642–4649. [Google Scholar] [CrossRef]
  58. Kanda, Y. Investigation of the freely available easy-to-use software ‘EZR’ for medical statistics. Bone Marrow Transplant. 2013, 48, 452–458. [Google Scholar] [CrossRef]
Figure 1. Outcomes after haploidentical and double-cord blood transplantations as the second transplantation. The cumulative incidences of (A) neutrophil recovery, (B) platelet recovery, (C) acute GVHD, (D) chronic GVHD, (E) non-relapse mortality, (F) relapse, and probabilities of (G) overall survival and (H) disease-free survival.
Figure 1. Outcomes after haploidentical and double-cord blood transplantations as the second transplantation. The cumulative incidences of (A) neutrophil recovery, (B) platelet recovery, (C) acute GVHD, (D) chronic GVHD, (E) non-relapse mortality, (F) relapse, and probabilities of (G) overall survival and (H) disease-free survival.
Cancers 15 00454 g001
Figure 2. Multivariate analysis of outcomes after second transplantation.
Figure 2. Multivariate analysis of outcomes after second transplantation.
Cancers 15 00454 g002
Figure 3. Multivariate analysis of outcomes after second transplantation for patients with available pre-transplant WT1-PCR (N = 38).
Figure 3. Multivariate analysis of outcomes after second transplantation for patients with available pre-transplant WT1-PCR (N = 38).
Cancers 15 00454 g003
Table 1. Patient characteristics.
Table 1. Patient characteristics.
Characteristics HIT (N = 32), N (%)dCBT (N = 20), N (%)p Value
AML MRC/treatment-related5 (16)/1 (3)5 (25)/0 (0)0.480
Age at SCT2, median (range)46.5 (23–67)49 (25–63)0.785
Male15 (47)13 (65)0.258
Hyperleukocytosis (≥50 × 109/L) at diagnosis
Unavailable CBC at diagnosis
4 (16) a
7
3 (17) a
5
1.000
Cytogenetics bFavorable
Intermediate
Adverse
4 (13)
25 (78)
3 (9)
3 (15)
10 (50)
7 (35)
0.053
FLT3-ITDPositive
Negative
Not available
4 (13)
20 (63)
8 (25)
3 (15)
14 (70)
3 (15)
0.697
Donor at SCT1Autologous
Matched-related
Matched-unrelated
Haploidentical
Cord blood
13 (41)
11 (34)
7 (22)
0 (0)
1 (3)
1 (5)
4 (20)
2 (10)
13 (65)
0 (0)
<0.001
SCT1 IntensityMAC
RTC c
RIC
25 (78)
3 (9)
4 (13)
5 (25)
12 (60)
3 (15)
<0.001
Remission duration after SCT1
≥9 months
24 (75)14 (70)0.754
Salvage therapyIntensive
FLANG d
FLAG-Ida e
MEC f
IA g
Non-intensive
Venetoclax-based
Sorafenib-based
With DLI
30 (94)
22 (69)
1 (3)
4 (13)
3 (9)
2 (6)
1 (3)
1 (3)
2 (6)
18 (90)
14 (70)
0 (0)
2 (10)
1 (5)
2 (10)
2 (10)
0 (0)
0 (0)
0.6340.517
Complete remission before SCT225 (78)17 (85)0.722
WT1 PCR (/104 ABL) before SCT2
CR, ≥250 (MRD-positive)
CR, <250 (MRD-negative)
CR, but data unavailable
Non-CR

5 (16)
18 (56)
2 (6)
7 (22)

3 (15)
13 (65)
1 (5)
3 (15)
0.960
HCT-CI0
1–2
≥3
5 (16)
12 (38)
15 (47)
5 (25)
3 (15)
12 (60)
0.207
Donor sexMatch
Partial mismatch
Full mismatch
16 (50)
-
16 (50)
3 (15)
12 (60)
5 (25)
Donor HLA match
(of eight loci, the lower value in dCBT)
4
5
6
≥7
11 (34)
16 (50)
5 (16)
0 (0)
3 (15)
7 (35)
6 (30)
4 (20)
<0.001
Total nucleated cells,
median (range), 108/kg
20.5 (8.7–40.3)0.39 (0.18–0.71)<0.001
CD34, median (range), 106/kg7.44 (1.77–28.96)0.07 (0.03–0.32)<0.001
Time between SCT1 and SCT2,
median months (range)
17.2 (6.6–142.0)15.7 (6.6–82.0)0.333
Time between relapse and SCT2,
median months (range)
3.7 (2.8–7.9)3.7 (2.8–7.1)0.631
Abbreviations: CBC: complete blood count; CR: complete remission; dCBT: double-cord blood transplantation; DLI: donor lymphocyte infusion; FLT3-ITD: fms-related tyrosine kinase 3-internal tandem duplication; GVHD: graft-versus-host disease; HCT-CI: hemotopoietic cell transplantation-specific comorbidity index; HIT: haploidentical donor transplant; HLA: human leukocyte antigen; MAC: myeloablative conditioning; MRC: myelodysplasia-related changes; PCR: polymerase chain reaction; RIC: reduced-intensity conditioning; RTC: reduced-toxicity conditioning; SCT1: first stem cell transplantation; SCT2: second stem cell transplantation; TNC: total nucleated cells. a Percentage was calculated as (number of patients with hyperleukocytosis at diagnosis)/(number of patients with available CBC at diagnosis) × 100 (%). b Cytogenetic classification was based on the International Working Group (IWG) response criteria for acute myeloid leukemia, published in 2003 [36]. c Reduced toxicity conditioning refers to fractionated TBI (8.0 Gy), fludarabine (150 mg/m2), and intravenous busulfan (6.4 mg/kg) [19]. d FLANG regimen consists of fludarabine 30 mg/m2/day, cytarabine 1 g/m2/day, mitoxantrone 10 mg/m2/day, and granulocyte-colony stimulating factor 300 μg/day for 5 days [37]. e FLAG-Ida regimen consists of fludarabine 30 mg/m2/day, cytarabine 1 g/m2/day, idarubicin 12 mg/m2/day and granulocyte-colony stimulating factor 300 μg/day for 5 days. f MEC regimen consists of cytarabine 2 g/m2/day, mitoxantrone 10 mg/m2/day for 4 days followed by etoposide 100 mg/m2/day for 3 days. g IA regimen refers to idarubicin 12 mg/m2/day for 3 days and cytarabine 100 mg/m2/day for 7 days.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lee, J.-H.; Cho, B.-S.; Kwag, D.; Min, G.-J.; Park, S.-S.; Park, S.; Yoon, J.-H.; Lee, S.-E.; Eom, K.-S.; Kim, Y.-J.; et al. Haploidentical versus Double-Cord Blood Stem Cells as a Second Transplantation for Relapsed Acute Myeloid Leukemia. Cancers 2023, 15, 454. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers15020454

AMA Style

Lee J-H, Cho B-S, Kwag D, Min G-J, Park S-S, Park S, Yoon J-H, Lee S-E, Eom K-S, Kim Y-J, et al. Haploidentical versus Double-Cord Blood Stem Cells as a Second Transplantation for Relapsed Acute Myeloid Leukemia. Cancers. 2023; 15(2):454. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers15020454

Chicago/Turabian Style

Lee, Jong-Hyuk, Byung-Sik Cho, Daehun Kwag, Gi-June Min, Sung-Soo Park, Silvia Park, Jae-Ho Yoon, Sung-Eun Lee, Ki-Seong Eom, Yoo-Jin Kim, and et al. 2023. "Haploidentical versus Double-Cord Blood Stem Cells as a Second Transplantation for Relapsed Acute Myeloid Leukemia" Cancers 15, no. 2: 454. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers15020454

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop