Next Article in Journal
Selective Targeting of α4β7/MAdCAM-1 Axis Suppresses Fibrosis Progression by Reducing Proinflammatory T Cell Recruitment to the Liver
Previous Article in Journal
Single-Cell RNA Sequencing Reveals Cardiac Fibroblast-Specific Transcriptomic Changes in Dilated Cardiomyopathy
Previous Article in Special Issue
Stem Cells Collection and Mobilization in Adult Autologous/Allogeneic Transplantation: Critical Points and Future Challenges
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Allogeneic Stem Cell Transplantation in Refractory Acute Myeloid Leukaemia

1
BMT Unit, AOR Villa Sofia-Vincenzo Cervello, 90146 Palermo, Italy
2
Onco-Hematology Unit, AOR Villa Sofia-Vincenzo Cervello, 90146 Palermo, Italy
3
Onco-Hematology and Cell Manipulation Laboratory Unit, AOR Villa Sofia-Vincenzo Cervello, 90146 Palermo, Italy
*
Author to whom correspondence should be addressed.
Cells 2024, 13(9), 755; https://doi.org/10.3390/cells13090755
Submission received: 19 February 2024 / Revised: 19 April 2024 / Accepted: 24 April 2024 / Published: 26 April 2024

Abstract

:
Refractory acute myeloid leukaemia is very difficult to treat and represents an unmet clinical need. In recent years, new drugs and combinations of drugs have been tested in this category, with encouraging results. However, all treated patients relapsed and died from the disease. The only curative option is allogeneic transplantation through a graft from a healthy donor immune system. Using myeloablative conditioning regimens, the median overall survival regimens is 19%. Several so-called sequential induction chemotherapies followed by allogeneic transplantation conditioned by reduced intensity regimens have been developed, improving the overall survival to 25–57%. In the allogeneic transplantation field, continuous improvements in practices, particularly regarding graft versus host disease prevention, infection prevention, and treatment, have allowed us to observe improvements in survival rates. This is true mainly for patients in complete remission before transplantation and less so for refractory patients. However, full myeloablative regimens are toxic and carry a high risk of treatment-related mortality. In this review, we describe the results obtained with the different modalities used in more recent retrospective and prospective studies. Based on these findings, we speculate how allogeneic stem cell transplantation could be modified to maximise its therapeutic effect on refractory acute myeloid leukaemia.

1. Introduction

In acute myeloid leukaemia (AML), induction chemotherapy (IC) based on daunorubicin plus cytarabine, with or without other drugs such as FLT3 inhibitors, conjugated anti-CD3 monoclonal antibodies, or purine analogues, is associated with a high overall response rate (ORR), defined as complete remission, complete remission with incomplete haematological recovery (CRi), or a morphological leukaemia-free state (MLFS). However, as reported in Figure 1, the CR rate was different based on the European leukaemia network (ELN) risk groups, and approximatively 10–40% of patients did not achieve a complete remission/complete remission with incomplete haematological recovery after IC. An important point is to appropriately define the type of unresponsiveness, because this can have consequences on the outcome after allogeneic stem cell transplantation (allo-SCT), which represents the only curative option. The Medical Research Council (MRC) proposed an operative definition of nonresponse based on the results obtained for 8907 AML patients included in several prospective studies [1]. They classified four unresponsive groups of patients as resistant (RES), with failure to achieve complete remission (CR) after C1, partial response (PR) after first IC with fewer than 15% blasts or a greater than 50% reduction in blast percentage; refractory to first IC (REF1) with more than 15% blasts and a less than 50% proportional reduction in blast percentage; and refractory to two courses of IC (REF2). The 5-year overall survival (OS) was greater for resistant and PR patients (17% and 21%, respectively) than for patients with REF1, REF2, or REF1 + 2 (9%, 8%, and 9%, respectively). As expected, the survival of the entire cohort of unresponsive patients was lower than that of patients who achieved a CR after one course of IC (40%). Allo-SCT improved outcomes in all these categories, except for in the PR group. More recently, the ELN defined primary refractory patients as those in which no CR, or CRi was documented after 2 courses of intensive IC [2]. Usually, if able, these patients receive reinduction chemotherapy to try to achieve a CR and to proceed to allo-SCT. The CR rate with several CT combinations is lower than that after IC and ranges from 10 to 50% [3]. Targeted therapies, such as FLT3 inhibitors, are more effective than conventional chemotherapy (CT) for inducing a response in patients with FLT3+ AML. Two randomised studies comparing FLT3 inhibitors to conventional CT in refractory AML patients showed that targeted therapy induced a greater response and improved survival [4,5]. More recently, the combination of venetoclax with other CT drugs has been used in this population of patients. In a small study, the overall response rate to venetoclax plus demethylating agents or low-dose cytarabine was 21% [6]. In an Italian study, venetoclax plus a demethylating agent was administered to 190 patients, and in the RR cohort, the CR rate was 44% [7]. Finally, venetoclax was associated with conventional CT (FLAGIda) in a prospective study, and the CR rate was 67% in the phase IIB arm [8]. Based on these unexhaustive data, most refractory AML patients do not achieve a response to reinduction therapies. These refractory patients are a true challenge for haematologists. In this paper, we summarise the data from retrospective and prospective studies, and we attempt to delineate how the outcome of refractory AML can be improved. Considering the large number of retrospective studies in the literature, we considered only studies including more than 100 patients published between 2010 and 2023. Studies were searched using the following items: refractory acute myeloid leukaemia, allogeneic stem cell transplantation, and a sequential therapy, myeloablative conditioning regimen. The search was performed by LC and RB, using PubMed. LC reviewed the papers selected, excluding those published in abstract form and reviews.

2. Results from Retrospective Studies

In the last few decades, several retrospective studies have reported results obtained in this population. The post-transplant course of patients not in CR at transplantation is worse than that of patients in CR [26,27]. Retrospective studies in which patients received a conventional transplant modality, myeloablative (MAC) or reduced-intensity conditioning (RIC) regimens, are reported in the Table 1. The overall survival obtained ranged from 20% to 40%, based on pre- and post-transplant characteristics. The leading cause of treatment failure is almost uniformly disease progression [28,29,30,31,32,33], and the cumulative incidence of relapse (CIR) is as high as 40–50%. Furthermore, in the setting of active disease, mainly after MAC, no-relapse mortality (NRM) is also of concern, reaching 40% of patients [34,35,36]. Studies including patients with conventional allo-SCT were reported in Table 1, while those treated with a sequential approach were reported in Table 2. In the first group (Table 1), the median OS, NRM, CIR, and leukaemia-free survival (LFS) were 27%, 24%, 49%, and 32%, respectively. Most of the included patients had primary induction failure (PIF) or relapsed (either untreated or secondary), with 20% blasts at the time of transplantation. In almost all the studies, the modality of immunosuppression tapering was not reported, nor was the use of prophylactic DLIs (pDLIs), except for two studies. Craddock et al. administered a pDLI to 28 patients and a therapeutic DLI to 15 patients to treat disease relapse [37]. In another paper, DLI was given to a total of 16 patients (12%) as early as day +120. The DLI treatment was deferred by the development of aGVHD in most patients [38]. Several predictive outcome factors were identified. In Table 3, we summarise the factors influencing OS, NRM, CIR, and LFS. Several factors, such as the AML stage, blast count, and the presence of adverse cytogenetic characteristics, were recurrent in several studies. Regarding the AML stage, patients treated with fewer than three cycles of CT had better OS, which was also reflected in better LFS. PIF was associated with a better LFS and reduced risk of relapse compared with relapsed disease [39]. The presence of leukaemic cells in the bone marrow or peripheral blood was predictive of poor prognosis, and a blast count of more than 20% was associated with shorter OS and LFS. Cytogenetics has also maintained its value in patients with more advanced disease and in predicting OS, CIR, and LFS [40]. A recipient age of more than 60 years was related to survival relapse and toxicity in three studies [33,39,40]. In two studies, the intensity of the conditioning regimens was found to be significant [39,40], and, as expected, intensive regimens were more effective against leukaemia, as they improved survival rates compared to less intensive regimens. While in early studies, transplantations were allocated from matched related or unrelated donors, and, more recently, allo-SCT from mismatched related and unrelated donors was frequently performed. This approach allows us to find a donor for virtually all patients, and, because of greater HLA disparities, a greater graft-versus-leukaemia effect can be assumed, also in the setting of RR AML. Baron et al. [41] reported that there was a trend towards better OS with mis-matched unrelated donors (mMUD) than with Haplo treatment. In this registry study, GVHD prophylaxis was administered uniformly via the PTCY platform. In several studies [33,37,42], prognostic scores (Table 4) were generated, confirming the heterogeneity of these refractory patients. Indeed, in the subgroup of patients without negative factors, the OS was relatively good (38–42%), and this data can aid in selecting candidates for transplantation.
Based on the dismal results with conventional allo-SCT, a different approach using sequential conditioning followed by RIC was introduced in clinical practice [52]. Overall, the OS and LFS seemed to be slightly greater than those obtained with the conventional approach. OS ranged from 24% to 53%, and LFS ranged from 22% to 70% (Table 2). However, comparisons between different studies must be considered with caution. Two retrospective studies comparing the FLAMSA-RIC to the MAC were published in the EBMT. In the first, FLAMSA-RIC was compared to fludarabine plus treosulfan (FT) or fludarabine plus thiotepa and busulfan (TBF). The NRM, CIR, OS, and LFS were all superimposable among the three treatment groups [53]. In the second, the FLAMSA-RIC score was compared to the MAC (both TBI and BU-based) in young AML patients. Patients treated with FLAMSA were regrouped into FLAMSA and RIC groups based on CT and total body irradiation (TBI) (FLAMSA-CT and FLAMSA-TBI, respectively). No patients were reported to be infused with pDLI, but a small fraction of them received pre-emptive DLI (8% in the FLAMSA-CT cohort, 16% in the FLAMSA-TBI cohort, and 7% in the MAC cohort). The 2-year NRM was lower in the FLAMSA-CT cohort than in the other two cohorts (7% vs. 18% in the FLAMSA-TBI cohort and 16–19% in the MAC cohort), and this was confirmed by multivariate analysis. Interestingly, compared with FLAMSA-TBI and MAC, FLAMSA-CT improved OS (50% vs. 36% vs. 34%) and LFS (40% vs. 27% vs. 28–30%), with similar relapse rates. Overall, these results, within the usual limits of registry studies, suggest better survival with FLAMSA-CT in RR AML patients [47]. Prophylactic DLI is considered an important part of the sequential approach. However, the feasibility of using a pDLI is questionable in retrospective studies. Indeed, as reported in Table 2, only three studies reported or planned administration of pDLIs [45,53], limiting the role of durable CRs, while the EBMT reported the efficacy of pDLIs in reducing relapse and improving survival in patients with acute leukaemia [54]. In the original sequential approach, pDLI treatment was planned to start on day +120 if patients were in CR, without GVHD or infection, or off of immunosuppression therapy (IS). This means that only a small, selected patient population can benefit from this therapeutic intervention, introducing the concept of immortal person-time bias. To address this, Weller et al. performed a retrospective analysis using an appropriate statistical method to control for this bias. The results from this analysis confirmed that, compared with pre-emptive DLI or no intervention, the FLAMSA-RIC protocol has a more positive impact on survival [51]. However, the place of pDLI in the sequential strategy was questioned. In a prospective study from England, patients with active AML and myelodysplasia, after debulking CT, were conditioned with fludarabine and cyclophosphamide without ATG, and without planned DLI. The 2 y OS was 39% and 1 y treatment related mortality (TRM) was 33%, and 2 y cumulative incidence of relapse was 30%. In this study, the median time to relapse was 95 days, and no patients relapsed after 2 years [55].
Several predictive factors for OS, NRM, CIR, and LFS were identified in studies using a sequential approach (Table 5). Overall, older patient age (with different cut-off values) frequently had a negative impact on OS, as did poor PS and recipient CMV-positivity. A higher recipient age was also significantly associated with NRM and, as expected, with the presence of comorbidities and previous infections. Interestingly, transplantation via the unrelated donor (UD) can protect against relapse but can also increase the risk of severe toxicity, with a negative impact on OS. However, these results could be due to the time of patient accrual. Indeed, in more recent years, the outcome after transplantation has significantly improved [56], and this could also be true in the setting of RR AML.

2.1. Results from Prospective Studies

Few prospective studies have been conducted on RR AML patients. The main results from these experiments are reported in Table 6.
In these studies, the OS ranged from 19% to 70%, the LFS from 19% to 62%, the CIR from 20% to 54%, and the NRM from 20% to 54%. This wide range of outcomes stems from large differences in terms of inclusion criteria (age, patients, and disease characteristics), transplant technique, GVHD prophylaxis, and use of pDLI. Indeed, three studies applied a sequential approach [52,57,60], one performed allo-SCT during the aplasia period after IC [58], and two used a more conventional approach with an MAC regimen [59,61]. The pDLI treatment was planned for four studies [52,57,59,60] after treatment with different modalities to reduce the amounts of immunosuppressive drugs needed. Finally, GVHD prophylaxis was variable, even if three studies were based on cyclosporin A (CyA) plus MMF [52,57,60], and in vivo T-cell depletion was used in three trials [52,57,60], independent of the donor type. In two trials [59,61], haploidentical donors were included; in all other trials, matched donors were related or unrelated, and cord blood was matched in one [61]. Nonetheless, these differences deserve some considerations. First, allo-SCT is an effective treatment for leukemic blasts that are not sensitive to conventional chemotherapy. The sequential approach, based on debulking CT, RIC regimen, and pDLI, seems to be feasible and more effective in this situation. It is difficult to determine which part of this strategy is essential. Considering that RIC regimens exhibit low activity in refractory patients and that durable CR is less than 10% after debulking CT alone, it could be speculated that the immunological reaction exercised by pDLI is the main factor involved. However, in the three trials containing pDLI in the protocol, only a small fraction of patients received it, minimizing the weight of the beneficial results reported. On the other hand, OS was exceptionally high in patients receiving at least one pDLI. In an Indian study [59], the role of pDLI was more clear and well supported. Indeed, in one cohort of patients, the MACs and 2 y OS and LFS were 35% and 25%, respectively, but when they used MACs plus very early pDLIs (from day +21), the 2 y OS and LFS increased to 70% and 62%, respectively. However, in this study, only haploidentical donors were included, and post-transplant immunosuppressive drugs were tapered on day +14 for MMF and on day +60 for CyA. These extraordinary results need to be confirmed in more patients and in multicentre settings. Several factors predictive of outcome were identified in these studies (Table 7).
Predictive factors were different due to the heterogeneous protocols used. In the German studies [52,57], high number CD34 positive (≥9.6 × 106/kg) was associated with better OS, LFS, and NRM. Furthermore, NRM was lower when the donor was matched related. Finally, less pre-treated patients (<2 induction chemotherapy) showed a better survival. The high intensity of the conditioning regimen plus the systematic infusion of donor lymphocytes can improve the survival, as well as the presence of chronic GVHD [55,58,59]. In the Italian study [61], a high comorbidities index (hematopoietic cell transplantation-comorbidity index) was associated with high NRM and low OS, while low-risk cytogenetic was associated with better OS.

2.2. How Can Allo-SCT Be Modified to Improve Outcomes?

Based on previous retrospective and prospective studies, it is obvious that there is room for improving the outcome of refractory AML patients treated with allo-SCT. It is clear that conventional allo-SCT performed in this setting has a small chance of being successful. To move forward from this, each segment of allo-SCT (conditioning regimen, GVHD prophylaxis, use of pDLI or pre-emptive DLI) should be re-evaluated and modified. Furthermore, the strategy of transplantation should be better scheduled (MAC vs. early sequential RIC vs. late sequential). The challenge, of course, is that every part modified in the process can have consequences for the others.
Starting from which strategy to use in a refractory AML setting, several retrospective studies and two prospective studies have shown that the relapse incidence and transplant-related mortality are high and that the OS is approximately 20% when MAC conditioning regimens are used (Table 1 and Table 6). In retrospective studies, conventional allo-SCT was basically structured as it was in patients with well-controlled disease and consisted of myeloablative conditioning regimens, regular GVHD prophylaxis, and immunosuppressive drug reduction. In one of the largest registry-based studies [42] reporting on patients transplanted from 1995 to 2004, the 3-year OS for AML patients was 19%. Several predictive factors (first CR duration ≤ 6 months, circulating blasts, donor other than HLA-identical sibling, Karnofsky or Lansky score ≤ 90, and poor-risk cytogenetics) were retrieved, identifying a subgroup of patients with a remarkable OS of 40% (Table 1). In this study, the conditioning regimens were myeloablative busulfan or TBI-based; 65% of the patients received bone marrow as a stem cell source, and GVHD prophylaxis consisted of cyclosporine A or tacrolimus plus methotrexate. In another registry-based study from Japan [38], OS was as low as that in other similar studies, with several predictive factors that can affect the outcome (Table 1). Similar results were reported by an Italian group (GITMO) in a more recent prospective study including only alternative donors conditioned with MAC regimens and with regular GVHD prophylaxis [61]. On the other hand, sequential strategies seem to improve the outcome in some but not in all studies (Table 2 and Table 6). The original protocol published by Schmid et al. has been modified in several ways, but the results were not significantly improved. Furthermore, the two approaches were compared in retrospective studies, and the clinical outcomes were not significantly different, only in one study [47] but not in the others [53,62].
The intensity of the conditioning regimen plays a role in the outcomes of AML patients [63], and, as a consequence, any improvement in terms of activity against leukaemic cells or tolerability could have a positive impact on the outcome. Thus, improving the treatment efficacy through the addition of synergistic agents may be more appealing. An alternative to this approach could be appropriate for optimising the use of regular drugs (i.e., busulfan) or other drugs. Based on in vitro studies at MD Andersson, several drugs were added to the BUFLU backbone. In a randomised trial, Alatrash et al. included 70 patients treated with different combinations of BUFLU + clofarabine. Eighty-one percent of the patients had active disease at transplantation. The OS and PFS improved in patients treated with the highest dose of clofarabine (30 mg/m2/day), and these improvements were not influenced by disease status. The tolerability was good, the NRM was 13%, and the CIR was 41% [64]. In another trial, the BUCY protocol was strengthened by the addition of a hypomethylating agent (decitabine 5 days before BUCY). In this study, the results obtained with decitabine added to BUCY were encouraging and better than in a historical cohort treated without decitabine [65]. In a retrospective study of the EBMT cohort, two of the most common MAC regimens were compared in an AML setting. The regimens were TBI or IV busulfan-based. Nevertheless, owing to the well-known limits of registry studies in terms of selection bias, the outcomes were similar, and the CT MAC was considered a valid alternative to the TBI MAC. Of course, the major message of this paper was addressed to those centres that did not have easy access to TBI [43]. Finally, the activity of a newer alkylating agent, such as treosulfan, was analysed and compared to the classical busulfan plus fludarabine association. Treosulfan and busulfan were administered at a myeloablative dose (42 g/m2 and 12.8 mg/kg, respectively). Overall, the safety profile of a treosulfan-based conditioning regimen was better, leading to an improved leukaemia-free survival and overall survival [66].
In addition to the intensity of the conditioning regimen, the proper exposure to drugs can play a central role in optimising antileukaemic activities. MD Andersson researchers were pioneers in this field. Several studies suggested that pharmacokinetic-based busulfan (BUPK) administration is more effective than the fixed-dose approach [67]. However, few centres, at least in Europe, were able to perform daily BUPK, even if the data from Houston were confirmed [68]. Another way to improve conditioning regimens without increasing toxicity is to introduce newer drugs, such as treosulfan. According to a randomised study, a reduced dose of treosulfan was more efficacious than a reduced dose of busulfan in aged patients with early-stage AML [69]. Treosulfan was also compared to TBF and FLAMSA-based in the setting of RR AML in a retrospective EBMT study [53]. In that study, treosulfan plus fludarabine (FT) had activity similar to that of TBF alone or in combination with the other sequential strategies (FLAMSA-based). However, there was a trend towards decreased early NRM and better survival in patients with FT and the sequential approach than in patients with TBF. Of course, it could be speculated that FT in a sequential scheme should improve the tolerability and activity in RR AML patients.
The use of pDLI seems to be pivotal for enhancing disease control [51,70], and it is an integral step of a sequential approach. However, in the studies where pDLI was planned, the feasibility of pDLI was low, because only one-quarter of the eligible patients received DLI [52,57,60]. There were several reasons, but these were mainly linked to the inclusion criteria, such as persistent CR, no GVHD, no IS drugs, and no infections or other complications. All these factors reduce the eligibility of patients for pDLIs and delay their infusions, exposing patients to leukaemia relapse. One way to improve the application of pDLI could be to start earlier after transplantation in parallel with IS reduction. As reported by Jaiswal, this schedule was also feasible in the context of transplantation via a haplo donor [59]. For example, cyclosporine A (CyA) could be tapered from day +60 to plan for the first pDLI at the same time to continue with CyA withdrawn on day +90, coupled with the second pDLI and third pDLI after 4–6 weeks. This schedule of pDLI for immunosuppression could improve the complexity of the relationship between time, incidence of GVHD post-DLI, timing of relapse, and incidence of GVHD after infusion [71]. Another means could be to use a modified DLI. Several Chinese groups used G-CSF-mobilised DLIs for immunosuppression, and, in a prospective trial, they showed that G-mobilised pDLIs were effective and well tolerated [72]. However, even in this study, patients were eligible if they achieved CR without GVHD. The consensus recommendations for DLI after haploidentical transplantation were published in 2020 [73]. A more sophisticated DLI could also be used to improve the efficacy/toxicity ratio. The negative selection of CD45RA+ CD3 cells was demonstrated to eliminate naïve CD3 cells responsible for GVHD [74]. In a small, proof-of-principle, prospective study, early DLI CD45RA+ depletion was well tolerated without an increase in GVHD frequency [75]. However, the anti-leukaemia activity of these compounds has not been fully analysed and, thus, must be evaluated in prospective studies. Ciurea et al. showed that prophylactic NK-DLI in high-risk AML patients was feasible and reduced the risk of relapse after haploidentical transplantation [76].
The reduction in the incidence of aGVHD via a sequential approach could be another way to improve the eligibility for prophylactic immunomodulation. Indeed, the occurrence of GVHD was one reason for not administering pDLI. Outside the haploidentical context, post-transplantation cyclophosphamide (PTCY) has been shown to be effective at reducing the incidence of GVHD in patients transplanted with matched-related or unrelated and mismatched unrelated donors [77,78,79]. Thus, PTCY plus other IS drugs can be integrated as part of an adapted approach to transplantation for RR AML patients, with the aim of reducing GVHD and allowing the timely use of the pDLI. Recently, the anti-CTLA4 inhibitor abatacept was tested in patients receiving transplants from MUDs or mMUDs, and it was found that abatacept was effective at reducing the incidence of grade 3–4 aGVHD, but the incidence of 2–4 aGVHD was still too high compared to that in historical ATG-free controls [80]. More interesting seems to be the introduction of 4-dose abatacept in the PTCY platform instead of MMF. This revised PTCY platform, after infusion of haploidentical peripheral stem cells, was promising for reducing the incidence of 2–4 and 3–4 aGVHD (17% and 4%, respectively) [81].
Finally, for the global prevention of relapse in ultrahigh-risk patients, any immunomodulatory manipulation can be associated with the use of target drugs such as azacitidine [82], anti-FLT3 drugs [83,84], and anti-IDH [85].

3. Conclusions

From this concise review, it is evident that the treatment of refractory AML is still an unmet clinical need and that, until recently, only allogenic stem cell transplantation has offered a chance to cure this disease. However, transplantation should be adapted to patients who are at a high risk of relapse. Each segment of the transplant process should be modified to maximise the antileukaemic effect without jeopardising tolerability.

Author Contributions

Conceptualization, L.C. and R.B.; methodology, L.C. and R.B.; software, not applicable; validation, L.C. and R.B.; formal analysis, L.C. and R.B.; investigation, not applicable; resources, not applicable; data curation, L.C.; writing—original draft preparation, L.C. and R.B.; writing—review and editing, L.C., R.B., G.S., S.T., C.R., A.M., V.C., A.S. and C.P.; visualization, not applicable; supervision, L.C.; project administration, not applicable; funding acquisition, not applicable. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Ferguson, P.; Hills, R.H.; Grech, A.; Betteridge, S.; Kjeldsen, L.; Dennis, M.; Vyas, P.; Goldstone, A.H.; Milligan, D.; Clark, R.E.; et al. An operational definition of primary refractory acute myeloid leukemia allowing early identification of patients who may benefit from allogeneic stem cell transplantation. Hematologica 2016, 101, 1351–1358. [Google Scholar] [CrossRef]
  2. Dohner, H.; Wei, A.H.; Appelbaum, F.R.; Craddock, C.; DiNardo, C.D.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Godley, L.A.; Hasserjian, R.P.; et al. Diagnosis management of AML in adults: 2022 recommendations from an International expert panel on behalf of the ELN. Blood 2022, 140, 1345–1377. [Google Scholar] [CrossRef] [PubMed]
  3. Thol, F.; Schlenk, R.F.; Heuser, M.; Ganser, A. How I treat refractory and early relapsed acute myeloid leukemia. Blood 2022, 126, 319–327. [Google Scholar] [CrossRef] [PubMed]
  4. Perl, A.E.; Martinelli, G.; Cortes, J.E.; Neubauer, A.; Berman, E.; Paolini, S.; Montesinos, P.; Baer, M.R.; Larson, R.A.; Ustun, C.; et al. Gilteritinib or Chemotherapy for Relapsed or Refractory FLT3-Mutated AML. N. Engl. J. Med. 2019, 381, 1728–1740. [Google Scholar] [CrossRef] [PubMed]
  5. Cortes, J.E.; Khaled, S.; Martinelli, G.; Perl, A.E.; Ganguly, S.; Russell, N.; Krämer, A.; Dombret, H.; Hogge, D.; Jonas, B.A.; et al. Quizartinib versus salvage chemotherapy in relapsed or refractory FLT3-ITD acute myeloid leukaemia (QuANTUM-R): A multicentre, randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2019, 20, 984–997. [Google Scholar] [CrossRef] [PubMed]
  6. DiNardo, C.D.; Pratz, K.; Pullarkat, V.; Jonas, B.A.; Arellano, M.; Becker, P.S.; Frankfurt, O.; Konopleva, M.; Wei, A.H.; Kantarjian, H.M.; et al. Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood 2019, 133, 7–17. [Google Scholar] [CrossRef]
  7. Todisco, E.; Papayannidis, C.; Fracchiolla, N.; Petracci, E.; Zingaretti, C.; Vetro, C.; Martelli, M.P.; Zappasodi, P.; Di Renzo, N.; Gallo, S.; et al. AVALON: The Italian cohort study on real-life efficacy of hypomethylating agents plus venetoclax in newly diagnosed or relapsed/refractory patients with acute myeloid leukemia. Cancer 2023, 129, 992–1004. [Google Scholar] [CrossRef]
  8. DiNardo, C.D.; Lachowiez, C.A.; Takahashi, K.; Loghavi, S.; Xiao, L.; Kadia, T.; Daver, N.; Adeoti, M.; Short, N.J.; Sasaki, K.; et al. Venetoclax Combined with FLAG-IDA Induction and Consolidation in Newly Diagnosed and Relapsed or Refractory Acute Myeloid Leukemia. J. Clin. Oncol. 2022, 39, 2768–2778. [Google Scholar] [CrossRef]
  9. Burnett, A.K.; Russell, N.H.; Hills, R.K.; Hunter, A.E.; Kjeldsen, L.; Yin, J.; Gibson, B.E.; Wheatley, K.; Milligan, D. Optimization of chemotherapy for younger patients with acute myeloid leukemia: Results of the medical research council AML15 trial. J. Clin. Oncol. 2013, 31, 3360–3368. [Google Scholar] [CrossRef]
  10. Burnett, A.K.; Russell, N.H.; Hills, R.K.; Kell, J.; Cavenagh, J.; Kjeldsen, L.; McMullin, M.F.; Cahalin, P.; Dennis, M.; Friis, L.; et al. A randomized comparison of daunorubicin 90 mg/m2 vs. 60 mg/m2 in AML induction: Results from the UK NCRI AML17 trial in 1206 patients. Blood 2015, 125, 3878–3885. [Google Scholar] [CrossRef]
  11. Burnett, A.; Cavenagh, J.; Russell, N.; Hills, R.; Kell, J.; Jones, G.; Nielsen, O.J.; Khwaja, A.; Thomas, I.; Clark, R.; et al. Defining the dose of gemtuzumab ozogamicin in combination with induction chemotherapy in acute myeloid leukemia: A comparison of 3 mg/m2 with 6 mg/m2 in the NCRI AML17 Trial. Haematologica 2016, 101, 724–731. [Google Scholar] [CrossRef]
  12. Castaigne, S.; Pautas, C.; Terré, C.; Raffoux, E.; Bordessoule, D.; Bastie, J.N.; Legrand, O.; Thomas, X.; Turlure, P.; Reman, O.; et al. Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): A randomised, open-label, phase 3 study. Lancet 2012, 379, 1508–1516. [Google Scholar] [CrossRef] [PubMed]
  13. Borthakur, G.; Cortes, J.E.; Estey, E.E.; Jabbour, E.; Faderl, S.; O’Brien, S.; Garcia-Manero, G.; Kadia, T.M.; Wang, X.; Patel, K.; et al. Gemtuzumab ozogamicin with fludarabine, cytarabine, and granulocyte colony stimulating factor (FLAG-GO) as front-line regimen in patients with core binding factor acute myelogenous leukemia. Am. J. Hematol. 2014, 89, 964–968. [Google Scholar] [CrossRef]
  14. Löwenberg, B.; Pabst, T.; Maertens, J.; van Norden, Y.; Biemond, B.J.; Schouten, H.C.; Spertini, O.; Vellenga, E.; Graux, C.; Havelange, V.; et al. Therapeutic value of clofarabine in younger and middle-aged (18–65 years) adults with newly diagnosed AML. Blood 2017, 129, 1636–1645. [Google Scholar] [CrossRef]
  15. Stone, R.M.; Mandrekar, S.J.; Sanford, B.L.; Laumann, K.; Geyer, S.; Bloomfield, C.D.; Thiede, C.; Prior, T.W.; Döhner, K.; Marcucci, G.; et al. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. N. Engl. J. Med. 2017, 377, 454–464. [Google Scholar] [CrossRef]
  16. Erba, H.P.; Montesinos, P.; Kim, H.J.; Patkowska, E.; Vrhovac, R.; Žák, P.; Wang, P.N.; Mitov, T.; Hanyok, J.; Kamel, Y.M.; et al. Quizartinib plus chemotherapy in newly diagnosed patients with FLT3-internal-tandem-duplication-positive acute myeloid leukaemia (QuANTUM-First): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2023, 401, 1571–1583. [Google Scholar] [CrossRef] [PubMed]
  17. Döhner, H.; Weber, D.; Krzykalla, J.; Fiedler, W.; Kühn, M.W.M.; Schroeder, T.; Mayer, K.; Lübbert, M.; Wattad, M.; Götze, K.; et al. Intensive chemotherapy with or without gemtuzumab ozogamicin in patients with NPM1-mutated acute myeloid leukaemia (AMLSG 09-09): A randomised, open-label, multicentre, phase 3 trial. Lancet Haematol. 2023, 10, e495–e509. [Google Scholar] [CrossRef]
  18. Russell, N.; Hills, R.; Kjeldsen, L.; Dennis, M.; Burnett, A. Treatment intensification with FLAG-Ida may improve disease control in younger patients with secondary acute myeloid leukaemia: Long-term follow up of the MRC AML15 trial. Br. J. Haematol. 2022, 196, 1344–1347. [Google Scholar] [CrossRef]
  19. Lancet, J.E.; Cortes, J.E.; Hogge, D.E.; Tallman, M.S.; Kovacsovics, T.J.; Damon, L.E.; Komrokji, R.; Solomon, S.R.; Kolitz, J.E.; Cooper, M.; et al. Phase 2 trial of CPX-351, a fixed 5:1 molar ratio of cytarabine/daunorubicin, vs cytarabine/daunorubicin in older adults with untreated AML. Blood 2014, 123, 3239–3246. [Google Scholar] [CrossRef]
  20. Lancet, J.E.; Uy, G.L.; Cortes, J.E.; Newell, L.F.; Lin, T.L.; Ritchie, E.K.; Stuart, R.K.; Strickland, S.A.; Hogge, D.; Solomon, S.R.; et al. CPX-351 (cytarabine and daunorubicin) Liposome for Injection Versus Conventional Cytarabine Plus Daunorubicin in Older Patients with Newly Diagnosed Secondary Acute Myeloid Leukemia. J. Clin. Oncol. 2018, 36, 2684–2692. [Google Scholar] [CrossRef]
  21. Rautenberg, C.; Stölzel, F.; Röllig, C.; Stelljes, M.; Gaidzik, V.; Lauseker, M.; Kriege, O.; Verbeek, M.; Unglaub, J.M.; Thol, F.; et al. Real-world experience of CPX-351 as first-line treatment for patients with acute myeloid leukemia. Blood Cancer J. 2021, 11, 164. [Google Scholar] [CrossRef]
  22. Chiche, E.; Rahmé, R.; Bertoli, S.; Dumas, P.Y.; Micol, J.B.; Hicheri, Y.; Pasquier, F.; Peterlin, P.; Chevallier, P.; Thomas, X.; et al. Real-life experience with CPX-351 and impact on the outcome of high-risk AML patients: A multicentric French cohort. Blood Adv. 2021, 5, 176–184. [Google Scholar] [CrossRef] [PubMed]
  23. Guolo, F.; Fianchi, L.; Minetto, P.; Clavio, M.; Gottardi, M.; Galimberti, S.; Rizzuto, G.; Rondoni, M.; Bertani, G.; Dargenio, M.; et al. CPX-351 treatment in secondary acute myeloblastic leukemia is effective and improves the feasibility of allogeneic stem cell transplantation: Results of the Italian compassionate use program. Blood Cancer J. 2020, 10, 96. [Google Scholar] [CrossRef] [PubMed]
  24. Othman, J.; Wilhelm-Benartzi, C.; Dillon, R.; Knapper, S.; Freeman, S.D.; Batten, L.M.; Canham, J.; Hinson, E.L.; Wych, J.; Betteridge, S.; et al. A randomized comparison of CPX-351 and FLAG-Ida in adverse karyotype AML and high-risk MDS: The UK NCRI AML19 trial. Blood Adv. 2023, 7, 4539–4549. [Google Scholar] [CrossRef] [PubMed]
  25. Minetto, P.; Candoni, A.; Guolo, F.; Clavio, M.; Zannier, M.E.; Miglino, M.; Dubbini, M.V.; Carminati, E.; Sicuranza, A.; Ciofini, S.; et al. Fludarabine, High-Dose Cytarabine and Idarubicin-Based Induction May Overcome the Negative Prognostic Impact of FLT3-ITD in NPM1 Mutated AML, Irrespectively of FLT3-ITD Allelic Burden. Cancers 2020, 13, 34. [Google Scholar] [CrossRef] [PubMed]
  26. Michallet, M.; Thomas, X.; Vernant, J.P.; Kuentz, M.; Socié, G.; Espérou-Bourdeau, H.; Milpied, N.; Blaise, D.; Rio, B.; Reiffers, J.; et al. Long-term outcome after allogeneic hematopoietic stem cell transplantation for advanced stage acute myeloblastic leukemia: A retrospective study of 379 patients reported to the Société Française de Greffe de Moelle (SFGM). Bone Marrow Transplant. 2000, 26, 1157–1163. [Google Scholar] [CrossRef] [PubMed]
  27. Abou Dalle, I.; Labopin, M.; Kröger, N.; Schroeder, T.; Finke, J.; Stelljes, M.; Neubauer, A.; Blaise, D.; Yakoub-Agha, I.; Salmenniemi, U.; et al. Impact of disease burden on clinical outcomes of AML patients receiving allogeneic hematopoietic cell transplantation: A study from the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation. Bone Marrow Transplant. 2023, 58, 784–790. [Google Scholar] [CrossRef] [PubMed]
  28. Stelljes, M.; Bornhauser, M.; Kroger, M.; Beyer, J.; Sauerland, M.C.; Heinecke, A.; Berning, B.; Scheffold, C.; Silling, G.; Buchner, T.; et al. Cooperative German Transplant Study Group. Conditioning with 8-Gy total body irradiation and fludarabine for allogeneic hematopoietic stem cell transplantation in acute myeloid leukemia. Blood 2005, 106, 3314–3321. [Google Scholar] [CrossRef] [PubMed]
  29. Ringdén, O.; Karlsson, H.; Olsson, R.; Omazic, B.; Uhlin, M. The allogeneic graft-versus-cancer effect. Br. J. Haematol. 2009, 147, 614–633. [Google Scholar] [CrossRef]
  30. Schlenk, R.F.; Döhner, K.; Mack, S.; Stoppel, M.; Király, F.; Götze, K.; Hartmann, F.; Horst, H.A.; Koller, E.; Petzer, A.; et al. Prospective evaluation of allogeneic hematopoietic stem-cell transplantation from matched related and matched unrelated donors in younger adults with high-risk acute myeloid leukemia: German-Austrian trial AMLHD98A. J. Clin. Oncol. 2010, 28, 4642–4648. [Google Scholar] [CrossRef]
  31. Luger, S.M.; Ringdén, O.; Zhang, M.J.; Pérez, W.S.; Bishop, M.R.; Bornhauser, M.; Bredeson, C.N.; Cairo, M.S.; Copelan, E.A.; Gale, R.P.; et al. Similar outcomes using myeloablative vs reduced-intensity allogeneic transplant preparative regimens for AML or MDS. Bone Marrow Transplant. 2012, 47, 203–211. [Google Scholar] [CrossRef] [PubMed]
  32. Pfrepper, C.; Klinke, A.; Behre, G.; Schenk, T.; Franke, G.N.; Jentzsch, M.; Schwind, S.; Al-Ali, H.K.; Hochhaus, A.; Niederwieser, D.; et al. Risk factors for outcome in refractory acute myeloid leukemia patients treated with a combination of fludarabine, cytarabine, and amsacrine followed by a reduced-intensity conditioning and allogeneic stem cell transplantation. J. Cancer Res. Cin. Oncol. 2016, 142, 317–324. [Google Scholar] [CrossRef] [PubMed]
  33. Todisco, E.; Ciceri, F.; Boschini, C.; Giglio, F.; Bacigalupo, A.; Patriarca, F.; Donnini, I.; Alessandrino, E.P.; Arcese, W.; Iori, A.P.; et al. Factors predicting outcome after allogeneic transplant in refractory acute myeloid leukemia: A retrospective analysis of Gruppo Italiano Trapianto di Midollo Osseo (GITMO). Bone Marrow Transplant. 2017, 52, 955–961. [Google Scholar] [CrossRef]
  34. Singhal, S.; Powles, R.; Henslee-Downey, P.J.; Chiang, K.Y.; Treleaven, J.; Godder, K.; Kulkarni, S.; van Rhee, F.; Sirohi, B.; Pinkerton, C.R.; et al. Allogeneic transplantation from HLA-matched sibling or partially HLA-mismatched related donors for primary refractory acute leukemia. Bone Marrow Transplant. 2002, 29, 291–295. [Google Scholar] [CrossRef] [PubMed]
  35. Oyekunle, A.A.; Kröger, N.; Zabelina, T.; Ayuk, F.; Schieder, H.; Renges, H.; Fehse, N.; Waschke, O.; Fehse, B.; Kabisch, H.; et al. Allogeneic stem-cell transplantation in patients with refractory acute leukemia: A long-term follow-up. Bone Marrow Transplant. 2006, 37, 45–50. [Google Scholar] [CrossRef] [PubMed]
  36. Craddock, C.; Labopin, M.; Pillai, S.; Finke, J.; Bunjes, D.; Greinix, H.; Ehninger, G.; Steckel, N.K.; Zander, A.R.; Schwerdtfeger, R.; et al. Factors predicting outcome after unrelated donor stem cell transplantation in primary refractory acute myeloid leukaemia. Leukemia 2011, 25, 808–813. [Google Scholar] [CrossRef] [PubMed]
  37. Hemmati, P.G.; Terwey, T.H.; Na, I.K.; Jehn, C.F.; le Coutre, P.; Vuong, L.G.; Dörken, B.; Arnold, R. Allogeneic stem cell transplantation for refractory acute myeloid leukemia: A single center analysis of long-term outcome. Eur. J. Haematol. 2015, 95, 498–506. [Google Scholar] [CrossRef]
  38. Yanada, M.; Harada, K.; Shimomura, Y.; Arai, Y.; Konuma, T. Conditioning regimens for allogeneic hematopoietic cell transplantation in acute myeloid leukemia: Real-world data from the Japanese registry studies. Front. Oncol. 2022, 12, 1050633. [Google Scholar] [CrossRef]
  39. Nagler, A.; Ngoya, A.; Galimard, J.E.; Labopin, M.; Bornhäuser, M.; Stelljes, M.; Finke, J.; Ganser, A.; Einsele, H.; Kröger, N.; et al. Longitudinal Outcome over Two Decades of Unrelated Allogeneic Stem Cell Transplantation for Relapsed/Refractory Acute Myeloid Leukemia: An ALWP/EBMT Analysis. Clin. Cancer Res. 2022, 28, 4258–4266. [Google Scholar] [CrossRef]
  40. Liu, N.; Ning, H.M.; Hu, L.D.; Jiang, M.; Xu, C.; Hu, J.W.; Wang, J.; Li, Y.H.; Li, B.T.; Lou, X.; et al. Outcome of myeloablative allogeneic peripheral blood hematopoietic stem cell transplantation for refractory/relapsed AML patients in NR status. Leuk. Res. 2015, 39, 1375–1381. [Google Scholar] [CrossRef]
  41. Baron, F.; Labopin, M.; Tischer, J.; Ciceri, F.; Raiola, A.M.; Blaise, D.; Sica, S.; Vydra, J.; Fanin, R.; Diez-Martin, J.L.; et al. Comparison of HLA-mismatched unrelated donor transplantation with post-transplant cyclophosphamide versus HLA-haploidentical transplantation in patients with active acute myeloid leukemia. Bone Marrow Transplant. 2022, 57, 1657–1663. [Google Scholar] [CrossRef]
  42. Duval, M.; Klein, J.; He, W.; Cahn, J.Y.; Cairo, M.; Camitta, B.M.; Kamble, R.; Copelan, E.; de Lima, M.; Gupta, V.; et al. Hematopoietic Stem-Cell Transplantation for Acute Leukemia in Relapse or Primary Induction Failure. J. Clin. Oncol. 2022, 28, 3730–3738. [Google Scholar] [CrossRef]
  43. Nagler, A.; Savani, B.N.; Labopin, M.; Polge, E.; Passweg, J.; Finke, J.; Kyrcz-Krzemien, S.; Volin, L.; Anagnostopoulos, A.; Aljurf, M.; et al. Outcomes after use of two standard ablative regimens in patients with refractory acute myeloid leukaemia: A retrospective, multicentre, registry analysis. Lancet Haematol. 2015, 2, e384–e392. [Google Scholar] [CrossRef] [PubMed]
  44. Ringdén, O.; Labopin, M.; Schmid, C.; Sadeghi, B.; Polge, E.; Tischer, J.; Ganser, A.; Michallet, M.; Kanz, L.; Schwerdtfeger, R.; et al. Sequential chemotherapy followed by reduced-intensity conditioning and allogeneic haematopoietic stem cell transplantation in adult patients with relapse or refractory acute myeloid leukaemia: A survey from the Acute Leukaemia Working Party of EBMT. Br. J. Haematol. 2017, 176, 431–439. [Google Scholar] [CrossRef] [PubMed]
  45. Duléry, R.; Ménard, A.L.; Chantepie, S.; El-Cheikh, J.; François, S.; Delage, J.; Giannotti, F.; Ruggeri, A.; Brissot, E.; Battipaglia, G.; et al. Sequential Conditioning with Thiotepa in T Cell- Replete Hematopoietic Stem Cell Transplantation for the Treatment of Refractory Hematologic Malignancies: Comparison with Matched Related, Haplo-Mismatched, and Unrelated Donors. Biol. Blood Marrow Transplant. 2018, 24, 1013–1021. [Google Scholar] [CrossRef] [PubMed]
  46. Steckel, N.K.; Groth, C.; Mikesch, J.H.; Trenschel, R.; Ottinger, H.; Kordelas, L.; Mueller-Tidow, C.; Schliemann, C.; Reicherts, C.; Albring, J.C.; et al. High-dose melphalan-based sequential conditioning chemotherapy followed by allogeneic haematopoietic stem cell transplantation in adult patients with relapsed or refractory acute myeloid leukaemia. Br. J. Haematol. 2018, 180, 840–853. [Google Scholar] [CrossRef] [PubMed]
  47. Rodríguez-Arbolí, E.; Labopin, M.; Tischer, J.; Brecht, A.; Ganser, A.; Finke, J.; Blau, I.W.; Kröger, N.; Kalhs, P.; Forcade, E.; et al. FLAMSA-Based Reduced-Intensity Conditioning versus Myeloablative Conditioning in Younger Patients with Relapsed/Refractory Acute Myeloid Leukemia with Active Disease at the Time of Allogeneic Stem Cell Transplantation: An Analysis from the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation. Biol. Blood Marrow Transplant. 2020, 26, 2165–2173. [Google Scholar] [PubMed]
  48. Le Bourgeois, A.; Labopin, M.; Marçais, A.; de Latour, R.P.; Blaise, D.; Chantepie, S.; N’Guyen, S.; Maillard, N.; Forcade, E.; Yakoub-Agha, I.; et al. Sequential allogeneic hematopoietic stem cell transplantation for active refractory/relapsed myeloid malignancies: Results of a reduced-intensity conditioning preceded by clofarabine and cytosine arabinoside, a retrospective study on behalf of the SFGM-TC. Ann. Hematol. 2020, 99, 1855–1862. [Google Scholar] [CrossRef] [PubMed]
  49. Sockel, K.; Stölzel, F.; Hönl, F.; Baldauf, H.; Röllig, C.; Wermke, M.; von Bonin, M.; Teipel, R.; Link-Rachner, C.; Brandt, K.; et al. Allogeneic Stem Cell Transplantation with Sequential Melphalan-Based Conditioning in AML: Residual Morphological Blast Count Determines the Risk of Relapse. Cancer Manag. Res. 2022, 14, 547–559. [Google Scholar] [CrossRef]
  50. Guijarro, F.; Bataller, A.; Diaz-Beyá, M.; Garrido, A.; Coll-Ferrà, C.; Vives, S.; Salamero, O.; Valcárcel, D.; Tormo, M.; Arnan, M.; et al. Long-term outcomes in patients with relapsed/refractory acute myeloid leukemia and other high-risk myeloid malignancies after undergoing sequential conditioning regimen based on IDA-FLAG and high-dose melphalan. Bone Marrow Transplant. 2022, 57, 1304–1312. [Google Scholar] [CrossRef]
  51. Weller, J.F.; Mezger, M.; Seifert, L.L.; Vogel, W.; Schneidawind, D.; Faul, C.; Bethge, W.; Lengerke, C.; Christopeit, M. Time-dependent analysis of adoptive immunotherapy following sequential FLAMSA-reduced intensity conditioning and allogeneic hematopoietic stem cell transplantation in patients with high-risk myeloid neoplasia. Eur. J. Haematol. 2022, 108, 244–263. [Google Scholar] [CrossRef]
  52. 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] [PubMed]
  53. Saraceni, F.; Labopin, M.; Brecht, A.; Kröger, N.; Eder, M.; Tischer, J.; Labussière-Wallet, H.; Einsele, H.; Beelen, D.; Bunjes, D.; et al. Fludarabine-treosulfan compared to thiotepa-busulfan-fludarabine or FLAMSA as conditioning regimen for patients with primary refractory or relapsed acute myeloid leukemia: A study from the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation (EBMT). J. Hematol. Oncol. 2019, 12, 44. [Google Scholar]
  54. Schmid, C.; Labopin, M.; Schaap, N.; Veelken, H.; Brecht, A.; Stadler, M.; Finke, J.; Baron, F.; Collin, M.; Bug, G.; et al. Long-term results and GvHD after prophylactic and preemptive donor lymphocyte infusion after allogeneic stem cell transplantation for acute leukemia. Bone Marrow Transplant. 2022, 57, 215–223. [Google Scholar] [CrossRef] [PubMed]
  55. Davies, J.K.; Hassan, S.; Sarker, S.J.; Besley, C.; Oakervee, H.; Smith, M.; Taussig, D.; Gribben, J.G.; Cavenagh, J.D. Durable graft-versus-leukaemia effects without donor lymphocyte infusions—Results of a phase II study of sequential T-replete allogeneic transplantation for high-risk acute myeloid leukaemia and myelodysplasia. Br. J. Haematol. 2018, 180, 346–355. [Google Scholar] [CrossRef]
  56. Penack, O.; Peczynski, C.; Mohty, M.; Yakoub-Agha, I.; Styczynski, J.; Montoto, S.; Duarte, R.F.; Kröger, N.; Schoemans, H.; Koenecke, C.; et al. How much has allogeneic stem cell transplant-related mortality improved since the 1980s? A retrospective analysis from the EBMT. Blood Adv. 2020, 4, 6283–6290. [Google Scholar] [CrossRef]
  57. Schmid, C.; Schleuning, M.; Schwerdtfeger, R.; Hertenstein, B.; Mischak-Weissinger, E.; Bunjes, D.; Harsdorf, S.V.; Scheid, C.; Holtick, U.; Greinix, H.; et al. Long-term survival in refractory acute myeloid leukemia after sequential treatment with chemotherapy and reduced-intensity conditioning for allogeneic stem cell transplantation. Blood 2006, 108, 1093–1099. [Google Scholar] [CrossRef]
  58. Middeke, J.M.; Herbst, R.; Parmentier, S.; Bug, G.; Hänel, M.; Stuhler, G.; Schäfer-Eckart, K.; Rösler, W.; Klein, S.; Bethge, W.; et al. Clofarabine salvage therapy before allogeneic hematopoietic stem cell transplantation in patients with relapsed or refractory AML: Results of the BRIDGE trial. Leukemia 2016, 30, 261–267. [Google Scholar] [CrossRef]
  59. Jaiswal, S.R.; Zaman, S.; Chakrabarti, A.; Sen, S.; Mukherjee, S.; Bhargava, S.; Ray, K.; O’Donnell, P.V.; Chakrabarti, S. Improved Outcome of Refractory/Relapsed Acute Myeloid Leukemia after Post-Transplantation Cyclophosphamide-Based Haploidentical Transplantation with Myeloablative Conditioning and Early Prophylactic Granulocyte Colony-Stimulating Factor–Mobilized Donor Lymphocyte Infusions. Blood Marrow Transplant. 2016, 22, 1867–1873. [Google Scholar]
  60. Mohty, M.; Malard, F.; Blaise, D.; Milpied, N.; Socié, G.; Huynh, A.; Reman, O.; Yakoub-Agha, I.; Furst, S.; Guillaume, T.; et al. Sequential regimen of clofarabine, cytosine arabinoside and reduced-intensity conditioned transplantation for primary refractory acute myeloid leukemia. Haematologica 2017, 102, 184–191. [Google Scholar] [CrossRef]
  61. Bonifazi, F.; Pavon, C.; Peccatori, J.; Giglio, F.; Arpinati, M.; Busca, A.; Bernasconi, P.; Grassi, A.; Iori, A.P.; Patriarca, F.; et al. Myeloablative conditioning with thiotepa-busulfan-fludarabine does not improve the outcome of patients transplanted with active leukemia: Final results of the GITMO prospective trial GANDALF-01. Bone Marrow Transplant. 2022, 57, 949–958. [Google Scholar] [CrossRef]
  62. Decroocq, J.; Itzykson, R.; Vigouroux, S.; Michallet, M.; Yakoub-Agha, I.; Huynh, A.; Beckerich, F.; Suarez, F.; Chevallier, P.; Nguyen-Quoc, S.; et al. Similar outcome of allogeneic stem cell transplantation after myeloablative and sequential conditioning regimen in patients with refractory or relapsed acute myeloid leukemia: A study from the Société Francophone de Greffe de Moelle et de Thérapie Cellulaire. Am. J. Hematol. 2018, 93, 416–423. [Google Scholar]
  63. Scott, B.L.; Pasquini, M.C.; Logan, B.R.; Wu, J.; Devine, S.M.; Porter, D.L.; Maziarz, R.T.; Warlick, E.D.; Fernandez, H.F.; Alyea, E.P.; et al. Myeloablative Versus Reduced-Intensity Hematopoietic Cell Transplantation for Acute Myeloid Leukemia and Myelodysplastic Syndromes. J. Clin. Oncol. 2017, 35, 1154–1161. [Google Scholar] [CrossRef]
  64. Alatrash, G.; Thall, P.F.; Valdez, B.C.; Fox, P.S.; Ning, J.; Garber, H.R.; Janbey, S.; Worth, L.L.; Popat, U.; Hosing, C.; et al. Long-Term Outcomes after Treatment with Clofarabine ± Fludarabine with Once-Daily Intravenous Busulfan as Pretransplant Conditioning Therapy for Advanced Myeloid Leukemia and Myelodysplastic Syndrome. Biol. Blood Marrow Transplant. 2016, 22, 1792–1800. [Google Scholar] [CrossRef]
  65. Tang, X.; Valdez, B.C.; Ma, Y.; Zhang, Q.; Qu, C.; Dai, H.; Yin, J.; Li, Z.; Xu, T.; Xu, Y.; et al. Low-dose decitabine as part of a modified Bu-Cy conditioning regimen improves survival in AML patients with active disease undergoing allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2021, 56, 1674–1682. [Google Scholar] [CrossRef] [PubMed]
  66. Gavriilaki, E.; Sakellari, I.; Labopin, M.; Bornhäuser, M.; Hamladji, R.M.; Casper, J.; Edinger, M.; Zák, P.; Yakoub-Agha, I.; Ciceri, F.; et al. Survival advantage of treosulfan plus fludarabine (FT14) compared to busulfan plus fludarabine (FB4) in active acute myeloid leukemia post allogeneic transplantation: An analysis from the European Society for Blood and Marrow Transplantation (EBMT) Acute Leukemia Working Party (ALWP). Bone Marrow Transplant. 2023, 58, 1084–1088. [Google Scholar] [PubMed]
  67. Andersson, B.S. Fludarabine-IV busulfan, dose-intensity and progression-free survival: Are we finally finding the way to reach a consensus opinion?: Higher busulfan dose intensity appears to improve leukemia-free and overall survival in AML allografted in CR2: An analysis from the acute leukemia working party of the European group for blood and marrow transplantation. Leuk. Res. 2016, 41, 5–6. [Google Scholar]
  68. Bramanti, S.; De Philippis, C.; Bartoli, A.; Giordano, L.; Mariotti, J.; Sarina, B.; Mannina, D.; Valli, V.; De Gregori, S.; Roperti, M.; et al. Feasibility and Efficacy of a Pharmacokinetics-Guided Busulfan Conditioning Regimen for Allogeneic Stem Cell Transplantation with Post-Transplantation Cyclophosphamide as Graft-versus-Host Disease Prophylaxis in Adult Patients with Hematologic Malignancies. Transplant. Cell Ther. 2021, 27, 912.e1–912.e6. [Google Scholar] [CrossRef]
  69. Beelen, D.W.; Stelljes, M.; Reményi, P.; Wagner-Drouet, E.M.; Dreger, P.; Bethge, W.; Ciceri, F.; Stölzel, F.; Junghanß, C.; Labussiere-Wallet, H.; et al. Treosulfan compared with reduced-intensity busulfan improves allogeneic hematopoietic cell transplantation outcomes of older acute myeloid leukemia and myelodysplastic syndrome patients: Final analysis of a prospective randomized trial. Am. J. Hematol. 2022, 97, 1023–1034. [Google Scholar] [CrossRef] [PubMed]
  70. Legrand, F.; Le Floch, A.C.; Granata, A.; Fürst, S.; Faucher, C.; Lemarie, C.; Harbi, S.; Bramanti, S.; Calmels, B.; El-Cheikh, J.; et al. Prophylactic donor lymphocyte infusion after allogeneic stem cell transplantation for high-risk AML. Bone Marrow Transplant. 2017, 52, 620–621. [Google Scholar] [CrossRef]
  71. Santoro, N.; Mooyaart, J.E.; Devillier, R.; Koc, Y.; Vydra, J.; Castagna, L.; Gülbas, Z.; Martin, J.D.; Araujo, M.C.; Kulagin, A.; et al. Donor lymphocyte infusions after haploidentical stem cell transplantation with PTCY: A study on behalf of the EBMT cellular therapy & immunobiology working party. Bone Marrow Transplant. 2023, 58, 54–60. [Google Scholar] [PubMed]
  72. Yan, C.H.; Liu, Q.F.; Wu, D.P.; Zhang, X.; Xu, L.P.; Zhang, X.H.; Wang, Y.; Huang, H.; Bai, H.; Huang, F.; et al. Prophylactic Donor Lymphocyte Infusion (DLI) Followed by Minimal Residual Disease and Graft-versus-Host Disease-Guided Multiple DLIs Could Improve Outcomes after Allogeneic Hematopoietic Stem Cell Transplantation in Patients with Refractory/Relapsed Acute Leukemia. Biol. Blood Marrow Transplant. 2017, 23, 1311–1319. [Google Scholar] [PubMed]
  73. Dholaria, B.; Savani, B.N.; Labopin, M.; Luznik, L.; Ruggeri, A.; Mielke, S.; Al Malki, M.M.; Kongtim, P.; Fuchs, E.; Huang, X.J.; et al. Clinical applications of donor lymphocyte infusion from an HLA-haploidentical donor: Consensus recommendations from the Acute Leukemia Working Party of the EBMT. Haematologica 2020, 105, 47–58. [Google Scholar] [CrossRef] [PubMed]
  74. Rheinländer, A.; Schraven, B.; Bommhardt, U. CD45 in human physiology and clinical medicine. Immunol. Lett. 2018, 196, 22–32. [Google Scholar] [CrossRef] [PubMed]
  75. Castagna, L.; Valli, V.; Timofeeva, I.; Capizzuto, R.; Bramanti, S.; Mariotti, J.; De Philippis, C.; Sarina, B.; Mannina, D.; Giordano, L.; et al. Feasibility and Efficacy of CD45RA+ Depleted Donor Lymphocytes Infusion after Haploidentical Transplantation with Post-Transplantation Cyclophosphamide in Patients with Hematological Malignancies. Transplant. Cell Ther. 2021, 27, 478.e1–478.e5. [Google Scholar] [CrossRef] [PubMed]
  76. Ciurea, S.O.; Schafer, J.R.; Bassett, R.; Denman, C.J.; Cao, K.; Willis, D.; Rondon, G.; Chen, J.; Soebbing, D.; Kaur, I.; et al. Phase 1 clinical trial using mbIL21 ex vivo-expanded donor-derived NK cells after haploidentical transplantation. Blood 2017, 130, 1857–1868. [Google Scholar] [CrossRef] [PubMed]
  77. Broers, A.E.C.; de Jong, C.N.; Bakunina, K.; Hazenberg, M.D.; van Marwijk Kooy, M.; de Groot, M.R.; van Gelder, M.; Kuball, J.; van der Holt, B.; Meijer, E.; et al. Posttransplant cyclophosphamide for prevention of graft-versus-host disease: Results of the prospective randomized HOVON-96 trial. Blood Adv. 2022, 6, 3378–3385. [Google Scholar] [CrossRef]
  78. Bolaños-Meade, J.; Hamadani, M.; Wu, J.; Al Malki, M.M.; Martens, M.J.; Runaas, L.; Elmariah, H.; Rezvani, A.R.; Gooptu, M.; Larkin, K.T.; et al. Post-Transplantation Cyclophosphamide-Based Graft-versus-Host Disease Prophylaxis. N. Engl. J. Med. 2023, 388, 2338–2348. [Google Scholar] [CrossRef]
  79. Shaw, B.E.; Jimenez-Jimenez, A.M.; Burns, L.J.; Logan, B.R.; Khimani, F.; Shaffer, B.C.; Shah, N.N.; Mussetter, A.; Tang, X.Y.; McCarty, J.M.; et al. National Marrow Donor Program-Sponsored Multicenter, Phase II Trial of HLA-Mismatched Unrelated Donor Bone Marrow Transplantation Using Post-Transplant Cyclophosphamide. J. Clin. Oncol. 2021, 39, 1971–1982. [Google Scholar] [CrossRef]
  80. Watkins, B.; Qayed, M.; McCracken, C.; Bratrude, B.; Betz, K.; Suessmuth, Y.; Yu, A.; Sinclair, S.; Furlan, S.; Bosinger, S.; et al. Phase II Trial of Costimulation Blockade with Abatacept for Prevention of Acute GVHD. J. Clin. Oncol. 2021, 39, 1865–1877. [Google Scholar] [CrossRef]
  81. Al-Homsi, A.S.; Cirrone, F.; Wo, S.; Cole, K.; Suarez-Londono, J.A.; Gardner, S.L.; Hsu, J.; Stocker, K.; Bruno, B.; Goldberg, J.D.; et al. PTCy, abatacept, and a short course of tacrolimus for GVHD prevention after haploidentical transplantation. Blood Adv. 2023, 7, 3604–3611. [Google Scholar] [CrossRef] [PubMed]
  82. Oran, B.; de Lima, M.; Garcia-Manero, G.; Thall, P.F.; Lin, R.; Popat, U.; Alousi, A.M.; Hosing, C.; Giralt, S.; Rondon, G.; et al. A phase 3 randomized study of 5-azacitidine maintenance vs observation after transplant in high-risk AML and MDS patients. Blood Adv. 2020, 4, 580–5588. [Google Scholar] [CrossRef] [PubMed]
  83. Burchert, A.; Bug, G.; Fritz, L.V.; Finke, J.; Stelljes, M.; Röllig, C.; Wollmer, E.; Wäsch, R.; Bornhäuser, M.; Berg, T.; et al. Sorafenib maintenance after allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia with FLT3-internal tandem duplication mutation (SORMAIN). J. Clin. Oncol. 2020, 38, 2993–3002. [Google Scholar] [CrossRef] [PubMed]
  84. Xuan, L.; Wang, Y.; Yang, K.; Shao, R.; Huang, F.; Fan, Z.; Chi, P.; Xu, Y.; Xu, N.; Deng, L.; et al. Sorafenib maintenance after allogeneic haemopoietic stem-cell transplantation in patients with FLT3-ITD acute myeloid leukaemia: Long-term follow-up of an open-label, multicentre, randomised, phase 3 trial. Lancet Haematol. 2023, 10, e600–e611. [Google Scholar] [CrossRef]
  85. DiNardo, C.D.; Stein, E.M.; de Botton, S.; Roboz, G.J.; Altman, J.K.; Mims, A.S.; Swords, R.; Collins, R.H.; Mannis, G.N.; Pollyea, D.A.; et al. Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML. N. Engl. J. Med. 2018, 378, 2386–2398. [Google Scholar] [CrossRef]
Figure 1. Complete remission rate in low-intermediate and high-risk AML using different induction chemotherapy. GO = gemtuzumab ozogamycin; FLAGIda: fludarabine, cytarabine, GCSF, idarubicine; CLAI: clofarabine, cytarabine, idarubicine; mido: midostaurin; quizar: quizartinib [9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25].
Figure 1. Complete remission rate in low-intermediate and high-risk AML using different induction chemotherapy. GO = gemtuzumab ozogamycin; FLAGIda: fludarabine, cytarabine, GCSF, idarubicine; CLAI: clofarabine, cytarabine, idarubicine; mido: midostaurin; quizar: quizartinib [9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25].
Cells 13 00755 g001
Table 1. Studies in refractory patients treated with standard conditioning.
Table 1. Studies in refractory patients treated with standard conditioning.
NAgeInclusion Criteria% Blasts (Median)Donor TypeConditioningGvHD ProphylaxisIS TaperingpDLICIRLFSOSNRM
Duval 2010 [42] 167338PIF, untreated, refractory
relapse
21%MRD/MUDMACFK/CyA +/− MTX
T-cell depletion (13%)
NRNoNRNR19%38%
Craddock 2011 [36] 16840PIF38%MUDMAC/RIC NRNoNR20%@5y22%@5yNR
Hemmati 2014 [37] 13152PIF and relapse22%MRD/MUDMAC/RIC
FLAMSA-RIC (21%)
CyA + MTX/MMFMRD +30
MUD +60
Yes48%@5y25%@5y/26%@3y
Liu 2015 [40] 13340, 30
21
PIF and relapse26%MRD/MUD
Haplo
MACCyA + MTX
CyA + MTX + MMF
GIAC protocol
NRNoNR36%@3y40%@3y19%@3y
Nagler 2015 [43] 85243
39
PIF and relapse20%
16%
MRD/MUDBUCY
TBICY
CyA + MTX
ATG
NRNo53%@2y
54%@2y
25%@2y
28%@2y
31%@2y
33%@2y
21%@2y
17%@2y
Todisco 2017 [33] 22749PIF>25%MRD/MUD
Haplo
CB
MAC 69%
RIC 31%
T-cell depletion 50%NRNo61%@3y23%@y14%@3y27%@3y
Nagler 2022 [39] 343055PIF and relapseNRMRD/MUDHaploMAC 54%
RIC 46%
FLAMSA-RIC 13%
CyA + MTX/MMF
ATG 78%
PTCY 4%
NRno48%@2y28%@2y36%@2y24%@2y
Baron 2022 [41] 21956PIF and relapseNRmMUD/HaploMAC/RICPTCY-basedNRno40%@2y
50%@2y
42%@2y
26%@2y
46%@2y
28%@2y
18%@2y
24%@2y
Yanada 2023 [38] 692753PIF and relapseNRMRD/MUD CBMAC 67%
RIC 33%
FK/CyA-basedNRno53%@5yNR23%@5y27%@5y
PIF = primary induction failure; MRD = matched related donor; MUD = matched unrelated donor; mMUD = mis-matched unrelated donor; MAC = myeloablative conditioning regimens; RIC = reduced intensity conditioning regimens; FK = tacrolimus; CyA = cyclosporine A; MTX = methotrexate; MMF = mycophenolate mofetil; ATG = anti-thymocyte globulin; PTCY = post-transplantation cyclophosphamide; GIAC = Granulocyte Intensified immunosuppression Antithymocyte globulin Combination pf peripheral blood and bone marrow; NR = nor reported; IS immunosuppression; pDLI = prophylactic donor lymphocyte infusion; CIR = cumulative incidence of relapse; LFS = leukaemia free survival; OS = overall survival; NRM = no-relapse mortality.
Table 2. Retrospective studies with sequential protocols.
Table 2. Retrospective studies with sequential protocols.
NAgeInclusion Criteria% Blasts (Median)DonorSequential CTRestConditioningGvHD ProphylaxisIS TaperingpDLICIROSLFSNRM
Ringden 2017 [44] 26751PIF and relapseNRMRD/MUDFLAMSA3dTBI4Gy, Cy
BU-based
PAM
CyA + MTX/MMF
ATG
NRno48%@3y30%@3y26%@3y26%@3y
Dulery 2018 [45] 7254PIF, first/second relapseNRMRD/MUD
haplo
TEC3dBU6.4FLUCyA + MMF
ATG
+60yes38%@2y57%@2yNR24%@2y
Steckel 2018 [46] 29256Primary refractory
Untreated relapse
32%MRD/MUDPAM1405dTBI8Gy/FLU
TREO/FLU
CyA ± MTX/MMF
ATG
NRno34%@1y34%@3y31%@3y36%@1y
Saraceni 2019 [47] 85651–58PIF and relapseNRMRD/MUDFLAMSA
/
/
NRBU/TBI-based
TREO/FLU
TBF
CyA + MTX/MMF
ATG
NRyes53%@2y
46%@2y
54%@2y
34%@2y
37%@2y
24%@2y
27%@2y
22%@2y
29%@2y
20%@2y
26%@2y
24%@2y
Rodrìguez-Arbolì, 2020 [47] 101839PIF and relapseNRMRD/MUDFLAMSANRTBI-based
CT-based
MAC
CyA+ MTX/MMF
ATG
NRNR55%@2y
53%@2y
51%@2y
36%@2y
50%@2y
33%@2y
27%@2y
40%@2y
30%@2y
18%@2y
7%@2y
19%@2y
Le Bourgeois 2020 [48] 13152PIF and relapseNRMRDClofaARAC3dBU9.6CYCyA + MTX/MMF
ATG
NRno45%@2y38%@2y29%@2y35%@2y
Sockel 2022 [49] 17356Relapse (36%)
first line
10%MRD/MUD
haplo
ClofaARAC/FLU-PAM
Clofa-PAM
CyA + MTX/MMF
PTCY
NRno30%@4y43%@4yNR36%@4y
Guijarro 2022 [50] 14055PIF or relapse20%MRD/MUD
haplo
FLAG-IDA3dPAM140 mg/m2CyA + MTX/MMF
ATG
PTCY
NRno30%@5y25%@5yNR45%@5y
Weller 2022 [51] 11460PIF or relapse17%MRD/MUD
haplo
FLAMSA3dRICCyA + MTX/MMF
ATG/PTCY
+90yes41%@2y45%@2y46%@2y (no DLI)
70%@2y (DLI)
27%@2y
TEC = thiotepa 5 mg/kg, etoposide 400 mg/m2, cyclophosphamide 2 g/m2. PAM = melphalan; PIF = primary induction failure; MRD = matched related donor; MUD = matched unrelated donor; mMUD = mis-matched unrelated donor; MAC = myeloablative conditioning regimens; RIC = reduced intensity conditioning regimens; FK = tacrolimus; CyA = cyclosporine A; MTX = methotrexate; MMF = mycophenolate mofetil; ATG = anti-thymocyte globulin; PTCY = post-transplantation cyclophosphamide; NR = nor reported; IS immunosuppression; pDLI = prophylactic donor lymphocyte infusion; CIR = cumulative incidence of relapse; LFS = leukaemia free survival; OS = overall survival; NRM = no-relapse mortality.
Table 3. Prognostic factors retrieved from standard conditioning studies.
Table 3. Prognostic factors retrieved from standard conditioning studies.
OSHRLFSHRCIRHRNRMHR
Duval 2010 [42] Duration of first CR < 6 months1.26Not done Not done Not done
Duration of first CR > 6 months0.83
Blasts PB at transplantation1.48
HLA familiar other related1.48
mMUD2.21
KPS > 90%0.65
Craddock 2011 [36] >3 CT1.66>3 CT1.63Not done Not done
BM blast > median (38%)1.49BM blast > median (38%)1.53
Recipient CMV+1.63Recipient CMV+1.67
Hemmati 2015 [37] Not done BM blast > 20%1.58BM blast > 20%1.7Not done
Any cGVHD0.21Any cGVHD0.18
Any aGVHD0.39
Nagler 2015 [43] Second relapse1.5Second relapse1.54First relapse1.24CYTBI0.69
Second relapse1.73Recipient age1.24
Todisco 2017 [33] >2 CT1.87Not done Not done Not done
BM blasts ≥ 25%/any level in PB1.75
KPS < 90%1.43
Recipient age > 601.77
Int/adverse cytogenetic1.44
Nagler 2022 [39] Recent period HSCT0.86Recent period HSCT0.87Recent period HSCT0.85mMUD 9/10
Recipient CMV+
1.31
1.39
Recipient age1.05mMUD 9/101.14Recipient age0.94
mMUD 9/101.2Recipient CMV+1.13TBI1.2
Recipient CMV+1.2Relapse1.1Relapse1.21
Poor Cytogenetic1.33Poor Cytogenetic1.51Poor Cytogenetic1.96
Yanada 2022 [38] Recipient age 40–49 y1.29Not done PS 2–41.13Recipient age 40–49 y1.38
Recipient age 50–59 y1.52Poor cytogenetic1.6Recipient age 50–59 y1.72
Recipient age > 60 y1.74Unevaluable cytogenetic1.23Recipient age > 60 y2.25
Recipient male1.22PB blasts 1–4%1.29Recipient male1.23
PS 2–41.79PB blasts 5–19%1.49PS 2–41.21
PIF0.93PB blasts > 20%1.77Poor cytogenetic0.84
Poor cytogenetic1.66CB0.8PB blasts > 20%0.85
Unevaluable cytogenetic1.5FK-based prophylaxis0.91CB1.20
PB blasts 1–4%1.18Year transplant 2016–20200.821Year transplant 2016–20200.781
PB blasts 5–19%1.38
PB blasts > 20%1.75
Year transplant 2011–20150.83
Year transplant 2016–20200.74
PIF: primary induction failure; UR: untreated relapse; SR: secondary relapse; MAC: myeloablative conditioning; RIC: reduced intensity conditioning. MUD, matched unrelated donor; MRD, matched related donor; PS, performance status; KPS: Karnofsky performance status; PB: peripheral blood; BM: bone marrow; mMUD = mis-matched unrelated donor; CB: cord blood.
Table 4. Prognostic scores from conventional allo-SCT studies.
Table 4. Prognostic scores from conventional allo-SCT studies.
NScore Components and PointsScoreOSLFS
Duval 2010 [42] 1673PIF or CR duration > 6 M = 0
CR duration < 6 M = 1
Cytogenetic good/INT = 0
Cytogenetic poor = 1
HLA match = 0
mMUD = 1
Haplo = 2
Circulating blasts yes = 1
KPS < 90 = 1
0 (1 point)
1 (1 point)
2 (2 points)
≥3 (3 points)
42%
28%
15%
6%
Not done
Craddock 2011 [36] 168>2 CT = 1
BM blasts > median = 1
R CMV+ = 1
0 (0 point)
1 (1 point)
2 (2 points)
3 (3 points)
44%
24%
10%
0%
40%
24%
12%
0%
Todisco 2017 [33] 227>2 CT = 1
BM blasts >25%/circulating any level = 1
Age > 60 = 1
Cytogenetic poor = 1
0 (0–1 points)
1 (2 points)
2 (>2 points)
32%@3y
10%@3y
0%@3y
Not done
PIF: primary induction failure; CR: complete remission; mMUD: mis-matched unrelated donor; KPS: Karnofsky performance status; CT: chemotherapy; R: recipient.
Table 5. Significant prognostic factors retrieved in sequential conditioning studies.
Table 5. Significant prognostic factors retrieved in sequential conditioning studies.
OSHRLFSHRCIRHRNRMHR
Ringden [48] In vivo T-cell depletion0.46In vivo T-cell depletion0.49UD (vs. MRD)0.6Recipient age
UD (vs. MRD)
In vivo T-cell depletion
1.33
1.96
0.35
Steckel [44] Recipient age > 59 y
Time from diagnosis to HSCT < 9M
>20% blasts (BM/PB)
mMUD
adverse
adverse
adverse
adverse
Not done Not done Recipient age > 59 y
HCT-CI ≥ 2
mMUD
Infection before conditioning
NR
Dulery [54] None Not done Not done KPS < 90%3
Saraceni [47] KPS ≥ 80%
Recipient CMV+
0.7
1.3
Recipient CMV+1.4Age
Relapse (vs. primary refr)
Recipient CMV+
0.9
1.3
1.3
Recipient age
mMUD
1.3
1.8
Rodrìguez-Arbolì [45] FLAMSA-CT
Adverse/failed cytogenetic
Second relapse
KPS > 90%
UD (vs. MRD)
0.65
2.13/2.02
1.88
0.54
1.23
FLAMSA-CT
Adverse/failed cytogenetic
Second relapse
KPS > 90%
0.73
2.07/1.99
1.78
0.64
Recipient age
Adverse/failed cytogenetic
Second relapse
KPS > 90%
0.85
2.93/2.55
1.94
0.67
FLAMSA-CT
Recipient age
KPS > 90%
UD (vs. MRD)
0.4
1.28
0.57
1.94
Le Bourgeois [50] CMV−/−1.75CMV−/−1.71CMV−/−2.49None
Sockel [9] >20% blasts (BM)
Recipient age
AML therapy related
1.8
1.26
2.10
Not done 5–20% blasts (BM)
>20% blasts (BM)
1.18
1.24
AML therapy related
Recipient age
UD
3.39
1.26
2.46
Guijarro [49] Recipient age > 55 y2.56Not done Adverse cytogenetic2.65Recipient age > 55 y2.4
UD: unrelated donor; mMUD: mis-matched unrelated donor; MRD: matched related donor; KPS: Karnofsky performance status; OS: overall survival; LFS: leukaemia free survival; CIR: cumulative incidence of relapse; NRM: no relapse mortality; HR: hazard ratio.
Table 6. Main results from prospective studies.
Table 6. Main results from prospective studies.
NMedian AgeInclusion Criteria% Blasts at ALLOCRSequentialRestCTXDonorGVHD ProphylaxisIS TaperingProphylactic DLICIROSLFSNRM
Schmid 2005 [52] 7552 y (18–65)No response to HD ARAC
Relapse 3 M after CR
Second relapse
Delayed response to IC
Relapse after auto
Secondary AML/MDS
NR11%FLAMSA-RIC *3 daysTBI4Gy, CyMRD 49%
MUD 51%
CSA day −1
MMF Day 0
ATG
CSA day +60 to 90
MMF day +50
24%
Day +120
Median day +160
20%42%@2y40%@2y33%@1y
Schmid 2006 [57] 10351 y (18–68)PIF after ≥2 IC
Relapse 6 M after CR
Refractory to salvage IC
≥2nd relapse
30% (0–90%)4%FLAMSA-RIC *3 daysTBI4Gy, CyMRD 40%
MUD 60%
CSA day −1
MMF Day 0
ATG
CSA day +60 to 90
MMF day +50
24%
Day +120
Median day +159
37%40@2y39%@2y17%@1y
Middeke 2016 [58] 8461 y (40–75)PIF after ≥2 IC
Relapsed
54% (5–92%)NoneClofaARACALLO in aplasiaClofaPAMMRD 18%
MUD 54%
mMUD 29%
CSA day −1
MMF Day 0
ATG (only in mMUD)
Not reportedNo26%@2y43@2yDFS 52%23%@2y
Jaiswal 2016 [59] ^4126 y (2–65)PIF after ≥2 IC
Relapsed refractory
14–16% (5–65%)Noneno/BUFLUPAM ^HaploPTCY day +3 and 4
CSA day +5
MMF day +5
CSA day +60
MMF from +14 to +21
90%
Day +21, +35, +60
43%
21% with DLI
53%@18 M
70% with DLI
35% w/out
44%@18 M
62% with DLI
25% w/out
19%@1y
Mohty 2017 [60] 2447 y (20–57)PIF after 2 IC
Persisting hypoplasia
20% (6–82%)NoneClofaARAC-RIC §3 daysBUCYMRD 63%
UD 37%
CSA day −1
MMF Day 0 (only in UD)
ATG
CSA day +90
MMF +62 to 90
25%
Day +120
54%@2y38%@2y29%@2y12%@2y
Davies 2018 [55] 4753 y (23–68)PIF after 1 IC
Relapse 6 M after CR
NRNoneDaunoARAC-RIC3 daysFLUCYMRD 49%
MUD 51%
CSA day −1
Short MTX
CSA day +90No30%@3y39%@2y39%@2y35%@1y
Bonifazi 2022 [61] 10154 y (16–69)PIF
Relapse
30% in PBNoneno/TBF °MUD 57%
Haplo 38%
CB 9%
ATG-based
PTCY-based
NRNo53%19%@2y19%@2y35%@1y
IC: induction chemotherapy; PB: peripheral blood; CTX: conditioning regimen; IS: immunosuppression; OS: overall survival; LFS: leukaemia-free survival; CIR: cumulative incidence of relapse; NRM: no relapse mortality; HR: hazard ratio. * RIC consisted of total body irradiation (4 Gy), ATG, and cyclophosphamide. § The RIC consisted of busulfan 6.4 mg/kg + cyclophosphamide 60 mg/kg. ^ excluding 10 patients treated with a nonmyeloablative conditioning regimen. The MAC consisted of fludarabine 150 mg/m2, busulfan 9.6 mg/kg, and malphalan 140 mg/m2. ° MAC consisted of thiotepa 10 mg/kg, busulfan 9.6 mg/kg, and fludarabine 150 mg/m2.
Table 7. Factors predictive of outcome in prospective studies.
Table 7. Factors predictive of outcome in prospective studies.
NSequentialpDLIOSLFSNRM
Schmid 2005 [52] 75FLAMSA-RICyesCD34CD34CD34
MRD
Schmid 2006 [57] 103FLAMSA-RICyesCD34
<2 IC
CD34
<2 IC
/
Middeke 2016 [58] 84ClofaARACNoAge
No response on day +15
//
Jaiswal 2016 [59] 41noyesMAC + pDLIMAC + pDLI/
Mohty 2017 [60] 24ClofaARAC-RICyes///
Davies 2018 [55] 47DaunoARAC-RICNocGVHDcGVHD/
Bonifazi 2022 [61] 101noNoHCT-CI > 0
Low risk cytogenetic
/HCT-CI > 0
pDLI = prophylactic DLI; HCT-CI: hematopoietic cell transplantation-comorbidity index; MAC: myeloablative conditioning regimen; cGVHD: chronic graft versus host disease.
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

Bono, R.; Sapienza, G.; Tringali, S.; Rotolo, C.; Patti, C.; Mulè, A.; Calafiore, V.; Santoro, A.; Castagna, L. Allogeneic Stem Cell Transplantation in Refractory Acute Myeloid Leukaemia. Cells 2024, 13, 755. https://doi.org/10.3390/cells13090755

AMA Style

Bono R, Sapienza G, Tringali S, Rotolo C, Patti C, Mulè A, Calafiore V, Santoro A, Castagna L. Allogeneic Stem Cell Transplantation in Refractory Acute Myeloid Leukaemia. Cells. 2024; 13(9):755. https://doi.org/10.3390/cells13090755

Chicago/Turabian Style

Bono, Roberto, Giuseppe Sapienza, Stefania Tringali, Cristina Rotolo, Caterina Patti, Antonino Mulè, Valeria Calafiore, Alessandra Santoro, and Luca Castagna. 2024. "Allogeneic Stem Cell Transplantation in Refractory Acute Myeloid Leukaemia" Cells 13, no. 9: 755. https://doi.org/10.3390/cells13090755

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