Next Article in Journal
Ocular Morbidity—A Critical Analysis to Improve Outpatient Services in an Eye Department in a Sub-Saharan Megacity
Next Article in Special Issue
Biologic and Therapeutic Implications of Genomic Alterations in Acute Lymphoblastic Leukemia
Previous Article in Journal
Spinal Cord Motion in Degenerative Cervical Myelopathy: The Level of the Stenotic Segment and Gender Cause Altered Pathodynamics
Previous Article in Special Issue
Antibody–Drug Conjugates for the Treatment of Acute Pediatric Leukemia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Allogeneic Hematopoietic Stem Cell Transplantation in Pediatric Leukemia

1
Department of Pediatric Hematology and Oncology, Scientific Institute for Research and Healthcare (IRCCS), Bambino Gesù Childrens’ Hospital, 00165 Rome, Italy
2
Department of Pediatrics, Sapienza University of Rome, 00185 Rome, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2021, 10(17), 3790; https://doi.org/10.3390/jcm10173790
Submission received: 5 July 2021 / Revised: 8 August 2021 / Accepted: 19 August 2021 / Published: 25 August 2021
(This article belongs to the Special Issue Advance in the Treatment of Pediatric Leukemia)

Abstract

:
Allogeneic hematopoietic stem cell transplantation (HSCT) offers potentially curative treatment for many children with high-risk or relapsed acute leukemia (AL), thanks to the combination of intense preparative radio/chemotherapy and the graft-versus-leukemia (GvL) effect. Over the years, progress in high-resolution donor typing, choice of conditioning regimen, graft-versus-host disease (GvHD) prophylaxis and supportive care measures have continuously improved overall transplant outcome, and recent successes using alternative donors have extended the potential application of allotransplantation to most patients. In addition, the importance of minimal residual disease (MRD) before and after transplantation is being increasingly clarified and MRD-directed interventions may be employed to further ameliorate leukemia-free survival after allogeneic HSCT. These advances have occurred in parallel with continuous refinements in chemotherapy protocols and the development of targeted therapies, which may redefine the indications for HSCT in the coming years. This review discusses the role of HSCT in childhood AL by analysing transplant indications in both acute lymphoblastic and acute myeloid leukemia, together with current and most promising strategies to further improve transplant outcome, including optimization of conditioning regimen and MRD-directed interventions.

1. Introduction

Despite the remarkable achievements obtained with frontline chemotherapy in the treatment of children with acute leukemia (AL), a significant proportion of patients still are treated with allogeneic hematopoietic stem cell transplantation (HSCT), either in first complete remission (CR1) or beyond, to achieve definitive disease eradication [1,2,3,4,5]. Over the years, the outcome of allogeneic HSCT for AL has continuously improved thanks to progress in high-resolution HLA typing, choice of conditioning regimen and supportive care measures. Furthermore, mainly owing to fundamental advances in graft manipulation techniques and graft-versus-host disease (GvHD) prophylaxis, results obtained using alternative donors are no longer inferior to those achieved using HLA-identical siblings and fully matched volunteers [6]. At the same time, successes in the transplant field are constantly paralleled by refinements in chemotherapy protocols, which take advantage of continuous breakthroughs of genomic medicine to achieve better treatment stratification and identify genetic lesions targetable with precision medicine approaches [7,8]. Such targeted approaches may be employed, in their turn, to further improve HSCT outcome by inducing a better remission status before transplant, especially in light of the robust evidence showing that pre- and post-HSCT minimal residual disease (MRD) represents one of the major determinants of subsequent relapse and long-term prognosis in AL [9,10]. This observation is particularly relevant in B-cell acute lymphoblastic leukemia (ALL), owing to the availability of highly effective immunotherapy approaches capable of inducing MRD-negative remissions of otherwise refractory and untreatable diseases [11]. The purpose of this review is to discuss the current role of HSCT in childhood AL by scrutinizing transplant indications in both ALL and acute myeloid leukemia (AML). We also discuss most promising practices to further improve transplant outcome, by analyzing (1) the predictive role of MRD and potential MRD-directed interventions, before and after HSCT; (2) the choice of conditioning regimen and (3) most recent results obtained with the use alternative donors.

2. HSCT Indications in ALL

Since most children with ALL have a good prognosis with current frontline protocols, only a minority of these patients are eligible for HSCT in CR1. On the contrary, most patients who experience a relapse are candidates for HSCT [12]. Despite the criteria used to assess this indication may vary between different cooperative groups, most studies/protocols consider eligible children with an estimated EFS probability lower than 50%, according to response to induction treatment and specific biologic features. Advances in biological characterization, improved MRD measurement capacity and its incorporation in treatment protocols to optimize risk stratification, and the upfront introduction of new biological agents (e.g., blinatumomab in AIEOP-BFM (EUDRACT 2016-001935-12) and COG protocols (AALL1731) and inotuzumab (AALL1732) and tisagenlecleucel (AALL1721/CASSIOPEIA) in COG protocol) have changed (and will continue to shape) the indications for HSCT over time [13]. Current HSCT indications in childhood ALL are summarized in Table 1. For example, criteria that were considered in past studies, such as a high white blood cell (WBC) count at presentation, Philadelphia chromosome positivity (Ph+), or poor-prednisone response, are no longer considered strict indications for transplantation [13]. The strongest criterion for HSCT indication is the response of the disease to induction therapy, this being a surrogate marker of leukemia sensitivity to chemotherapy. Indeed, MRD at selected time points is currently the single most powerful prognostic factor in childhood ALL [14]. Both multiparametric flow cytometry or quantitative PCR for specific rearrangements of immune-receptor genes IgH/TCR are currently used for MRD assessment [15]. The role of MRD for identification of HSCT candidates has been clearly proven by several groups, including The Italian Association of Pediatric Haematology–Oncology (AIEOP)/Berlin–Frankfurt–Muenster (BFM) group [16].
Additional criteria are employed to identify those patients who would benefit from HSCT in the first CR. Differing from AML, PIF is infrequent in pediatric ALL (being observed in less than 2% of patients); regardless, its occurrence represents a strong indication for HSCT in CR1, as shown by Schrappe and colleagues in a large retrospective study [17]. Hypodiploidy (<44 chromosomes) defines a high-risk group of patients with a poor outcome (the lowest the number of chromosome, the worst the prognosis) [18]. Thus, hypodiplody has been considered a strong indication to HSCT, although two recent retrospective studies, one from COG [19], the other from 16 cooperative groups [20] did not show an advantage for patients transplanted in CR1. Patients carrying the t(17;19)(q22;p13) translocation, resulting into the fusion gene TCF3-HLF (E2A-HLF), have a dismal outcome, irrespectively of MRD clearance [21]. For this reason, these patients are considered eligible for experimental therapies (e.g., BCL-2 inhibitors or anti-CD19-directed immunotherapies) and for HSCT in CR1 [22]. Deletions of Ikaros (IKZF1), a zinc-finger transcription factor required for the development of all lymphoid lineages, have been shown to confer an increased risk of recurrence [23]. In a recent paper, Stanulla and colleagues showed that IKZF1 deletions that co-occurred with deletions in CDKN2A, CDKN2B, PAX5 or PAR1 in the absence of ERG deletion were associated with the worst outcome and were grouped as IKZF1plus [24]. The IKZF1plus prognostic effect differed dramatically according to the MRD levels after induction treatment; in particular, IKZF1plus patients with intermediate and high-risk MRD had a miserable outcome and are now considered candidates to receive HSCT in CR1. Rearrangements involving the KMT2A (MLL) gene on chromosome 11q23 are observed in a large part of infants ALL; on the contrary, in children older than 1 year, they are much less frequent [25]. KMT2A-rearrangments represent a high-risk feature [26] justifying the use of HSCT in CR1, although for children with these abnormalities the indication to HSCT is determined in combination with (i) age less than 6 months and either poor response to steroids or leukocytes ≥300 × 109/L at diagnosis [27] for infant ALL, and (ii) MRD clearance. Intrachromosomal amplification of chromosome 21 is a rare recurrent lesion (found in less than 3% cases of pediatric ALL). The sole use of standard chemotherapy in this population has been associated with an increased risk of relapse [28]. Studies conducted in the UK have reported improved survival with the use of front-line HSCT [29]. Since patients with T-ALL cannot still benefit from immunotherapy approaches (including CAR-T cells and BiTEs), they have a lower chance of being rescued when a relapse occurs. In addition, for this reason, patients with poor MRD clearance at the end of induction are considered candidates for HSCT in CR1.
Indications to HSCT for children with ALL who experience a first relapse are currently based on disease immunophenotype (T/B ALL) time elapsing between diagnosis and recurrence and site of relapse [30,31,32,33]. Since persistence of MRD after induction/consolidation therapy has been proven to influence outcome in relapsed children [33,34,35,36], MRD measurement has been incorporated in the decision process for treatment stratification and indication to HSCT. In addition, patients with TP53 mutations, t(1;19), t(17;19), hypodiploidy and KMT2A-rearragments have a dismal prognosis regardless of the time elapsing between diagnosis and relapse and should be offered HSCT once a new CR is achieved. Overall, two-thirds of children in CR2 proceed to HSCT. Finally, all patients who have experienced two or more relapses are considered candidates to receive a HSCT, regardless of the type of donor available.

3. HSCT Indications in AML

The use of HSCT as a consolidation strategy for pediatric patients with AML in CR1 has been the subject of much debate [37,38,39]. Historically, HSCT in CR1 was reserved for those children who had an available, HLA-identical sibling donor. Early studies adopting this Mendelian/genetic randomization strategy reported a reduction in leukemia relapse-rate (RR) in patients receiving transplantation, which was counterbalanced by the occurrence of transplant-related mortality (TRM) [39,40,41]. The presence of a matched sibling donor as a HSCT indication has been replaced, in most contemporary protocols, by risk assessments based upon disease characteristics and response-related factors. Current HSCT indications in childhood AML are summarized in Table 2. Considering recent improvements in chemotherapy and the potential risk of acute and late toxicities after HSCT, the current practice restricts the use of HSCT in CR1 only to those AML patients with high-risk (HR) features. However, there is no universal agreement on the definition of HR disease and different criteria have been, and continue to be, used by different cooperative groups. There is general consensus that standard-risk patients should not be transplanted in CR1 but only after the first relapse and achievement of a second complete remission. In this regard, the underlying genetic and molecular aberrations represent a major criterion for risk group stratification and allocation to HSCT. In particular, genomic alterations involving core-binding factor (CBF) transcription factors, namely inversion16(p13;1q22), t(16;16)(p13;q22) and t(8;21)(q22;q22), are widely recognized by all study groups as favorable risk group markers. Recently, a further chromosomal aberration involving CBF, t(16;21)(q24;q22), resulting in RUNX1-CBFA2T3 fusion, has been shown to be associated with good prognosis [42]. Noteworthy, a different translocation involving chromosomes 16 and 21, t(16;21)(p11;q22) (FUS-ERG), identifies instead a rare subgroup of AML with extremely poor prognosis, which should be considered for HSCT in CR1 [42]. In patients harboring t(8;21)RUNX1-RUNX1T1, an MRD reduction of less than 2 Log with respect to diagnosis, is associated with a significant higher risk of relapse when compared to patients who reduce MRD more than 3 Log. Although these subjects should be considered candidates for more aggressive therapies, additional studies are needed to determine whether they could benefit from an allogeneic HSCT in CR1 [43]. Less frequent genetic abnormalities, such as biallelic mutations of CEBPα, mutations in nucleophosmin1 (NPM1) with a normal karyotype, are also associated with a favorable prognosis in pediatric AML [44,45]. Patients with acute promyelocytic leukemia (APL) and t(15;17), owing to the advent of ATRA and arsenic trioxide, currently represent a group with an extremely favorable prognosis [46].
In the pioneering work from the Medical Research Council (MRC)-AML group, adverse cytogenetic features were originally defined as −5, −7, del(5q), abnormal 3q or complex karyotype [47]. Additional MRC studies focused on childhood AML identified abnormalities of 12p as a new cytogenetic group associated with poor prognosis [48]. Although these alterations cumulatively account for <5% of cases of childhood AML, several groups have demonstrated that their identification correlates with high rates of induction failure and poor survival, and there is general agreement that these patients should be offered allogeneic HSCT in CR1 [3,49,50,51,52]. The t(6;9)(p22;q34), which leads to the formation of a leukemia-associated fusion protein DEK-NUP214, occurs infrequently in children (less than 1% of AML cases) and is associated with FLT3 ITD in approximately 40% of cases. AML patients harboring t(6;9) have a high risk of treatment failure, particularly those not proceeding to allogeneic HSCT [48,53,54]. FLT3 internal tandem duplication (FLT3/ITD) occurs in approximately 10% to 20% of pediatric AML cases and conveys a poor prognosis, which is favorably modified by the presence of a low allelic ratio or concomitant NPM1 mutations. By contrast, patients harboring FLT3-ITD with a high allelic ratio and without an NMP1 mutation have a very high risk of relapse and benefit from HSCT in CR1 [55,56,57,58,59]. KMT2A (MLL) rearrangements (which occur in 20% to 24% of all patients with childhood AML) have a different prognostic value depending on the specific fusion partner. Results of a large, retrospective analysis including data from 756 pediatric patients with KMT2A-rearranged AML, showed that the t(4;11)(q21;q23.3)/KMT2A-MLLT2, t(6;11)(q27;q23)/KMT2A-MLLT4, t(10;11)(p12;q23)/KMT2A-AF10 and t(10;11)(p11.2;q23)/KMT2A-ABI1 were associated with a dismal outcome [60]. Patients with such abnormalities are almost unanimously considered candidates for allo-HSCT in CR1 [48,50,61,62]. Cryptic gene fusions, including NUP98-rearrangements (the most common being t(5;11)(q35;p15)/NUP98-NSD1 and t(11;12)(p15;p13)/NUP98-KDM5A) [63,64,65], CBFA2T3-GLIS2 (resulting from a cryptic inversion of chromosome 16) [65,66,67] and t(7;12)(q36;p13)/MNX1-ETV6 (occurring virtually exclusively in children younger than 2 years) [50,68] have been shown to predict poor outcome and warrant consolidation with allogeneic HSCT in CR1 [66,68,69,70]. Finally, the t(8;16)(p11;p13) rearrangement, fusing KAT6A to the CREBBP gene, has an age-dependent impact on prognosis. While in very young children this translocation is associated with spontaneous remission encouraging a watch-and-wait strategy, in older children the prognosis is poor and consolidation with allotransplant in CR1 should be seriously considered [71].
MRD measurement by multiparametric flow cytometry (MFC-MRD) has been recently shown as a strong and independent prognostic marker of relapse in pediatric AML. In a prospective study including 232 children with AML in which MFC-MRD was adopted as risk-stratification criteria together with genetic features, high-level MRD positivity (≥1% leukemic cells) after first induction was associated with a greater cumulative incidence of relapse MRD compared to low-level (<1%) MRD (49.2% ± 7.4% vs. 16.7% ± 7.8%, p < 0.0001). While the outcome for patients with low levels of MRD after first induction was identical to that of patients with negative MRD, detectable MRD levels below 1% after second induction were associated with a poor outcome [5]. Subsequent studies performed by different cooperative groups have confirmed the strong prognostic relevance of MRD after the first and the second induction course, and MFC-MRD is being increasingly employed to refine the current strategies of disease risk stratification, including identification of candidates for HSCT in CR1 [72,73,74,75].
Regardless of the initial risk classification, in relapsed AML allogeneic HSCT offers the best chance of cure, ideally after the achievement of second CR (CR2) [2,62,76]. In subjects who relapse after a previous HSCT, a second transplant can offer remarkable long-term disease-free survival (DFS) probability, even in those cases not obtaining a further remission [77,78,79,80]. The proportion of children with refractory AML, defined as failure to achieve a morphological remission after two courses of chemotherapy, is estimated to be as high as 10% [3,81]. Allogeneic HSCT is currently considered as the only curative strategy in these subjects, being capable of producing long-term DFS in up to 50% of cases [82]. Patients below the age of 10 years and those with low leukemia burden or in CR at the time transplant have the highest chance of cure [82,83].

4. The Choice of Conditioning Regimen

Since maximal reduction of leukemia cells is of highest importance for post-HSCT outcome in childhood AL, myeloablative conditioning (MAC) is still considered the gold standard [84].
For what concerns patients affected by ALL, total body irradiation (TBI)-containing regimens are used preferentially over chemotherapy-based ones. This was based on: (i) historical data [85]; (ii) retrospective/registry studies [84,86,87,88,89]; (iii) a small randomized controlled trial [90]. Hyperfractionated TBI, combined with cyclophosphamide (CY), was firstly used by investigators at Memorial Sloan Kettering Cancer Center [85]. A number of studies analyzed the effect of different chemotherapy agents in combination with TBI. Thiotepa (TT) in combination with CY was shown to be safe and effective [91]. In a CIBMTR study, the use of high-dose etoposide in combination with TBI compared favorably with TBI and CY for children with ALL in CR2 [86]. The same conditioning regimen was employed in the ALL-SCT-BFM-2003 study with good efficacy and manageable toxicity, resulting into low incidence of TRM [92]. For this reason, this association was chosen for the prospective randomized FORUM trial (see below). Interestingly, in another retrospective study, melphalan was identified as the best single agent in association with TBI, because of lower relapse incidence as compared with other drug combinations [93]. In view of notable well-known late side effects related to the use of TBI (including growth impairment, gonadal dysfunction, cognitive dysfunction, cataracts and secondary malignancies) [94], an important question is if TBI-based regimens can be replaced by chemotherapy-based conditioning. The combination of TBI and CY resulted into improved OS and leukemia-free survival (LFS) as compared to busulfan (Bu) and CY in a retrospective study on 627 patients [87]. A small, randomized, controlled trial in 43 children with ALL showed higher event-free survival (EFS) when a TBI-based regimen was used as compared to chemotherapy alone [90]. Interestingly, in a recent retrospective registry study on more than 3000 patients conducted by the Pediatric Disease Working Party of EBMT, although TBI was confirmed to be superior to chemotherapy in the whole cohort (for OS, LFS, TRM and relapse), a subgroup analysis showed comparable outcomes of the different type of conditioning regimens for patients in CR1 [84]. In order to obtain a definitive answer on the optimal preparative regimen to be employed in children with ALL, an international, prospective, open-label, randomized, controlled trial to investigate whether chemo-conditioning regimens could replace TBI in pediatric patients with high-risk ALL (For Omitting Radiation Under Majority age (FORUM), NCT01949129) was conducted in 21 countries in patients aged 4–21 years at HSCT, in CR pre-HSCT, and with an HLA-compatible related or unrelated donor [95]. In detail, fractionated 12 Gy TBI combined with etoposide was compared to fludarabine, TT, and either Bu or treosulfan (according to the country preference chosen by national coordinators of the study). The statistical design was a noninferiority study with an 8% margin, with an estimated sample size of 1000 patients randomly assigned in 5 years with 2-year minimum follow-up. However, the trial was terminated early because of application of a stopping rule, since at interim analysis chemo-conditioning resulted significantly inferior to TBI. In particular, 2-year OS (91% versus 75%, p < 0.0001) and EFS (86% versus 58%, p < 0.0001) were significantly higher for the 212 patients randomly assigned to receive TBI as compared to 201 who were randomized to chemotherapy alone (intention-to-treat analysis). Cumulative incidence of relapse and, more surprisingly, TRM were lower in the TBI group than in the chemotherapy-based preparative regimen. The outcome of children given fludarabine, TT and either Bu or treosulfan was comparable. Notably, in subgroup analyses, TBI remained superior to chemotherapy for almost all the different variables analyzed (including disease status) [95]. Thus, this kind of conditioning is now considered the gold standard for pediatric patients affected by ALL.
In the context of acute myeloid leukemia (AML), TBI and non-TBI regimens have never been compared in a prospective, randomized fashion. The superiority of TBI-based compared to Bu-based regimens, initially reported in older studies present in literature [96,97,98] has not been confirmed in other reports. A retrospective study from the Japanese Society for Hematopoietic Cell Transplantation comparing TBI/Cy and intravenous Bu/Cy in pediatric AML patients in first or second remission (CR1/CR2) confirmed no significant differences in terms of relapse and TRM, this resulting in a similar 3-year OS (68% and 72% for TBI/Cy and Bu/Cy, respectively) [99]. Similar results have also been reported from the French Group in a retrospective study evaluating TBI/Cy versus Bu/Cy for AML patients in CR1 [100]. Recently, TBI and non-TBI regimens were compared in 624 children transplanted between 2008 and 2016 and reported to CIBMTR registry. Five-year NRM was higher with TBI regimens (22% vs. 11%, p < 0.0001), but relapse was lower (23% vs. 37%, p < 0.0001) compared to non-TBI conditioning. Consequently, OS (62% vs. 60%, p = 1.00) and LFS (55% vs. 52%, p = 0.42) did not differ between treatment groups. TBI regimens were associated with higher incidence of grade II–IV aGVHD and greater 3-year incidence of gonadal failure or growth hormone deficiency while no differences in cGVHD and late pulmonary, cardiac or renal impairment were observed [101].
The first association of Bu and high-dose cyclophosphamide (Cy) (200 mg/kg), reported from Santos et al., resulted into a low relapse rate, but unacceptable transplant related mortality (TRM): for this reason, the Cy dosage was reduced to 120 mg/kg in the subsequent studies, with a lower toxicity and the same efficacy profile [88]. The addition of a third alkylating agent (Melphalan) to a standard Bu/Cy backbone has been shown to be effective with an acceptable toxicity profile, and this conditioning regimen has been adopted by many cooperative groups [61,102]. Results from the Italian protocol AIEOP AML 2001/02, in which 243 patients given either allo- or auto-HSCT after myeloablative regimen including Bu/Cy/Mel were analyzed, showed a 8-year OS and DFS of 75% and 74%, the with cumulative incidence of TRM being of 7% for the allo-HSCT group [61]. Similar results were observed in the AML SCT-BFM 2007 trial [62]. A recent large retrospective study on behalf of the Pediatric Disease Working Party of the EBMT compared the outcomes of pediatric AML patients receiving three different conditioning regimens (TBI/Cy, BU/Cy, BU/Cy and melphalan). Among the three options, Bu/Cy/Mel was associated with a lower incidence of relapse and the higher LFS as compared with the other chemo/radiotherapy combinations [103].
In most recent years, the use of reduced intensity conditioning (RIC) has been explored in pediatric patients with AL as a strategy to maintain a good balance between high anti-leukemic effect and reduced acute/late chemotherapy toxicity. In this regard, the reduced-intensity approach could offer better outcome and minor toxicity to pediatric patients with AML who are ineligible for myeloablative therapy because of heavy pretreatment, comorbidities or genetic conditions at high-risk of transplant-related toxicities, including Fanconi anemia or Down syndrome [104,105]. Recently, in a large cohort of 180 AML patients, Bitan et al. reported no significant differences in of acute and chronic GvHD and 5-year OS and FFS between patients given or RIC and MAC regimens [106]. Treosulfan-based conditioning regimen appears an interesting alternative to busulfan [107,108]. EBMT data regarding 198 pediatric patients with hematological malignancies including 50 AML conditioned with Treosulfan-based regimens showed an optimal safety profile; the 3-year OS and EFS were 54% and 45%, respectively, with better results if Treosulfan was associated with fludarabine and an alkylating drug such as Tiothepa or Melphalan [108]. In a prospective II phase study evaluating safety and efficacy of a conditioning regimen based on Treosulfan, Fludarabine and Thiotepa in 29 pediatric patients with AML, the 3-year PFS and OS were 79% and 84%, respectively [109]. Results on other RIC or reduced toxicity regimens have been published in smaller cohorts of pediatric patients, reporting promising safety and efficacy results [110,111,112,113]. Of note, use of RIC or reduced toxicity regimen was explored in children with relapsed/refractory (R/R) AML in association with Clofarabine. Six patients with R/R AML underwent a reinduction with Clofarabine or high-dose Cytarabine and subsequently received RIC regimen and HSCT; at 21 months after the procedure, 5/6 patients were alive without relapse [114]. More recently, a large study including 103 CR1 AML pediatric patients compared safety and efficacy of Bu/Cy, Bu/Cy/Mel and Clofarabine/Flu/Bu; 5-year LFS was 43%, 59% and 67%, respectively, while 5-year TRM was 14%, 7% and 6%, respectively [115].
Anti-human T-lymphocyte globulin (ATLG) is largely used during the preparation to an allograft from donors other than an HLA-identical sibling to regulate bidirectional alloreactivity; indeed, this type of serotherapy reduces the incidence and severity of the two main immune-mediated complications of the procedure, namely GvHD and graft rejection. Several randomized trials conducted in adults have demonstrated a significant reduction in the incidence of chronic GvHD when ATLG is added to the standard GvHD prophylaxis [116]. Only one randomized controlled trial, comparing different doses of ATLG, has been conducted in children with hematological malignancies transplanted from an unrelated volunteer [117]. This study demonstrated that low-dose ATLG can reduce the incidence of life-threatening infections, without significantly affecting the incidence of acute and chronic GvHD; in addition, a lower dose of ATLG resulted into an improved OS and EFS. Thus, a low-dose ATLG (namely 15 mg/kg of Grafalon®, Neovii Biotech, Rapperswil, Switzerland; formerly ATG Fresenius®), should be regarded as the standard serotherapy regimen for HSCT from UD in pediatric patients with malignant disorders.

5. The Role of Pre-Transplant Minimal Residual Disease: Better Remission for Better HSCT Outcome?

During the last two decades, MRD quantification has emerged as a crucial assessment in the evaluation of early treatment response and in the definition of patient risk stratification in both ALL and AML [5,16,118,119,120]. At the same time, accumulating evidence has shown that pre-transplant MRD status correlates with the risk of relapse and OS after HSCT. In the late 1990s, Knechtli et al. provided the first demonstration of the key predictive role exerted by pre-transplant PCR-based MRD in a retrospective analysis of 64 pediatric patients undergoing allogeneic HSCT [121]. This observation was confirmed prospectively by the ALL-REZ BFM Study Group in 91 children with relapsed ALL. In this study, children with pre-HSCT PCR-MRD ≥10−4 had a higher cumulative incidence of relapse as compared with patients having PCR-MRD <10−4 (57% vs. 13% respectively, p < 0.001) [122]. The best cutoff value of pre-transplant PCR-MRD for prognosis prediction in ALL is still a matter of debate. In a large cohort of high-risk relapsed ALL children transplanted in CR2, subjects with PCR-MRD ≥10−3 before HSCT had a significantly worse probability of DFS as compared with patients with PCR-MRD <10−3, while no differences were observed between patients with MRD <10−4 and those having an MRD within the range of 10−4 and 10−3 [123]. By contrast, a retrospective analysis of 119 ALL patients, performed by the Italian Association for Paediatric Haematology/Oncology (AIEOP), showed that the level of pre-HSCT MRD positivity has a different impact on EFS according to disease phase at HSCT. Indeed, in patients transplanted in CR1, only an high PCR-MRD level (≥10−3) was associated with an increased risk of relapse, while in subjects transplanted in CR2, even a low-level MRD positivity (<10−3) determined a high relapse rate and poor outcome [124]. In the context of childhood AML, the prognostic value of molecular MRD before HSCT is less defined. In a recent I-BFM-AML collaborative study, the role of PCR-MRD collected within 5 weeks prior to HSCT was evaluated in 108 pediatric AML patients harboring one of the main recurrent AML gene rearrangements (t(8;21)(q22;q22); RUNX1-RUNX1T1, inv(16)(p13.1q22)/t(16;16)(p13.1;q22); CBFB-MYH11, t(9;11)(p22;q23); KMT2A-MLLT3 or FLT3-ITD). In this study, 5-year OS after HSCT was significantly higher in patients with low PCR-MRD (defined as a value below 2.1 × 10−4 calculated by ROC curve analysis with respect to diagnosis or relapse), as compared with subjects having PCR-MRD levels above the cutoff (83% vs. 57%; p = 0.012) [125].
The assessment of MRD by means of MFC (MFC-MRD) showed similar results in terms of prognostic value in both ALL and AML. In a cohort of 122 children with very high-risk ALL (n = 64) or AML (N = 58), Leung and colleagues showed that the 5-year cumulative incidence of relapse after HCT was 40% in the patients with high levels of FCM-MRD (≥0.1% in ALL and ≥1.0% in AML), 16% among those with low level of MRD (0.01% to <0.1% in ALL and 0.1% to <1% in AML) and 6% in those with no MRD (p = 0.0002). High MRD was also confirmed as an independent adverse prognostic factor for survival in multivariate analysis (p = 0.0035) [126]. In a Children’s Oncology Group (COG)/Pediatric Bone Marrow Transplant Consortium (PBMTC) multicenter phase III trial evaluating the addition of sirolimus to standard GvHD prophylaxis in children with ALL, patients with MFC-MRD ≥0.1% had a higher relapse risk as compared to subjects whose MRD was negative or <0.1% [127]. In the context of AML, detectable MFC-MRD immediately prior to HSCT has been consistently associated with increased risk of post-HSCT relapse and worse OS in both children and adults [10,128,129]. These observations are further supported by a meta-analysis of 19 studies evaluating pre-HSCT MRD (mainly assessed by FCM) in 1431 pediatric and adult AML patients; MRD positivity was associated with decreased DFS, OS and increased cumulative incidence of relapse [130]. Several groups have also shown the value of next-generation sequencing (NGS) technologies for MRD detection in both ALL and AML [131,132,133,134]. When NGS-MRD was compared with MFC-MRD in 56 pediatric B-ALL patients, NGS-MRD predicted relapse and survival more accurately than MFC-MRD (p < 0.0001), especially in the MRD negative cohort (2-year relapse probability, 0% vs. 16%; p = 0.02; 2-year OS, 96% vs. 77%; p = 0.003) [132]. Given the importance of pre-HSCT MRD in determining the probability of cure, modern ALL and AML therapy approaches have focused on strategies to induce MRD-negativity before transplant, especially for patients with high-risk disease features. These approaches are particularly attractive in B-cell ALL (B-ALL) where children may benefit from the tremendous improvements obtained over the past decade owing to the clinical application of different immunotherapy agents. Indeed, before the advent of immunotherapy, strategies to achieve pre-transplant MRD-negative remission in relapsed B-ALL relied on intensive multidrug chemotherapy regimens, which are commonly associated with the occurrence of toxicities that may be fatal or reduce the likelihood of proceeding to allogeneic HSCT [135,136].
In a phase I/II study conducted in children with R/R B-ALL, blinatumomab, a bispecific T-cell engager antibody targeting CD19, was able to induce cytomorphological remission in 39% of subjects, which was MRD-negative in 52% of cases [137]. Higher CR rates were observed in the expanded access study, likely because of the greater proportion of patients with <50% blasts enrolled, these data reflecting the association between lower leukemia burden and clinical response [138]. Recently, the results of two international randomized clinical trials, comparing blinatumomab with conventional chemotherapy as pre-transplant consolidation therapy in children with high-risk first relapse of B-ALL, were published. Randomization was performed after three cycles of chemotherapy in one study and after a single cycle of induction chemotherapy in the other one. In both studies, treatment with blinatumomab resulted in less severe toxicities, higher MRD remission rate, greater probability of proceeding to allogeneic HSCT and improved outcome [139,140].
In the dose-finding part of a phase 1/2 study promoted by the Innovative Therapies for Children With Cancer in Europe (ITCC) consortium, inotuzumab ozogamicin (InO), an antibody–drug conjugate composed of a CD22-directed monoclonal antibody linked to calicheamicin, was able induce a CR/CRi (CR with incomplete hematologic recovery) in 80% of children with relapsed and refractory B-ALL. Among the 19 responders with available MFC-MRD data, 16 (84%) obtained MRD-negativity [141]. Similar results were observed in a retrospective analysis of InO compassionate-use program, which reported a CR rate of 67% with 71% of responders achieving MRD negativity [142]. One possible toxicity concern, regarding the use of InO before HSCT, is related to the risk of sinusoidal obstruction syndrome (SOS), especially in heavily pretreated patients [141,142]. While blinatumomab and InO are mainly recognized as bridge-to-transplant (or, at least, bridge-to-consolidation) strategies in children, the benefit of a consolidative HSCT after chimeric antigen receptor T-cell (CAR T) therapy is currently the object of considerable debate [13]. Anti-CD19 CAR T-cell therapy has produced impressive MRD-negative CR rates, ranging from 56% to 93%. However, durability of response after CAR T is variable and influenced by CAR T-cell expansion, persistence and characteristics of co-stimulatory domains [11,143,144]. Studies with CD28-based anti-CD19 CAR T cells, which are characterized by short persistence after infusion, have a higher tendency of referral to HSCT compared to studies with 41BB-based CAR T cells. In the long-term follow-up study of 50 children and young adults (CAYA) treated with a CD28-based anti-CD19 CAR T, 62.0% patients achieved a CR, which was FCM-MRD negative in 90.3% of cases. Noteworthy, MRD-negative patients proceeding to allo-HSCT had a 5-year EFS of 61.9%, while all MRD-negative subjects who did not proceed to a consolidative HSCT experienced leukemia relapse [143]. In the global phase II trial of tisagenlecleucel in 75 CAYA with R/R B-ALL, only eight patients among those who obtained disease remission underwent allo-HSCT. Despite that, 59% of patients who received tisagenlecleucel remained in remission; the majority of those who relapsed experiencing CD19-negative disease [11]. A benefit from consolidative HSCT after anti-CD19 CAR T cell therapy has also been observed with a different 4-1BB-co-stimulated CAR [144],particularly in patients with rapid loss of B-cell aplasia and those who were not transplanted before CAR-T cells [145]. Consolidation with HSCT was shown to be favorably associated with better EFS (p = 0.016) also in a phase I trial evaluating a CD22-targeted/4-1BB CAR T-cell in CAYA with B-ALL [146]. Emerging data suggest that more sensitive NGS-MRD testing may help identify which patients need HSCT consolidation after CAR-T cell therapy [147]. In the near future, the dramatic increase in the number of B-ALL patients who achieve MRD-negative complete remissions owing to immunotherapy approaches, will likely offer the opportunity to re-evaluate the role of HSCT as consolidation strategy in this setting.
For patients with AML and T-cell ALL, the availability of immunotherapy is currently much more limited as compared with B-ALL. In the context of pediatric AML, modest benefit has been observed by the incorporation of CD33-targeted therapy with gemtuzumab ozogamicin (GO) in addition to standard chemotherapy [148]. Despite that, there is evidence that certain subsets of patients, particularly those with FLT3-ITD mutations, KMT2A rearrangements, single-nucleotide polymorphisms in ABCB1 and CD33, and high CD33 expression are more likely to profit from GO administration and GO may be effective at reducing MRD levels before HSCT [5,149,150,151,152,153]. Concerns regarding the risk of increased toxicity of GO in conjunction with HSCT warrant further investigation regarding optimal dosing and timing to improve overall outcomes [151]. Several immunotherapies are in various stages of preclinical and clinical development for AML, including antibody–drug conjugates, bispecific antibodies, cellular therapies and checkpoint inhibitors [154,155]. Currently, most early-phase cellular immunotherapy studies for children with AML are intended as a bridge to transplant in order to achieve more profound remission status. It is too early to speculate whether such approaches will also be able decrease the need for subsequent HSCT [154,155]. Daratumumab, a CD38-targeting monoclonal antibody, is the most promising antibody-based approach in T-ALL treatment [156,157]. Currently, daratumumab in addition to standard chemotherapy is under investigation in a phase II trial for pediatric and young adult patients with R/R T- or B-cell ALL (NCT03384654). Developing CAR-T cell therapies into the setting of T-ALL has been hampered by the risk of fratricide because of the shared expression of target antigens between CAR-T cells and T-leukemia cells, and of severe life-threatening immunodeficiency due to the elimination of normal T lymphocytes [158]. Fratricide-resistant CD7, CD5 and CD1a-targeted CAR-T [159,160,161], and universal allotolerant off-the-shelf CAR-T cells generated by genome editing [162,163] have been proposed as potential strategies to overcome these limitations.
Although accumulating evidence shows a correlation between the presence of pre-HSCT MRD and the risk or relapse in children with leukemia, merely having a detectable disease prior to HCT does not necessarily indicate an inability to cure the disease [126]. For this reason, especially in settings such as T-ALL and AML where the availability of immunotherapy approaches outside clinical trials is still limited, the benefit of repeated efforts aimed at achieving MRD-negativity before transplant should be carefully weighed against the risks of inducing additional toxicities affecting post-transplant outcome.

6. Post-Transplant Minimal Residual Disease: Is There Room for Intervention?

MRD assessment before transplantation cannot effectively identify all individuals with impending post-transplantation relapse who might benefit for pre-emptive intervention. For this reason, the predictive role of post-transplant MRD has been investigated by several groups, especially in ALL [124,164,165,166]. In a BFM study evaluating 113 pediatric patients transplanted for relapsed ALL, the level of PCR-MRD was inversely correlated with EFS and positively correlated with the cumulative incidence of relapse at all time points after transplant. In multivariable analysis, MRD ≥ 10−4 leukemic cells was consistently correlated with inferior EFS [164]. Although high levels of post-transplant MRD are strongly predictive of disease recurrence, low level MRD positivity after transplantation was not invariably associated with relapse, especially if detected early after HSCT [164]. By contrast, this and other studies showed that the further the patient is from HSCT, the more likely even low levels of MRD predict a poor prognosis [124,164,165,166]. These findings support the assumption that low levels of residual leukemia cells could be controlled by an immunologic GvL effect in the early post-transplant period, before the graft becomes tolerant toward the recipient. In a recent multicenter study, Bader et al. defined the relative risk of pre- and post-HSCT MRD in pediatric ALL, and their relationship with other independent risk factors [9]. When considered individually, however, both pre-HSCT and post-HSCT had significant prognostic value, if the two measures were simultaneously evaluated in a bivariate analysis, pre-HSCT MRD became less important in determining risk compared with the post-HSCT MRD. In this study, both MRD negative or positive patients had an approximately threefold decrease in relapse if they developed aGvHD and patients who had positive MRD recorded post-HSCT and developed aGvHD had relapse rates similar to those who were MRD negative and did not develop aGvHD [9]. The beneficial effect exerted by the occurrence of aGvHD on relapse risk and survival of children with ALL has been documented by several reports. In a COG/PBMTC study, patients with pre-HCT MFC-MRD ≥0.1% who did not develop aGvHD compared with those with MFC-MRD <0.1% who did develop aGvHD had much worse 2-year DFS (18% vs. 71%; p = 0.001). Patients with pre-HCT MRD <0.1% who did not experience aGvHD had higher rates of relapse than those who did develop aGvHD (40% vs. 13%; p = 0.008) [166]. Zecca et al. showed that children with hematologic malignancies who developed chronic GvHD (cGvHD) after transplant had a reduced relapse probability (16% ± 3% vs. 39% ± 3%, p = 0.0001) and a better DFS (68% ± 4% vs. 54% ± 3%, p = 0.01) compared with children without cGVHD, The anti-leukemic effect of cGvHD was observed mainly in patients with ALL [167]. In patients with B-ALL, post-HSCT NGS-MRD positivity performed better than FCM-MRD for predicting relapse, especially early after HCT. Any post-HSCT NGS positivity resulted in an increase in relapse risk by multivariate analysis (hazard ratio, 7.7; p = 0.05) [132]. The role of MRD after transplant in pediatric AML is less defined, although there is evidence that post-HCT positivity of MFC-MRD can predict imminent relapse [129]. Noteworthy, a recent study evaluated the predictive role of NGS-MRD in pediatric and adult AML patients undergoing HSCT. In this study, variant allele frequency (VAF) more than 0.2% on day +21 post-HSCT, was associated with decreased 3-year OS and an increased risk of relapse [133]. Finally, although less sensitive than MRD, close chimerism monitoring on peripheral blood has proven useful for the early detection of impending relapse in both ALL and AML [168,169,170].
For patients with post-transplant evidence of MRD, additional interventions could influence outcomes by favouring the development of a GvL effect. This goal has been pursued through several strategies, including rapid discontinuation (or abrupt cessation) of immune suppression [168,171,172,173], administration of cytokines [174] and infusion of donor-derived lymphocytes or cytokine-stimulated immune effector cells [170,175]. Since the benefit derived from GvL may be offset by the increased TRM associated with severe GvHD, caution should be adopted when adopting interventions that stimulate excessive GvHD [9]. In patients with Philadelphia chromosome positive ALL, or FLT3/ITD-positive AML, post-transplant administration of tyrosine kinase inhibitors, such as imatinib and sorafenib or midostaurin, respectively, can improve outcomes, especially for patients showing molecular MRD recurrence after-HSCT [176,177,178]. In the context of AML, the combination of azacytidine and donor-lymphocyte infusion (DLI) could represent an effective strategy to prevent overt disease recurrence in MRD-positive patients [179]. Recently, Ofran et al. described three patients with T-ALL in whom residual MRD after transplant was eradicated following administration of daratumumab, provided on a compassionate basis, in combination with DLI or rapid tapering of immunosuppression [157]. For patients with B-cell ALL, the availability of highly effective immunotherapy agents, such as blinatumomab and inotuzumab, also makes them attractive strategies to tackle MRD-recurrence in the post-transplant period. Ongoing clinical trials are evaluating these approaches in both children and adults (NCT04785547, NCT04044560, NCT03913559, NCT03104491).

7. Transplantation from Alternative Donors: No Longer a Second Choice?

Eligibility criteria for HSCT in both ALL and AML have been varying over time not only according to disease characteristics and response to treatment, but also depending on type of available donor. The use of HLA-matched related donors is still considered the preferred option [40,92,180,181]. However, more than 70% of children with AL who might benefit from an allograft lack an HLA-identical family donor. With the establishment of donor registries, many patients are able to locate a suitable unrelated donor (UD). Results in children transplanted for either ALL or AML suggest that fully matched UD selected through high-resolution typing of HLA loci offer minimal or possibly no disadvantages in terms of disease outcome compared with HLA-identical siblings [182,183,184,185,186,187]. In the context of ALL, it has been shown that results obtained using a 9/10 or a 10/10 allelic matched donor, either related or unrelated, are not inferior to those observed after HSCT from an HLA-identical sibling in terms of EFS, OS and CIR [92]. For several years, a wider degree of HLA mismatch has been considered acceptable only in the presence of poor prognostic features indicating an increased risk of leukemia recurrence/progression without HSCT consolidation [92,188], although recent results obtained with different alternative donor approaches strongly suggest that this paradigm could no longer be valid.
UCB transplantation (UCBT) has been largely employed in the past for patients lacking an HLA-matched donor and several reports have demonstrated that unrelated UCBT is able to offer long-term outcomes similar to those observed using an UD in both ALL and AML [189,190]. Eapen et al. compared results observed in 503 children with AL given unrelated UCBT with those of 282 bone marrow transplant (BMT) recipients. DFS was superior in recipients of HLA matched cord blood (p = 0.040), while it did not differ between BMT and one or two HLA-disparate UCBTs [190]. The results of UCBT in both childhood ALL and AML have been extensively analyzed by retrospective registry-based studies performed by the Eurocord group. In the context of ALL, a lower risk of relapse (24% vs. 39%; hazard ratio 0.4; p = 0.01) and better DFS (54% vs. 29%; hazard ratio 2, p = 0.003) were observed in children with negative MRD before UCBT as compared with those with positive MRD [191]. In children given UCBT for AML, the results were particularly promising for children with poor prognostic features, namely secondary leukaemia, high-risk karyotype, or transplanted in CR2 after an early relapse, for whom the DFS and risk of relapse were similar to those of the other patients [192]. Analysis of AML patients who underwent autologous or allogeneic HSCT within the AIEOP AML 2002/01 showed a remarkable DFS overcoming 90% in the subgroup who received UCBT [61]. Subsequent studies have demonstrated that better HLA-matching strategies between donor and recipient (at high-resolution level for HLA-A, -B, -C and -DRB1 loci) may further improve the outcome of patients with AL given a single UCBT [193,194]. Transplantation of two UCB grafts has been proposed to overcome the limitations related to the low cell dose infused; unfortunately, in two prospective randomized studies, the double UCBT strategy did not improve the overall outcome of children and young adults with AL in the presence of a single unit of adequate cell dose, being instead associated with an increased risk of GvHD [195,196]. Despite that, it has been suggested that the double-unit strategy may enhance the GvL effect and be of particular benefit in patients with positive pre-transplant MRD [197,198]. Strategies aimed at expanding the number of UCB progenitors ex vivo and favouring stem-cell-homing in vivo are being developed with encouraging preliminary results, holding the potential to revitalize the field of UCBT in the near future [199,200,201].
One of the main reasons for the continue decline in the utilization of UCBT observed in the last decade, beyond the intrinsic limitations of the approach (i.e., delayed engraftment, increased risk of TRM), is the emergence of highly effective T-cell depleted and T-cell repleted HLA-haploidentical transplantation platforms [202]. Indeed, HSCT from an HLA-haploidentical relative offers an immediate transplant treatment virtually to any patient lacking a matched donor or a suitable UCB unit and, in addition, easy access to the donor for post-transplant adoptive cell therapies [203,204].
In the setting of T-cell depleted (TCD) haploidentical (haplo) HSCT, a remarkable advance has been obtained through the development of more sophisticated graft manipulation strategies (with respect to the original positive selection of CD34+ cells), based on the selective depletion of CD3+ and CD19+ cells [187,205,206] or, more recently, of TCRα/β+ and CD19+ cells [207]. With the latter approach, in particular, the graft transfers to the recipient not only high numbers of CD34+ cells, but also of mature donor NK cells and TCRγ/δ+ T cells that can display their protective effect against leukemia regrowth and life-threatening infections [208]. By contrast, T cells expressing the αβ chains of the T cell receptor, which are responsible for the development of GvHD, are removed with a median depletion efficiency of 4 Log [209]. Our group has reported the outcome of a cohort of 80 children with AL (ALL, n = 56; AML, n = 24) transplanted from an haploidentical relative after αβ+ T-cell and CD19+ B-cell depletion [210]. All children received a fully myeloablative preparative regimen and received anti-T-lymphocyte globulin for preventing graft rejection and GvHD; no patient received any post-transplant GVHD prophylaxis. The cumulative incidence of TRM was remarkably low (5%). Nineteen subjects relapsed, the cumulative incidence of recurrence being 24%. The 5-year OS probability was 72% (95% CI, 62–82) for the whole study population. Overall, the 5-year DFS was 71% (95% CI, 61–81), without differences between ALL and AML patients. The cumulative incidence of skin-only, grade I–II acute GvHD (aGvHD) was 30% and no patient developed GvHD with visceral involvement, grade III–IV aGvHD or extensive chronic GvHD. TBI-containing preparative regimen was the only variable favourably influencing relapse incidence and chronic GVHD-free/relapse-free survival (GRFS) [210]. More recently, the results of 98 αβ-TCD haplo-HSCT recipients were compared with those of 127 matched UD (MUD), 118 mismatched UD (MMUD) in a multicenter retrospective analysis. MUD and MMUD-HSCT were characterized by a higher cumulative incidence of grade II–IV and grade III–IV aGvHD (35% vs. 44% and 6% vs. 18%, respectively) compared with 16% and 0% in αβ-TCD haplo-HSCT (p < 0.001). Children treated with αβ-TCD haplo-HSCT also had a significantly lower incidence of overall and extensive chronic GVHD (p < 0.01). While the GRFS probability of survival of MUD-HSCT and αβhaplo-HSCT recipients was superimposable (61% and 58%, respectively), the choice of MMUD-HSCT had a detrimental effect on this composite end-point (34%; p < 0.001) [6]. Noteworthy, although the haploidentical donor was mainly selected privileging NK immunological features [211,212], no favourable influence of NK alloreactivity and of donor KIR B-haplotype was observed in both studies [6,210]. This is in contrast with the observations derived from studies on the infusion of CD34+ cells [213,214,215,216], likely because the NK-mediated GVL effect was partially obscured by other cells present in the graft, including γδ T cells [217]. In a retrospective Spanish study performed in children with ALL given ex vivo TCD grafts using different manipulation strategies, the presence of donor–recipient KIR mismatch provided no advantage, while a donor with KIR A haplotype was associated with an increased probability of relapse [218]. Although the benefits of an NK alloreactive asset appear less clear when using refined graft-manipulation strategies, the selection of a donor with favourable NK features continues to be recommended for patients receiving TCD-HSCT [212]. Since preclinical data showed that bisphosphonates can enhance TcRγδ-mediated anti-leukemia activity [219], the administration of zoledronic acid (ZOL) after αβ-TCD haplo-HSCT has been explored as potential strategy for further improving patient outcome, showing encouraging results in terms of better OS, reduced GvHD incidence and lower TRM [220,221]. In addition, TCD haplo-HSCT approaches represent an ideal platform for post-transplant adoptive cell therapies because no (or minimal) immunosuppression is given after HSCT. An intriguing approach to accelerate the recovery of adaptive immunity and to promote anti-tumour activity relies on the administration of suicide gene-modified T cells, which offer the possibility of limiting the risk of uncontrolled GvHD by triggering T-cell apoptosis [222]. In the adult TCD haplo-HSCT setting, post-transplantation infusion of donor T-cells modified with the insertion of the herpes simplex thymidine kinase suicide gene (to achieve in vivo susceptibility to ganciclovir) enabled regulation of GvHD, while promoting immune reconstitution [223,224]. An alternative strategy, developed by the Baylor group, is based on T cells engineered to express the caspase 9 (iC9) suicide gene [225]. Post-transplant infusion of iC9-T cells has been shown to accelerate immune recovery [226,227]. Administration of iC9-T cells after αβ+ TCD-haplo-HSCT was associated with better OS and DFS when compared with an historical cohort of “pure” αβ+ TCD-haplo-HSCT [228]. In order to promote the recovery of pathogen-specific immunity, other groups have investigated the infusion of low-dose memory CD45RA-depleted donor lymphocytes after HSCT with αβ T-cell depletion, with encouraging results [229]. The rationale for this strategy is based on experimental data demonstrating that mouse CD4 memory T-cells, and effector memory CD8 T-cells, are devoid of GvH reactivity [230]. Removal of CD45RA+ naıve T lymphocytes has been also tested as TCD strategy before haploidentical HSCT. In a recent publication describing the preliminary outcomes of 50 children receiving a CD45RA-depleted haplo-HSCT (of whom 47 having AL), the St. Jude investigators reported moderately high rates of grade III–IV aGvHD (28%) and chronic GvHD (26%). Despite that, GvHD was successfully treated in most patients, and NRM mortality among patients in CR at the time of HCST was 5.6%, the 3-year OS and EFS being 78.9% and 77.7%, respectively [231].
T-cell repleted haplo-HSCT approaches based on the post-transplant administration of cyclophosphamide (PTCy), pioneered by the Johns Hopkins group [232], have been extensively investigated in adult patients [233,234]. In the last few years, accumulating evidence suggests that this strategy may also be successfully employed in children. A recent retrospective analysis compared the outcome of pediatric patients with AL undergoing T-replete haplo-HSCT with PTCy after nonmyeloablative (NMA) conditioning with that of a contemporary cohort of children receiving transplantation from MUD and MMUD. The 5-year OS rates were not different between MUD, MMUD and Haplo patients, while an increased incidence of graft failure was observed in the haplo-group, likely derived from the adoption of an NMA preparative regimen [235]. A full-myeloablative regimen, either busulfan-based or TBI-based, has been employed with success in 29 children and young adults undergoing HLA-haplo-HSCT with PTCy for malignant conditions (of whom 18 with AL), with 3-year OS and EFS of 79% (95% CI, 66, 96) and 69% (95% CI, 54, 88), respectively. In this small cohort, relative high rates of cGvHD were observed (cumulative incidence of cGvHD at 1-year 28% (95% CI, 0.1, 0.4), moderate to severe 14% (95% CI, 0.01, 0.3)), but the limited sample size prevents drawing definitive conclusions [236]. The results of a retrospective analysis of 180 children with ALL who received a HLA-haplo-HSCT with PTCy, were recently reported by Pediatric Disease Working Party (PDWP) of the European Society for Blood and Marrow Transplantation (EBMT). Cumulative incidence of day-100 grade II–IV aGvHD was 28%, and 2-year cGvHD was 21.9%. The 2-year NRM was 19.6% and the estimated 2-year DFS was 65%, 44% and 18.8% for patients transplanted in CR1, CR2 and CR3, respectively. In multivariable analysis for patients in CR1 and CR2, disease status (CR2 (hazard ratio 2.19; p = 0.04)), age at HCST greater than 13 years (hazard ratio 2.07; p = 0.03) and use of peripheral blood stem cell (PBSC) (hazard ratio 1.98; p = 0.04) were independently associated with decreased OS [237]. Recent evidence also suggests that refinements in pharmacological GvHD prophylaxis strategies may improve the outcome of patients receiving unmanipulated HLA-mismatched grafts from UD. In particular, in a prospective multicenter phase II trial, the addition of abatacept, an anti-CTLA-4 monoclonal antibody, to standard calcineurin inhibitor (CNI)/methotrexate (MTX)-based GvHD prophylaxis was able to reduce the incidence of grade III–IV aGvHD in children and adults with haematological malignancies undergoing HSCT from an 8/8 MUD or 7/8 MMUD. Patients receiving abatacept had better severe aGvHD-free survival (SGFS) as compared with controls receiving only CNI/MTX (93.2% vs. 82, p = 0.05 in the 8/8 arm; 97.7% vs. 58.7%, p < 0.001, in the 7/8 arm) [238]. The impressive results observed in the 7/8 arm are of particular interest for those patients (often belonging to ethnic groups poorly represented in the registries) for whom the possibility of UD-HSCT is mainly restricted to an HLA-mismatched setting [239].
Collectively, most recent transplant results obtained with MUD, MMUD, haploidentical donors and UCB units indicate that all these options are able to offer the chance of transplant to virtually every child with AL in need of an allograft and lacking an HLA-identical sibling, without significant differences in terms of outcome. Each of these strategies has advantages and limitations, but rather than being considered competing alternatives, they should be regarded as complementary options and the final choice should be based on patient’s characteristics, clinician/Centre expertise, the urgency of the transplant and specific features of the unrelated/haploidentical donor or UCB unit.

8. Conclusions

Results of HSCT in AL have improved substantially, in particular during the last two decades, and progress using alternative donors has extended the potential application of allotransplantation to most patients. For several years, donor characteristics have played a relevant role in determining eligibility for HSCT, with alternative donors being accepted only in those cases deemed at the highest risk of leukemia relapse. However, current results with alternative donors suggest that this paradigm could no longer be valid and encourage not restricting the indications for HSCT upon availability of a fully matched donor. In this regard, the importance of the specific transplant infrastructure and experience of the team in determining the outcome of HSCT from alternative donors must not be neglected [240]. The importance of MRD before and after transplant is being increasingly clarified and MRD-directed interventions may further improve the outcome of allogeneic HSCT in AL, particularly in view of the growing availability of precision medicine approaches. In addition, incorporation of MRD data in risk stratification models before allo-HSCT may allow better identifying which patients should be offered an allograft and possibly inform how to transplant these subjects [241]. In the last few years, successes obtained by combination chemotherapy and the development of highly effective immunotherapy agents have raised the question of whether HSCT will continue to play a role in modern therapy of childhood AL, particularly in B-ALL [13]. Although the answer to this question should necessarily come from the results of well-designed and randomized clinical trials, the number of variables that may influence transplant outcome and the quantity of innovative approaches that have entered, or are about to enter, into the clinical arena, makes the design and conduction of such studies anything but trivial. For these reasons, determining the appropriate role of HSCT in childhood AL will continue to be a challenging and dynamic process, requiring constant and careful assessment of the likelihood of cure with conventional chemotherapy or novel targeted therapies to identify the subset of children for whom transplant offers a better treatment option. In this regard, close cooperation between chemotherapy cooperative study groups and transplant/cell therapy societies, and monitoring of treatment-related late effects with long-term patient follow-up is crucial.

Author Contributions

Conceptualization, M.A., P.M., F.L. and D.P.; writing—original draft preparation, M.A., P.M. and D.P.; writing—final review, F.L. All authors have read and approved the published version of the manuscript.

Funding

This paper was supported by grants from Accelerator Award – Cancer Research UK/AIRC – INCAR project (F.L.), Associazione Italiana Ricerca per la Ricerca sul Cancro (AIRC)-Special Project 5 × 1000 no. 9962 (F.L.), AIRC IG 2018 id. 21724 (F.L.), Ministero dell’Università e della Ricerca (Grant PRIN 2017 to F.L.); Italian Healthy Ministry project on CAR T RCR-2019-23669115 (Coordinator F.L.)

Institutional Review Board Statement

Not Applicable.

Informed Consent Statement

Not Applicable.

Data Availability Statement

Not Applicable.

Conflicts of Interest

F.L. reports advisory board membership for Amgen, Novartis, Bellicum Pharmaceutical, Neovii, and is on the Speakers’ Bureau for Amgen, Novartis, Miltenyi, Medac, Jazz Pharmaceutical, Gilead, Sanofi and Takeda, outside the submitted work.

References

  1. Hunger, S.P.; Lu, X.; Devidas, M.; Camitta, B.M.; Gaynon, P.S.; Winick, N.J.; Reaman, G.H.; Carroll, W.L. Improved survival for children and adolescents with acute lymphoblastic leukemia between 1990 and 2005: A report from the children’s oncology group. J. Clin. Oncol. 2012, 30, 1663–1669. [Google Scholar] [CrossRef] [PubMed]
  2. Bunin, N.J.; Davies, S.M.; Aplenc, R.; Camitta, B.M.; DeSantes, K.B.; Goyal, R.K.; Kapoor, N.; Kernan, N.A.; Rosenthal, J.; Smith, F.O.; et al. Unrelated donor bone marrow transplantation for children with acute myeloid leukemia beyond first remission or refractory to chemotherapy. J. Clin. Oncol. 2008, 26, 4326–4332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Pession, A.; Masetti, R.; Rizzari, C.; Putti, M.C.; Casale, F.; Fagioli, F.; Luciani, M.; Lo Nigro, L.; Menna, G.; Micalizzi, C.; et al. Results of the AIEOP AML 2002/01 multicenter prospective trial for the treatment of children with acute myeloid leukemia. Blood 2013, 122, 170–178. [Google Scholar] [CrossRef] [Green Version]
  4. Rasche, M.; Zimmermann, M.; Borschel, L.; Bourquin, J.P.; Dworzak, M.; Klingebiel, T.; Lehrnbecher, T.; Creutzig, U.; Klusmann, J.H.; Reinhardt, D. Successes and challenges in the treatment of pediatric acute myeloid leukemia: A retrospective analysis of the AML-BFM trials from 1987 to 2012. Leukemia 2018, 32, 2167–2177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Rubnitz, J.E.; Inaba, H.; Dahl, G.; Ribeiro, R.C.; Bowman, W.P.; Taub, J.; Pounds, S.; Razzouk, B.I.; Lacayo, N.J.; Cao, X.; et al. Minimal residual disease-directed therapy for childhood acute myeloid leukaemia: Results of the AML02 multicentre trial. Lancet Oncol. 2010, 11, 543–552. [Google Scholar] [CrossRef] [Green Version]
  6. Bertaina, A.; Zecca, M.; Buldini, B.; Sacchi, N.; Algeri, M.; Saglio, F.; Perotti, C.; Gallina, A.M.; Bertaina, V.; Lanino, E.; et al. Unrelated donor vs HLA-haploidentical alpha/beta T-cell- and B-cell-depleted HSCT in children with acute leukemia. Blood 2018, 132, 2594–2607. [Google Scholar] [CrossRef] [Green Version]
  7. Iacobucci, I.; Mullighan, C.G. Genetic Basis of Acute Lymphoblastic Leukemia. J. Clin. Oncol. 2017, 35, 975–983. [Google Scholar] [CrossRef]
  8. Iacobucci, I.; Wen, J.; Meggendorfer, M.; Choi, J.K.; Shi, L.; Pounds, S.B.; Carmichael, C.L.; Masih, K.E.; Morris, S.M.; Lindsley, R.C.; et al. Genomic subtyping and therapeutic targeting of acute erythroleukemia. Nat. Genet. 2019, 51, 694–704. [Google Scholar] [CrossRef]
  9. Bader, P.; Salzmann-Manrique, E.; Balduzzi, A.; Dalle, J.H.; Woolfrey, A.E.; Bar, M.; Verneris, M.R.; Borowitz, M.J.; Shah, N.N.; Gossai, N.; et al. More precisely defining risk peri-HCT in pediatric ALL: Pre- vs. post-MRD measures, serial positivity, and risk modeling. Blood Adv. 2019, 3, 3393–3405. [Google Scholar] [CrossRef] [Green Version]
  10. Walter, R.B.; Gooley, T.A.; Wood, B.L.; Milano, F.; Fang, M.; Sorror, M.L.; Estey, E.H.; Salter, A.I.; Lansverk, E.; Chien, J.W.; et al. Impact of pretransplantation minimal residual disease, as detected by multiparametric flow cytometry, on outcome of myeloablative hematopoietic cell transplantation for acute myeloid leukemia. J. Clin. Oncol. 2011, 29, 1190–1197. [Google Scholar] [CrossRef] [Green Version]
  11. Maude, S.L.; Laetsch, T.W.; Buechner, J.; Rives, S.; Boyer, M.; Bittencourt, H.; Bader, P.; Verneris, M.R.; Stefanski, H.E.; Myers, G.D.; et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 439–448. [Google Scholar] [CrossRef]
  12. Locatelli, F.; Schrappe, M.; Bernardo, M.E.; Rutella, S. How I treat relapsed childhood acute lymphoblastic leukemia. Blood 2012, 120, 2807–2816. [Google Scholar] [CrossRef] [Green Version]
  13. Diorio, C.; Maude, S.L. CAR T cells vs. allogeneic HSCT for poor-risk ALL. Hematol. Am. Soc. Hematol. Educ. Program. 2020, 2020, 501–507. [Google Scholar] [CrossRef]
  14. Borowitz, M.J.; Devidas, M.; Hunger, S.P.; Bowman, W.P.; Carroll, A.J.; Carroll, W.L.; Linda, S.; Martin, P.L.; Pullen, D.J.; Viswanatha, D.; et al. Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia and its relationship to other prognostic factors: A Children’s Oncology Group study. Blood 2008, 111, 5477–5485. [Google Scholar] [CrossRef] [Green Version]
  15. van Dongen, J.J.; van der Velden, V.H.; Bruggemann, M.; Orfao, A. Minimal residual disease diagnostics in acute lymphoblastic leukemia: Need for sensitive, fast, and standardized technologies. Blood 2015, 125, 3996–4009. [Google Scholar] [CrossRef] [Green Version]
  16. Conter, V.; Bartram, C.R.; Valsecchi, M.G.; Schrauder, A.; Panzer-Grumayer, R.; Moricke, A.; Arico, M.; Zimmermann, M.; Mann, G.; De Rossi, G.; et al. Molecular response to treatment redefines all prognostic factors in children and adolescents with B-cell precursor acute lymphoblastic leukemia: Results in 3184 patients of the AIEOP-BFM ALL 2000 study. Blood 2010, 115, 3206–3214. [Google Scholar] [CrossRef] [PubMed]
  17. Schrappe, M.; Hunger, S.P.; Pui, C.H.; Saha, V.; Gaynon, P.S.; Baruchel, A.; Conter, V.; Otten, J.; Ohara, A.; Versluys, A.B.; et al. Outcomes after induction failure in childhood acute lymphoblastic leukemia. N. Engl. J. Med. 2012, 366, 1371–1381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Nachman, J.B.; Heerema, N.A.; Sather, H.; Camitta, B.; Forestier, E.; Harrison, C.J.; Dastugue, N.; Schrappe, M.; Pui, C.H.; Basso, G.; et al. Outcome of treatment in children with hypodiploid acute lymphoblastic leukemia. Blood 2007, 110, 1112–1115. [Google Scholar] [CrossRef] [Green Version]
  19. McNeer, J.L.; Devidas, M.; Dai, Y.; Carroll, A.J.; Heerema, N.A.; Gastier-Foster, J.M.; Kahwash, S.B.; Borowitz, M.J.; Wood, B.L.; Larsen, E.; et al. Hematopoietic Stem-Cell Transplantation Does Not Improve the Poor Outcome of Children with Hypodiploid Acute Lymphoblastic Leukemia: A Report From Children’s Oncology Group. J. Clin. Oncol. 2019, 37, 780–789. [Google Scholar] [CrossRef]
  20. Pui, C.H.; Rebora, P.; Schrappe, M.; Attarbaschi, A.; Baruchel, A.; Basso, G.; Cave, H.; Elitzur, S.; Koh, K.; Liu, H.C.; et al. Outcome of Children with Hypodiploid Acute Lymphoblastic Leukemia: A Retrospective Multinational Study. J. Clin. Oncol. 2019, 37, 770–779. [Google Scholar] [CrossRef] [PubMed]
  21. Fischer, U.; Forster, M.; Rinaldi, A.; Risch, T.; Sungalee, S.; Warnatz, H.J.; Bornhauser, B.; Gombert, M.; Kratsch, C.; Stutz, A.M.; et al. Genomics and drug profiling of fatal TCF3-HLF-positive acute lymphoblastic leukemia identifies recurrent mutation patterns and therapeutic options. Nat. Genet. 2015, 47, 1020–1029. [Google Scholar] [CrossRef] [PubMed]
  22. Mouttet, B.; Vinti, L.; Ancliff, P.; Bodmer, N.; Brethon, B.; Cario, G.; Chen-Santel, C.; Elitzur, S.; Hazar, V.; Kunz, J.; et al. Durable remissions in TCF3-HLF positive acute lymphoblastic leukemia with blinatumomab and stem cell transplantation. Haematologica 2019, 104, e244–e247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Mullighan, C.G.; Su, X.; Zhang, J.; Radtke, I.; Phillips, L.A.; Miller, C.B.; Ma, J.; Liu, W.; Cheng, C.; Schulman, B.A.; et al. Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. N. Engl. J. Med. 2009, 360, 470–480. [Google Scholar] [CrossRef] [PubMed]
  24. Stanulla, M.; Dagdan, E.; Zaliova, M.; Moricke, A.; Palmi, C.; Cazzaniga, G.; Eckert, C.; Te Kronnie, G.; Bourquin, J.P.; Bornhauser, B.; et al. IKZF1(plus) Defines a New Minimal Residual Disease-Dependent Very-Poor Prognostic Profile in Pediatric B-Cell Precursor Acute Lymphoblastic Leukemia. J. Clin. Oncol. 2018, 36, 1240–1249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Biondi, A.; Cimino, G.; Pieters, R.; Pui, C.H. Biological and therapeutic aspects of infant leukemia. Blood 2000, 96, 24–33. [Google Scholar] [CrossRef]
  26. Pieters, R.; Schrappe, M.; De Lorenzo, P.; Hann, I.; De Rossi, G.; Felice, M.; Hovi, L.; LeBlanc, T.; Szczepanski, T.; Ferster, A.; et al. A treatment protocol for infants younger than 1 year with acute lymphoblastic leukaemia (Interfant-99): An observational study and a multicentre randomised trial. Lancet 2007, 370, 240–250. [Google Scholar] [CrossRef]
  27. Mann, G.; Attarbaschi, A.; Schrappe, M.; De Lorenzo, P.; Peters, C.; Hann, I.; De Rossi, G.; Felice, M.; Lausen, B.; Leblanc, T.; et al. Improved outcome with hematopoietic stem cell transplantation in a poor prognostic subgroup of infants with mixed-lineage-leukemia (MLL)-rearranged acute lymphoblastic leukemia: Results from the Interfant-99 Study. Blood 2010, 116, 2644–2650. [Google Scholar] [CrossRef]
  28. Heerema, N.A.; Carroll, A.J.; Devidas, M.; Loh, M.L.; Borowitz, M.J.; Gastier-Foster, J.M.; Larsen, E.C.; Mattano, L.A., Jr.; Maloney, K.W.; Willman, C.L.; et al. Intrachromosomal amplification of chromosome 21 is associated with inferior outcomes in children with acute lymphoblastic leukemia treated in contemporary standard-risk children’s oncology group studies: A report from the children’s oncology group. J. Clin. Oncol. 2013, 31, 3397–3402. [Google Scholar] [CrossRef]
  29. Moorman, A.V.; Robinson, H.; Schwab, C.; Richards, S.M.; Hancock, J.; Mitchell, C.D.; Goulden, N.; Vora, A.; Harrison, C.J. Risk-directed treatment intensification significantly reduces the risk of relapse among children and adolescents with acute lymphoblastic leukemia and intrachromosomal amplification of chromosome 21: A comparison of the MRC ALL97/99 and UKALL2003 trials. J. Clin. Oncol. 2013, 31, 3389–3396. [Google Scholar] [CrossRef]
  30. Bailey, L.C.; Lange, B.J.; Rheingold, S.R.; Bunin, N.J. Bone-marrow relapse in paediatric acute lymphoblastic leukaemia. Lancet Oncol. 2008, 9, 873–883. [Google Scholar] [CrossRef]
  31. Einsiedel, H.G.; von Stackelberg, A.; Hartmann, R.; Fengler, R.; Schrappe, M.; Janka-Schaub, G.; Mann, G.; Hahlen, K.; Gobel, U.; Klingebiel, T.; et al. Long-term outcome in children with relapsed ALL by risk-stratified salvage therapy: Results of trial acute lymphoblastic leukemia-relapse study of the Berlin-Frankfurt-Munster Group 87. J. Clin. Oncol. 2005, 23, 7942–7950. [Google Scholar] [CrossRef]
  32. Lawson, S.E.; Harrison, G.; Richards, S.; Oakhill, A.; Stevens, R.; Eden, O.B.; Darbyshire, P.J. The UK experience in treating relapsed childhood acute lymphoblastic leukaemia: A report on the medical research council UKALLR1 study. Br. J. Haematol. 2000, 108, 531–543. [Google Scholar] [CrossRef]
  33. Borgmann, A.; von Stackelberg, A.; Hartmann, R.; Ebell, W.; Klingebiel, T.; Peters, C.; Henze, G.; Berlin-Frankfurt-Munster Relapse Study, G. Unrelated donor stem cell transplantation compared with chemotherapy for children with acute lymphoblastic leukemia in a second remission: A matched-pair analysis. Blood 2003, 101, 3835–3839. [Google Scholar] [CrossRef]
  34. Parker, C.; Waters, R.; Leighton, C.; Hancock, J.; Sutton, R.; Moorman, A.V.; Ancliff, P.; Morgan, M.; Masurekar, A.; Goulden, N.; et al. Effect of mitoxantrone on outcome of children with first relapse of acute lymphoblastic leukaemia (ALL R3): An open-label randomised trial. Lancet 2010, 376, 2009–2017. [Google Scholar] [CrossRef] [Green Version]
  35. Eckert, C.; Biondi, A.; Seeger, K.; Cazzaniga, G.; Hartmann, R.; Beyermann, B.; Pogodda, M.; Proba, J.; Henze, G. Prognostic value of minimal residual disease in relapsed childhood acute lymphoblastic leukaemia. Lancet 2001, 358, 1239–1241. [Google Scholar] [CrossRef]
  36. Coustan-Smith, E.; Gajjar, A.; Hijiya, N.; Razzouk, B.I.; Ribeiro, R.C.; Rivera, G.K.; Rubnitz, J.E.; Sandlund, J.T.; Andreansky, M.; Hancock, M.L.; et al. Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia after first relapse. Leukemia 2004, 18, 499–504. [Google Scholar] [CrossRef] [Green Version]
  37. Creutzig, U.; Reinhardt, D. Current controversies: Which patients with acute myeloid leukaemia should receive a bone marrow transplantation? A European view. Br. J. Haematol. 2002, 118, 365–377. [Google Scholar] [CrossRef] [PubMed]
  38. Chen, A.R.; Alonzo, T.A.; Woods, W.G.; Arceci, R.J. Current controversies: Which patients with acute myeloid leukaemia should receive a bone marrow transplantation? An American view. Br. J. Haematol. 2002, 118, 378–384. [Google Scholar] [CrossRef]
  39. Hasle, H. A critical review of which children with acute myeloid leukaemia need stem cell procedures. Br. J. Haematol. 2014, 166, 23–33. [Google Scholar] [CrossRef]
  40. Woods, W.G.; Neudorf, S.; Gold, S.; Sanders, J.; Buckley, J.D.; Barnard, D.R.; Dusenbery, K.; DeSwarte, J.; Arthur, D.C.; Lange, B.J.; et al. A comparison of allogeneic bone marrow transplantation, autologous bone marrow transplantation, and aggressive chemotherapy in children with acute myeloid leukemia in remission. Blood 2001, 97, 56–62. [Google Scholar] [CrossRef] [Green Version]
  41. Lie, S.O.; Abrahamsson, J.; Clausen, N.; Forestier, E.; Hasle, H.; Hovi, L.; Jonmundsson, G.; Mellander, L.; Gustafsson, G. Treatment stratification based on initial in vivo response in acute myeloid leukaemia in children without Down’s syndrome: Results of NOPHO-AML trials. Br. J. Haematol. 2003, 122, 217–225. [Google Scholar] [CrossRef]
  42. Noort, S.; Zimmermann, M.; Reinhardt, D.; Cuccuini, W.; Pigazzi, M.; Smith, J.; Ries, R.E.; Alonzo, T.A.; Hirsch, B.; Tomizawa, D.; et al. Prognostic impact of t(16;21)(p11;q22) and t(16;21)(q24;q22) in pediatric AML: A retrospective study by the I-BFM Study Group. Blood 2018, 132, 1584–1592. [Google Scholar] [CrossRef] [Green Version]
  43. Pigazzi, M.; Manara, E.; Buldini, B.; Beqiri, V.; Bisio, V.; Tregnago, C.; Rondelli, R.; Masetti, R.; Putti, M.C.; Fagioli, F.; et al. Minimal residual disease monitored after induction therapy by RQ-PCR can contribute to tailor treatment of patients with t(8;21) RUNX1-RUNX1T1 rearrangement. Haematologica 2015, 100, e99–e101. [Google Scholar] [CrossRef]
  44. Ho, P.A.; Alonzo, T.A.; Gerbing, R.B.; Pollard, J.; Stirewalt, D.L.; Hurwitz, C.; Heerema, N.A.; Hirsch, B.; Raimondi, S.C.; Lange, B.; et al. Prevalence and prognostic implications of CEBPA mutations in pediatric acute myeloid leukemia (AML): A report from the Children’s Oncology Group. Blood 2009, 113, 6558–6566. [Google Scholar] [CrossRef] [Green Version]
  45. Hollink, I.H.; Zwaan, C.M.; Zimmermann, M.; Arentsen-Peters, T.C.; Pieters, R.; Cloos, J.; Kaspers, G.J.; de Graaf, S.S.; Harbott, J.; Creutzig, U.; et al. Favorable prognostic impact of NPM1 gene mutations in childhood acute myeloid leukemia, with emphasis on cytogenetically normal AML. Leukemia 2009, 23, 262–270. [Google Scholar] [CrossRef]
  46. Testi, A.M.; Pession, A.; Diverio, D.; Grimwade, D.; Gibson, B.; de Azevedo, A.C.; Moran, L.; Leverger, G.; Elitzur, S.; Hasle, H.; et al. Risk-adapted treatment of acute promyelocytic leukemia: Results from the International Consortium for Childhood APL. Blood 2018, 132, 405–412. [Google Scholar] [CrossRef] [PubMed]
  47. Grimwade, D.; Walker, H.; Oliver, F.; Wheatley, K.; Harrison, C.; Harrison, G.; Rees, J.; Hann, I.; Stevens, R.; Burnett, A.; et al. The importance of diagnostic cytogenetics on outcome in AML: Analysis of 1612 patients entered into the MRC AML 10 trial. The Medical Research Council Adult and Children’s Leukaemia Working Parties. Blood 1998, 92, 2322–2333. [Google Scholar] [CrossRef] [Green Version]
  48. Harrison, C.J.; Hills, R.K.; Moorman, A.V.; Grimwade, D.J.; Hann, I.; Webb, D.K.; Wheatley, K.; de Graaf, S.S.; van den Berg, E.; Burnett, A.K.; et al. Cytogenetics of childhood acute myeloid leukemia: United Kingdom Medical Research Council Treatment trials AML 10 and 12. J. Clin. Oncol. 2010, 28, 2674–2681. [Google Scholar] [CrossRef] [PubMed]
  49. Johnston, D.L.; Alonzo, T.A.; Gerbing, R.B.; Hirsch, B.; Heerema, N.A.; Ravindranath, Y.; Woods, W.G.; Lange, B.J.; Gamis, A.S.; Raimondi, S.C. Outcome of pediatric patients with acute myeloid leukemia (AML) and -5/5q- abnormalities from five pediatric AML treatment protocols: A report from the Children’s Oncology Group. Pediatr. Blood Cancer 2013, 60, 2073–2078. [Google Scholar] [CrossRef]
  50. von Neuhoff, C.; Reinhardt, D.; Sander, A.; Zimmermann, M.; Bradtke, J.; Betts, D.R.; Zemanova, Z.; Stary, J.; Bourquin, J.P.; Haas, O.A.; et al. Prognostic impact of specific chromosomal aberrations in a large group of pediatric patients with acute myeloid leukemia treated uniformly according to trial AML-BFM 98. J. Clin. Oncol. 2010, 28, 2682–2689. [Google Scholar] [CrossRef]
  51. Hasle, H.; Alonzo, T.A.; Auvrignon, A.; Behar, C.; Chang, M.; Creutzig, U.; Fischer, A.; Forestier, E.; Fynn, A.; Haas, O.A.; et al. Monosomy 7 and deletion 7q in children and adolescents with acute myeloid leukemia: An international retrospective study. Blood 2007, 109, 4641–4647. [Google Scholar] [CrossRef] [Green Version]
  52. Rasche, M.; von Neuhoff, C.; Dworzak, M.; Bourquin, J.P.; Bradtke, J.; Gohring, G.; Escherich, G.; Fleischhack, G.; Graf, N.; Gruhn, B.; et al. Genotype-outcome correlations in pediatric AML: The impact of a monosomal karyotype in trial AML-BFM 2004. Leukemia 2017, 31, 2807–2814. [Google Scholar] [CrossRef] [Green Version]
  53. Sandahl, J.D.; Coenen, E.A.; Forestier, E.; Harbott, J.; Johansson, B.; Kerndrup, G.; Adachi, S.; Auvrignon, A.; Beverloo, H.B.; Cayuela, J.M.; et al. t(6;9)(p22;q34)/DEK-NUP214-rearranged pediatric myeloid leukemia: An international study of 62 patients. Haematologica 2014, 99, 865–872. [Google Scholar] [CrossRef] [PubMed]
  54. Tarlock, K.; Alonzo, T.A.; Moraleda, P.P.; Gerbing, R.B.; Raimondi, S.C.; Hirsch, B.A.; Ravindranath, Y.; Lange, B.; Woods, W.G.; Gamis, A.S.; et al. Acute myeloid leukaemia (AML) with t(6;9)(p23;q34) is associated with poor outcome in childhood AML regardless of FLT3-ITD status: A report from the Children’s Oncology Group. Br. J. Haematol. 2014, 166, 254–259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Meshinchi, S.; Alonzo, T.A.; Stirewalt, D.L.; Zwaan, M.; Zimmerman, M.; Reinhardt, D.; Kaspers, G.J.; Heerema, N.A.; Gerbing, R.; Lange, B.J.; et al. Clinical implications of FLT3 mutations in pediatric AML. Blood 2006, 108, 3654–3661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Zwaan, C.M.; Meshinchi, S.; Radich, J.P.; Veerman, A.J.; Huismans, D.R.; Munske, L.; Podleschny, M.; Hahlen, K.; Pieters, R.; Zimmermann, M.; et al. FLT3 internal tandem duplication in 234 children with acute myeloid leukemia: Prognostic significance and relation to cellular drug resistance. Blood 2003, 102, 2387–2394. [Google Scholar] [CrossRef] [Green Version]
  57. Brunet, S.; Labopin, M.; Esteve, J.; Cornelissen, J.; Socie, G.; Iori, A.P.; Verdonck, L.F.; Volin, L.; Gratwohl, A.; Sierra, J.; et al. Impact of FLT3 internal tandem duplication on the outcome of related and unrelated hematopoietic transplantation for adult acute myeloid leukemia in first remission: A retrospective analysis. J. Clin. Oncol. 2012, 30, 735–741. [Google Scholar] [CrossRef]
  58. Pratcorona, M.; Brunet, S.; Nomdedeu, J.; Ribera, J.M.; Tormo, M.; Duarte, R.; Escoda, L.; Guardia, R.; Queipo de Llano, M.P.; Salamero, O.; et al. Favorable outcome of patients with acute myeloid leukemia harboring a low-allelic burden FLT3-ITD mutation and concomitant NPM1 mutation: Relevance to post-remission therapy. Blood 2013, 121, 2734–2738. [Google Scholar] [CrossRef] [Green Version]
  59. DeZern, A.E.; Sung, A.; Kim, S.; Smith, B.D.; Karp, J.E.; Gore, S.D.; Jones, R.J.; Fuchs, E.; Luznik, L.; McDevitt, M.; et al. Role of allogeneic transplantation for FLT3/ITD acute myeloid leukemia: Outcomes from 133 consecutive newly diagnosed patients from a single institution. Biol Blood Marrow Transpl. 2011, 17, 1404–1409. [Google Scholar] [CrossRef] [Green Version]
  60. Balgobind, B.V.; Raimondi, S.C.; Harbott, J.; Zimmermann, M.; Alonzo, T.A.; Auvrignon, A.; Beverloo, H.B.; Chang, M.; Creutzig, U.; Dworzak, M.N.; et al. Novel prognostic subgroups in childhood 11q23/MLL-rearranged acute myeloid leukemia: Results of an international retrospective study. Blood 2009, 114, 2489–2496. [Google Scholar] [CrossRef] [Green Version]
  61. Locatelli, F.; Masetti, R.; Rondelli, R.; Zecca, M.; Fagioli, F.; Rovelli, A.; Messina, C.; Lanino, E.; Bertaina, A.; Favre, C.; et al. Outcome of children with high-risk acute myeloid leukemia given autologous or allogeneic hematopoietic cell transplantation in the aieop AML-2002/01 study. Bone Marrow Transpl. 2015, 50, 181–188. [Google Scholar] [CrossRef] [Green Version]
  62. Sauer, M.G.; Lang, P.J.; Albert, M.H.; Bader, P.; Creutzig, U.; Eyrich, M.; Greil, J.; Gruhn, B.; Holter, W.; Klingebiel, T.; et al. Hematopoietic stem cell transplantation for children with acute myeloid leukemia-results of the AML SCT-BFM 2007 trial. Leukemia 2020, 34, 613–624. [Google Scholar] [CrossRef]
  63. Struski, S.; Lagarde, S.; Bories, P.; Puiseux, C.; Prade, N.; Cuccuini, W.; Pages, M.P.; Bidet, A.; Gervais, C.; Lafage-Pochitaloff, M.; et al. NUP98 is rearranged in 3.8% of pediatric AML forming a clinical and molecular homogenous group with a poor prognosis. Leukemia 2017, 31, 565–572. [Google Scholar] [CrossRef]
  64. Bisio, V.; Zampini, M.; Tregnago, C.; Manara, E.; Salsi, V.; Di Meglio, A.; Masetti, R.; Togni, M.; Di Giacomo, D.; Minuzzo, S.; et al. NUP98-fusion transcripts characterize different biological entities within acute myeloid leukemia: A report from the AIEOP-AML group. Leukemia 2017, 31, 974–977. [Google Scholar] [CrossRef]
  65. de Rooij, J.D.; Branstetter, C.; Ma, J.; Li, Y.; Walsh, M.P.; Cheng, J.; Obulkasim, A.; Dang, J.; Easton, J.; Verboon, L.J.; et al. Pediatric non-Down syndrome acute megakaryoblastic leukemia is characterized by distinct genomic subsets with varying outcomes. Nat. Genet. 2017, 49, 451–456. [Google Scholar] [CrossRef] [Green Version]
  66. Masetti, R.; Pigazzi, M.; Togni, M.; Astolfi, A.; Indio, V.; Manara, E.; Casadio, R.; Pession, A.; Basso, G.; Locatelli, F. CBFA2T3-GLIS2 fusion transcript is a novel common feature in pediatric, cytogenetically normal AML, not restricted to FAB M7 subtype. Blood 2013, 121, 3469–3472. [Google Scholar] [CrossRef] [Green Version]
  67. Gruber, T.A.; Larson Gedman, A.; Zhang, J.; Koss, C.S.; Marada, S.; Ta, H.Q.; Chen, S.C.; Su, X.; Ogden, S.K.; Dang, J.; et al. An Inv(16)(p13.3q24.3)-encoded CBFA2T3-GLIS2 fusion protein defines an aggressive subtype of pediatric acute megakaryoblastic leukemia. Cancer Cell 2012, 22, 683–697. [Google Scholar] [CrossRef] [Green Version]
  68. von Bergh, A.R.; van Drunen, E.; van Wering, E.R.; van Zutven, L.J.; Hainmann, I.; Lonnerholm, G.; Meijerink, J.P.; Pieters, R.; Beverloo, H.B. High incidence of t(7;12)(q36;p13) in infant AML but not in infant ALL, with a dismal outcome and ectopic expression of HLXB9. Genes Chromosomes Cancer 2006, 45, 731–739. [Google Scholar] [CrossRef]
  69. Shiba, N.; Ichikawa, H.; Taki, T.; Park, M.J.; Jo, A.; Mitani, S.; Kobayashi, T.; Shimada, A.; Sotomatsu, M.; Arakawa, H.; et al. NUP98-NSD1 gene fusion and its related gene expression signature are strongly associated with a poor prognosis in pediatric acute myeloid leukemia. Genes Chromosomes Cancer 2013, 52, 683–693. [Google Scholar] [CrossRef]
  70. Noort, S.; Wander, P.; Alonzo, T.A.; Smith, J.; Ries, R.E.; Gerbing, R.B.; Dolman, M.E.M.; Locatelli, F.; Reinhardt, D.; Baruchel, A.; et al. The clinical and biological characteristics of NUP98-KDM5A in pediatric acute myeloid leukemia. Haematologica 2021, 106, 630–634. [Google Scholar] [CrossRef]
  71. Coenen, E.A.; Zwaan, C.M.; Reinhardt, D.; Harrison, C.J.; Haas, O.A.; de Haas, V.; Mihal, V.; De Moerloose, B.; Jeison, M.; Rubnitz, J.E.; et al. Pediatric acute myeloid leukemia with t(8;16)(p11;p13), a distinct clinical and biological entity: A collaborative study by the International-Berlin-Frankfurt-Munster AML-study group. Blood 2013, 122, 2704–2713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Tierens, A.; Bjorklund, E.; Siitonen, S.; Marquart, H.V.; Wulff-Juergensen, G.; Pelliniemi, T.T.; Forestier, E.; Hasle, H.; Jahnukainen, K.; Lausen, B.; et al. Residual disease detected by flow cytometry is an independent predictor of survival in childhood acute myeloid leukaemia; Results of the NOPHO-AML 2004 study. Br. J. Haematol. 2016, 174, 600–609. [Google Scholar] [CrossRef]
  73. Buldini, B.; Rizzati, F.; Masetti, R.; Fagioli, F.; Menna, G.; Micalizzi, C.; Putti, M.C.; Rizzari, C.; Santoro, N.; Zecca, M.; et al. Prognostic significance of flow-cytometry evaluation of minimal residual disease in children with acute myeloid leukaemia treated according to the AIEOP-AML 2002/01 study protocol. Br. J. Haematol. 2017, 177, 116–126. [Google Scholar] [CrossRef]
  74. Loken, M.R.; Alonzo, T.A.; Pardo, L.; Gerbing, R.B.; Raimondi, S.C.; Hirsch, B.A.; Ho, P.A.; Franklin, J.; Cooper, T.M.; Gamis, A.S.; et al. Residual disease detected by multidimensional flow cytometry signifies high relapse risk in patients with de novo acute myeloid leukemia: A report from Children’s Oncology Group. Blood 2012, 120, 1581–1588. [Google Scholar] [CrossRef]
  75. Inaba, H.; Coustan-Smith, E.; Cao, X.; Pounds, S.B.; Shurtleff, S.A.; Wang, K.Y.; Raimondi, S.C.; Onciu, M.; Jacobsen, J.; Ribeiro, R.C.; et al. Comparative analysis of different approaches to measure treatment response in acute myeloid leukemia. J. Clin. Oncol. 2012, 30, 3625–3632. [Google Scholar] [CrossRef] [Green Version]
  76. Hoffman, A.E.; Schoonmade, L.J.; Kaspers, G.J. Pediatric relapsed acute myeloid leukemia: A systematic review. Expert Rev. Anticancer Ther. 2021, 21, 45–52. [Google Scholar] [CrossRef]
  77. Meshinchi, S.; Leisenring, W.M.; Carpenter, P.A.; Woolfrey, A.E.; Sievers, E.L.; Radich, J.P.; Sanders, J.E. Survival after second hematopoietic stem cell transplantation for recurrent pediatric acute myeloid leukemia. Biol. Blood Marrow Transpl. 2003, 9, 706–713. [Google Scholar] [CrossRef] [Green Version]
  78. Naik, S.; Martinez, C.; Leung, K.; Sasa, G.; Nguyen, N.Y.; Wu, M.F.; Gottschalk, S.; Brenner, M.; Heslop, H.; Krance, R. Outcomes after Second Hematopoietic Stem Cell Transplantations in Pediatric Patients with Relapsed Hematological Malignancies. Biol. Blood Marrow Transpl. 2015, 21, 1266–1272. [Google Scholar] [CrossRef] [Green Version]
  79. Taga, T.; Murakami, Y.; Tabuchi, K.; Adachi, S.; Tomizawa, D.; Kojima, Y.; Kato, K.; Koike, K.; Koh, K.; Kajiwara, R.; et al. Role of Second Transplantation for Children with Acute Myeloid Leukemia Following Posttransplantation Relapse. Pediatr. Blood Cancer 2016, 63, 701–705. [Google Scholar] [CrossRef]
  80. Uden, T.; Bertaina, A.; Abrahamsson, J.; Ansari, M.; Balduzzi, A.; Bourquin, J.P.; Gerhardt, C.; Bierings, M.; Hasle, H.; Lankester, A.; et al. Outcome of children relapsing after first allogeneic haematopoietic stem cell transplantation for acute myeloid leukaemia: A retrospective I-BFM analysis of 333 children. Br. J. Haematol. 2020, 189, 745–750. [Google Scholar] [CrossRef] [PubMed]
  81. Rubnitz, J.E.; Inaba, H. Childhood acute myeloid leukaemia. Br. J. Haematol. 2012, 159, 259–276. [Google Scholar] [CrossRef]
  82. O’Hare, P.; Lucchini, G.; Cummins, M.; Veys, P.; Potter, M.; Lawson, S.; Vora, A.; Wynn, R.; Peniket, A.; Kirkland, K.; et al. Allogeneic stem cell transplantation for refractory acute myeloid leukemia in pediatric patients: The UK experience. Bone Marrow Transpl. 2017, 52, 825–831. [Google Scholar] [CrossRef] [Green Version]
  83. Quarello, P.; Fagioli, F.; Basso, G.; Putti, M.C.; Berger, M.; Luciani, M.; Rizzari, C.; Menna, G.; Masetti, R.; Locatelli, F. Outcome of children with acute myeloid leukaemia (AML) experiencing primary induction failure in the AIEOP AML 2002/01 clinical trial. Br. J. Haematol. 2015, 171, 566–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Willasch, A.M.; Peters, C.; Sedlacek, P.; Dalle, J.H.; Kitra-Roussou, V.; Yesilipek, A.; Wachowiak, J.; Lankester, A.; Prete, A.; Hamidieh, A.A.; et al. Myeloablative conditioning for allo-HSCT in pediatric ALL: FTBI or chemotherapy?-A multicenter EBMT-PDWP study. Bone Marrow Transpl. 2020, 55, 1540–1551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Brochstein, J.A.; Kernan, N.A.; Groshen, S.; Cirrincione, C.; Shank, B.; Emanuel, D.; Laver, J.; O’Reilly, R.J. Allogeneic bone marrow transplantation after hyperfractionated total-body irradiation and cyclophosphamide in children with acute leukemia. N. Engl. J. Med. 1987, 317, 1618–1624. [Google Scholar] [CrossRef]
  86. Marks, D.I.; Forman, S.J.; Blume, K.G.; Perez, W.S.; Weisdorf, D.J.; Keating, A.; Gale, R.P.; Cairo, M.S.; Copelan, E.A.; Horan, J.T.; et al. A comparison of cyclophosphamide and total body irradiation with etoposide and total body irradiation as conditioning regimens for patients undergoing sibling allografting for acute lymphoblastic leukemia in first or second complete remission. Biol. Blood Marrow Transpl. 2006, 12, 438–453. [Google Scholar] [CrossRef] [Green Version]
  87. Davies, S.M.; Ramsay, N.K.; Klein, J.P.; Weisdorf, D.J.; Bolwell, B.; Cahn, J.Y.; Camitta, B.M.; Gale, R.P.; Giralt, S.; Heilmann, C.; et al. Comparison of preparative regimens in transplants for children with acute lymphoblastic leukemia. J. Clin. Oncol. 2000, 18, 340–347. [Google Scholar] [CrossRef]
  88. Tutschka, P.J.; Copelan, E.A.; Klein, J.P. Bone marrow transplantation for leukemia following a new busulfan and cyclophosphamide regimen. Blood 1987, 70, 1382–1388. [Google Scholar] [CrossRef] [Green Version]
  89. Tracey, J.; Zhang, M.J.; Thiel, E.; Sobocinski, K.A.; Eapen, M. Transplantation conditioning regimens and outcomes after allogeneic hematopoietic cell transplantation in children and adolescents with acute lymphoblastic leukemia. Biol. Blood Marrow Transpl. 2013, 19, 255–259. [Google Scholar] [CrossRef] [Green Version]
  90. Bunin, N.; Aplenc, R.; Kamani, N.; Shaw, K.; Cnaan, A.; Simms, S. Randomized trial of busulfan vs. total body irradiation containing conditioning regimens for children with acute lymphoblastic leukemia: A Pediatric Blood and Marrow Transplant Consortium study. Bone Marrow Transpl. 2003, 32, 543–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Zecca, M.; Pession, A.; Messina, C.; Bonetti, F.; Favre, C.; Prete, A.; Cesaro, S.; Porta, F.; Mazzarino, I.; Giorgiani, G.; et al. Total body irradiation, thiotepa, and cyclophosphamide as a conditioning regimen for children with acute lymphoblastic leukemia in first or second remission undergoing bone marrow transplantation with HLA-identical siblings. J. Clin. Oncol. 1999, 17, 1838–1846. [Google Scholar] [CrossRef]
  92. Peters, C.; Schrappe, M.; von Stackelberg, A.; Schrauder, A.; Bader, P.; Ebell, W.; Lang, P.; Sykora, K.W.; Schrum, J.; Kremens, B.; et al. Stem-cell transplantation in children with acute lymphoblastic leukemia: A prospective international multicenter trial comparing sibling donors with matched unrelated donors-The ALL-SCT-BFM-2003 trial. J. Clin. Oncol. 2015, 33, 1265–1274. [Google Scholar] [CrossRef] [PubMed]
  93. Kato, M.; Ishida, H.; Koh, K.; Inagaki, J.; Kato, K.; Goto, H.; Kaneko, T.; Cho, Y.; Hashii, Y.; Kurosawa, H.; et al. Comparison of chemotherapeutic agents as a myeloablative conditioning with total body irradiation for pediatric acute lymphoblastic leukemia: A study from the pediatric ALL working group of the Japan Society for Hematopoietic Cell Transplantation. Pediatr. Blood Cancer 2015, 62, 1844–1850. [Google Scholar] [CrossRef] [PubMed]
  94. Saglio, F.; Zecca, M.; Pagliara, D.; Giorgiani, G.; Balduzzi, A.; Calore, E.; Favre, C.; Faraci, M.; Prete, A.; Tambaro, F.P.; et al. Occurrence of long-term effects after hematopoietic stem cell transplantation in children affected by acute leukemia receiving either busulfan or total body irradiation: Results of an AIEOP (Associazione Italiana Ematologia Oncologia Pediatrica) retrospective study. Bone Marrow Transpl. 2020, 55, 1918–1927. [Google Scholar] [CrossRef]
  95. Peters, C.; Dalle, J.H.; Locatelli, F.; Poetschger, U.; Sedlacek, P.; Buechner, J.; Shaw, P.J.; Staciuk, R.; Ifversen, M.; Pichler, H.; et al. Total Body Irradiation or Chemotherapy Conditioning in Childhood ALL: A Multinational, Randomized, Noninferiority Phase III Study. J. Clin. Oncol. 2021, 39, 295–307. [Google Scholar] [CrossRef] [PubMed]
  96. Blaise, D.; Maraninchi, D.; Archimbaud, E.; Reiffers, J.; Devergie, A.; Jouet, J.P.; Milpied, N.; Attal, M.; Michallet, M.; Ifrah, N.; et al. Allogeneic bone marrow transplantation for acute myeloid leukemia in first remission: A randomized trial of a busulfan-Cytoxan versus Cytoxan-total body irradiation as preparative regimen: A report from the Group d’Etudes de la Greffe de Moelle Osseuse. Blood 1992, 79, 2578–2582. [Google Scholar] [CrossRef] [PubMed]
  97. Gupta, T.; Kannan, S.; Dantkale, V.; Laskar, S. Cyclophosphamide plus total body irradiation compared with busulfan plus cyclophosphamide as a conditioning regimen prior to hematopoietic stem cell transplantation in patients with leukemia: A systematic review and meta-analysis. Hematol. Oncol. Stem Cell Ther. 2011, 4, 17–29. [Google Scholar] [CrossRef] [Green Version]
  98. Ringden, O.; Ruutu, T.; Remberger, M.; Nikoskelainen, J.; Volin, L.; Vindelov, L.; Parkkali, T.; Lenhoff, S.; Sallerfors, B.; Ljungman, P.; et al. A randomized trial comparing busulfan with total body irradiation as conditioning in allogeneic marrow transplant recipients with leukemia: A report from the Nordic Bone Marrow Transplantation Group. Blood 1994, 83, 2723–2730. [Google Scholar] [CrossRef]
  99. Ishida, H.; Kato, M.; Kudo, K.; Taga, T.; Tomizawa, D.; Miyamura, T.; Goto, H.; Inagaki, J.; Koh, K.; Terui, K.; et al. Comparison of Outcomes for Pediatric Patients with Acute Myeloid Leukemia in Remission and Undergoing Allogeneic Hematopoietic Cell Transplantation with Myeloablative Conditioning Regimens Based on Either Intravenous Busulfan or Total Body Irradiation: A Report from the Japanese Society for Hematopoietic Cell Transplantation. Biol. Blood Marrow Transpl. 2015, 21, 2141–2147. [Google Scholar] [CrossRef] [Green Version]
  100. de Berranger, E.; Cousien, A.; Petit, A.; Peffault de Latour, R.; Galambrun, C.; Bertrand, Y.; Salmon, A.; Rialland, F.; Rohrlich, P.S.; Vannier, J.P.; et al. Impact on long-term OS of conditioning regimen in allogeneic BMT for children with AML in first CR: TBI + CY versus BU + CY: A report from the Societe Francaise de Greffe de Moelle et de Therapie Cellulaire. Bone Marrow Transpl. 2014, 49, 382–388. [Google Scholar] [CrossRef] [Green Version]
  101. Dandoy, C.E.; Davies, S.M.; Woo Ahn, K.; He, Y.; Kolb, A.E.; Levine, J.; Bo-Subait, S.; Abdel-Azim, H.; Bhatt, N.; Chewing, J.; et al. Comparison of total body irradiation versus non-total body irradiation containing regimens for de novo acute myeloid leukemia in children. Haematologica 2021, 106, 1839–1845. [Google Scholar] [CrossRef] [PubMed]
  102. Phillips, G.L.; Shepherd, J.D.; Barnett, M.J.; Lansdorp, P.M.; Klingemann, H.G.; Spinelli, J.J.; Nevill, T.J.; Chan, K.W.; Reece, D.E. Busulfan, cyclophosphamide, and melphalan conditioning for autologous bone marrow transplantation in hematologic malignancy. J. Clin. Oncol. 1991, 9, 1880–1888. [Google Scholar] [CrossRef] [PubMed]
  103. Lucchini, G.; Labopin, M.; Beohou, E.; Dalissier, A.; Dalle, J.H.; Cornish, J.; Zecca, M.; Samarasinghe, S.; Gibson, B.; Locatelli, F.; et al. Impact of Conditioning Regimen on Outcomes for Children with Acute Myeloid Leukemia Undergoing Transplantation in First Complete Remission. An Analysis on Behalf of the Pediatric Disease Working Party of the European Group for Blood and Marrow Transplantation. Biol. Blood Marrow Transpl. 2017, 23, 467–474. [Google Scholar] [CrossRef] [Green Version]
  104. Muramatsu, H.; Sakaguchi, H.; Taga, T.; Tabuchi, K.; Adachi, S.; Inoue, M.; Kitoh, T.; Suminoe, A.; Yabe, H.; Azuma, E.; et al. Reduced intensity conditioning in allogeneic stem cell transplantation for AML with Down syndrome. Pediatr. Blood Cancer 2014, 61, 925–927. [Google Scholar] [CrossRef]
  105. Giardino, S.; de Latour, R.P.; Aljurf, M.; Eikema, D.J.; Bosman, P.; Bertrand, Y.; Tbakhi, A.; Holter, W.; Bornhauser, M.; Rossig, C.; et al. Outcome of patients with Fanconi anemia developing myelodysplasia and acute leukemia who received allogeneic hematopoietic stem cell transplantation: A retrospective analysis on behalf of EBMT group. Am. J. Hematol. 2020, 95, 809–816. [Google Scholar] [CrossRef]
  106. Bitan, M.; He, W.; Zhang, M.J.; Abdel-Azim, H.; Ayas, M.F.; Bielorai, B.; Carpenter, P.A.; Cairo, M.S.; Diaz, M.A.; Horan, J.T.; et al. Transplantation for children with acute myeloid leukemia: A comparison of outcomes with reduced intensity and myeloablative regimens. Blood 2014, 123, 1615–1620. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Strocchio, L.; Zecca, M.; Comoli, P.; Mina, T.; Giorgiani, G.; Giraldi, E.; Vinti, L.; Merli, P.; Regazzi, M.; Locatelli, F. Treosulfan-based conditioning regimen for allogeneic haematopoietic stem cell transplantation in children with sickle cell disease. Br. J. Haematol. 2015, 169, 726–736. [Google Scholar] [CrossRef]
  108. Boztug, H.; Zecca, M.; Sykora, K.W.; Veys, P.; Lankester, A.; Slatter, M.; Skinner, R.; Wachowiak, J.; Potschger, U.; Glogova, E.; et al. Treosulfan-based conditioning regimens for allogeneic HSCT in children with acute lymphoblastic leukaemia. Ann. Hematol. 2015, 94, 297–306. [Google Scholar] [CrossRef] [Green Version]
  109. Kalwak, K.; Mielcarek, M.; Patrick, K.; Styczynski, J.; Bader, P.; Corbacioglu, S.; Burkhardt, B.; Sykora, K.W.; Drabko, K.; Gozdzik, J.; et al. Treosulfan-fludarabine-thiotepa-based conditioning treatment before allogeneic hematopoietic stem cell transplantation for pediatric patients with hematological malignancies. Bone Marrow Transpl. 2020, 55, 1996–2007. [Google Scholar] [CrossRef] [Green Version]
  110. Satwani, P.; Bhatia, M.; Garvin, J.H., Jr.; George, D.; Dela Cruz, F.; Le Gall, J.; Jin, Z.; Schwartz, J.; Duffy, D.; van de Ven, C.; et al. A Phase I study of gemtuzumab ozogamicin (GO) in combination with busulfan and cyclophosphamide (Bu/Cy) and allogeneic stem cell transplantation in children with poor-risk CD33+ AML: A new targeted immunochemotherapy myeloablative conditioning (MAC) regimen. Biol. Blood Marrow Transpl. 2012, 18, 324–329. [Google Scholar] [CrossRef] [Green Version]
  111. Nemecek, E.R.; Hilger, R.A.; Adams, A.; Shaw, B.E.; Kiefer, D.; Le-Rademacher, J.; Levine, J.E.; Yanik, G.; Leung, W.; Talano, J.A.; et al. Treosulfan, Fludarabine, and Low-Dose Total Body Irradiation for Children and Young Adults with Acute Myeloid Leukemia or Myelodysplastic Syndrome Undergoing Allogeneic Hematopoietic Cell Transplantation: Prospective Phase II Trial of the Pediatric Blood and Marrow Transplant Consortium. Biol. Blood Marrow Transpl. 2018, 24, 1651–1656. [Google Scholar] [CrossRef] [Green Version]
  112. Oshrine, B.; Adams, L.; Nguyen, A.T.H.; Amankwah, E.; Shyr, D.; Hale, G.; Petrovic, A. Comparison of melphalan- And busulfan-based myeloablative conditioning in children undergoing allogeneic transplantation for acute myeloid leukemia or myelodysplasia. Pediatr. Transpl. 2020, 24, e13672. [Google Scholar] [CrossRef] [PubMed]
  113. Kato, K.; Yoshida, N.; Matsumoto, K.; Matsuyama, T. Fludarabine, cytarabine, granulocyte colony-stimulating factor and melphalan (FALG with L-PAM) as a reduced toxicity conditioning regimen in children with acute leukemia. Pediatr. Blood Cancer 2014, 61, 712–716. [Google Scholar] [CrossRef] [PubMed]
  114. Kussman, A.; Shyr, D.; Hale, G.; Oshrine, B.; Petrovic, A. Allogeneic hematopoietic cell transplantation in chemotherapy-induced aplasia in children with high-risk acute myeloid leukemia or myelodysplasia. Pediatr. Blood Cancer 2019, 66, e27481. [Google Scholar] [CrossRef]
  115. Versluys, A.B.; Boelens, J.J.; Pronk, C.; Lankester, A.; Bordon, V.; Buechner, J.; Ifversen, M.; Jackmann, N.; Sundin, M.; Vettenranta, K.; et al. Hematopoietic cell transplant in pediatric acute myeloid leukemia after similar upfront therapy; a comparison of conditioning regimens. Bone Marrow Transpl. 2021, 56, 1426–1432. [Google Scholar] [CrossRef]
  116. Locatelli, F.; Merli, P.; Bertaina, A. Rabbit anti-human T-lymphocyte globulin and hematopoietic transplantation. Oncotarget 2017, 8, 96460–96461. [Google Scholar] [CrossRef]
  117. Locatelli, F.; Bernardo, M.E.; Bertaina, A.; Rognoni, C.; Comoli, P.; Rovelli, A.; Pession, A.; Fagioli, F.; Favre, C.; Lanino, E.; et al. Efficacy of two different doses of rabbit anti-T-lymphocyte globulin to prevent graft-versus-host disease in children with haematological malignancies transplanted from an unrelated donor: A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2017, 18, 1126–1136. [Google Scholar] [CrossRef]
  118. Schrappe, M.; Valsecchi, M.G.; Bartram, C.R.; Schrauder, A.; Panzer-Grumayer, R.; Moricke, A.; Parasole, R.; Zimmermann, M.; Dworzak, M.; Buldini, B.; et al. Late MRD response determines relapse risk overall and in subsets of childhood T-cell ALL: Results of the AIEOP-BFM-ALL 2000 study. Blood 2011, 118, 2077–2084. [Google Scholar] [CrossRef]
  119. Vora, A.; Goulden, N.; Mitchell, C.; Hancock, J.; Hough, R.; Rowntree, C.; Moorman, A.V.; Wade, R. Augmented post-remission therapy for a minimal residual disease-defined high-risk subgroup of children and young people with clinical standard-risk and intermediate-risk acute lymphoblastic leukaemia (UKALL 2003): A randomised controlled trial. Lancet Oncol. 2014, 15, 809–818. [Google Scholar] [CrossRef]
  120. San Miguel, J.F.; Vidriales, M.B.; Lopez-Berges, C.; Diaz-Mediavilla, J.; Gutierrez, N.; Canizo, C.; Ramos, F.; Calmuntia, M.J.; Perez, J.J.; Gonzalez, M.; et al. Early immunophenotypical evaluation of minimal residual disease in acute myeloid leukemia identifies different patient risk groups and may contribute to postinduction treatment stratification. Blood 2001, 98, 1746–1751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Knechtli, C.J.; Goulden, N.J.; Hancock, J.P.; Grandage, V.L.; Harris, E.L.; Garland, R.J.; Jones, C.G.; Rowbottom, A.W.; Hunt, L.P.; Green, A.F.; et al. Minimal residual disease status before allogeneic bone marrow transplantation is an important determinant of successful outcome for children and adolescents with acute lymphoblastic leukemia. Blood 1998, 92, 4072–4079. [Google Scholar] [CrossRef] [PubMed]
  122. Bader, P.; Kreyenberg, H.; Henze, G.H.; Eckert, C.; Reising, M.; Willasch, A.; Barth, A.; Borkhardt, A.; Peters, C.; Handgretinger, R.; et al. Prognostic value of minimal residual disease quantification before allogeneic stem-cell transplantation in relapsed childhood acute lymphoblastic leukemia: The ALL-REZ BFM Study Group. J. Clin. Oncol. 2009, 27, 377–384. [Google Scholar] [CrossRef] [PubMed]
  123. Eckert, C.; Hagedorn, N.; Sramkova, L.; Mann, G.; Panzer-Grumayer, R.; Peters, C.; Bourquin, J.P.; Klingebiel, T.; Borkhardt, A.; Cario, G.; et al. Monitoring minimal residual disease in children with high-risk relapses of acute lymphoblastic leukemia: Prognostic relevance of early and late assessment. Leukemia 2015, 29, 1648–1655. [Google Scholar] [CrossRef] [PubMed]
  124. Lovisa, F.; Zecca, M.; Rossi, B.; Campeggio, M.; Magrin, E.; Giarin, E.; Buldini, B.; Songia, S.; Cazzaniga, G.; Mina, T.; et al. Pre- and post-transplant minimal residual disease predicts relapse occurrence in children with acute lymphoblastic leukaemia. Br. J. Haematol. 2018, 180, 680–693. [Google Scholar] [CrossRef] [PubMed]
  125. Pigazzi, M.; Benetton, M.; Walter, C.; Hansen, M.; Skou, A.-S.; Da Ros, A.; Marchetti, A.; Polato, K.; Belloni, M.; Tregnago, C.; et al. Impact of Minimal Residual Disease (MRD) Assessed before Transplantation on the Outcome of Children with Acute Myeloid Leukemia Given an Allograft: A Retrospective Study By the I-BFM Study Group. Blood 2020, 136, 38–39. [Google Scholar] [CrossRef]
  126. Leung, W.; Pui, C.H.; Coustan-Smith, E.; Yang, J.; Pei, D.; Gan, K.; Srinivasan, A.; Hartford, C.; Triplett, B.M.; Dallas, M.; et al. Detectable minimal residual disease before hematopoietic cell transplantation is prognostic but does not preclude cure for children with very-high-risk leukemia. Blood 2012, 120, 468–472. [Google Scholar] [CrossRef] [Green Version]
  127. Pulsipher, M.A.; Langholz, B.; Wall, D.A.; Schultz, K.R.; Bunin, N.; Carroll, W.L.; Raetz, E.; Gardner, S.; Gastier-Foster, J.M.; Howrie, D.; et al. The addition of sirolimus to tacrolimus/methotrexate GVHD prophylaxis in children with ALL: A phase 3 Children’s Oncology Group/Pediatric Blood and Marrow Transplant Consortium trial. Blood 2014, 123, 2017–2025. [Google Scholar] [CrossRef]
  128. Zhou, Y.; Othus, M.; Araki, D.; Wood, B.L.; Radich, J.P.; Halpern, A.B.; Mielcarek, M.; Estey, E.H.; Appelbaum, F.R.; Walter, R.B. Pre- and post-transplant quantification of measurable (‘minimal’) residual disease via multiparameter flow cytometry in adult acute myeloid leukemia. Leukemia 2016, 30, 1456–1464. [Google Scholar] [CrossRef]
  129. Jacobsohn, D.A.; Loken, M.R.; Fei, M.; Adams, A.; Brodersen, L.E.; Logan, B.R.; Ahn, K.W.; Shaw, B.E.; Kletzel, M.; Olszewski, M.; et al. Outcomes of Measurable Residual Disease in Pediatric Acute Myeloid Leukemia before and after Hematopoietic Stem Cell Transplant: Validation of Difference from Normal Flow Cytometry with Chimerism Studies and Wilms Tumor 1 Gene Expression. Biol. Blood Marrow Transpl. 2018, 24, 2040–2046. [Google Scholar] [CrossRef] [Green Version]
  130. Buckley, S.A.; Wood, B.L.; Othus, M.; Hourigan, C.S.; Ustun, C.; Linden, M.A.; DeFor, T.E.; Malagola, M.; Anthias, C.; Valkova, V.; et al. Minimal residual disease prior to allogeneic hematopoietic cell transplantation in acute myeloid leukemia: A meta-analysis. Haematologica 2017, 102, 865–873. [Google Scholar] [CrossRef] [Green Version]
  131. Faham, M.; Zheng, J.; Moorhead, M.; Carlton, V.E.; Stow, P.; Coustan-Smith, E.; Pui, C.H.; Campana, D. Deep-sequencing approach for minimal residual disease detection in acute lymphoblastic leukemia. Blood 2012, 120, 5173–5180. [Google Scholar] [CrossRef] [Green Version]
  132. Pulsipher, M.A.; Carlson, C.; Langholz, B.; Wall, D.A.; Schultz, K.R.; Bunin, N.; Kirsch, I.; Gastier-Foster, J.M.; Borowitz, M.; Desmarais, C.; et al. IgH-V(D)J NGS-MRD measurement pre- and early post-allotransplant defines very low- and very high-risk ALL patients. Blood 2015, 125, 3501–3508. [Google Scholar] [CrossRef] [Green Version]
  133. Kim, T.; Moon, J.H.; Ahn, J.S.; Kim, Y.K.; Lee, S.S.; Ahn, S.Y.; Jung, S.H.; Yang, D.H.; Lee, J.J.; Choi, S.H.; et al. Next-generation sequencing-based posttransplant monitoring of acute myeloid leukemia identifies patients at high risk of relapse. Blood 2018, 132, 1604–1613. [Google Scholar] [CrossRef] [Green Version]
  134. Thol, F.; Gabdoulline, R.; Liebich, A.; Klement, P.; Schiller, J.; Kandziora, C.; Hambach, L.; Stadler, M.; Koenecke, C.; Flintrop, M.; et al. Measurable residual disease monitoring by NGS before allogeneic hematopoietic cell transplantation in AML. Blood 2018, 132, 1703–1713. [Google Scholar] [CrossRef] [Green Version]
  135. Sun, W.; Orgel, E.; Malvar, J.; Sposto, R.; Wilkes, J.J.; Gardner, R.; Tolbert, V.P.; Smith, A.; Hur, M.; Hoffman, J.; et al. Treatment-related adverse events associated with a modified UK ALLR3 induction chemotherapy backbone for childhood relapsed/refractory acute lymphoblastic leukemia. Pediatr. Blood Cancer 2016, 63, 1943–1948. [Google Scholar] [CrossRef]
  136. Oskarsson, T.; Soderhall, S.; Arvidson, J.; Forestier, E.; Frandsen, T.L.; Hellebostad, M.; Lahteenmaki, P.; Jonsson, O.G.; Myrberg, I.H.; Heyman, M.; et al. Treatment-related mortality in relapsed childhood acute lymphoblastic leukemia. Pediatr. Blood Cancer 2018, 65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. von Stackelberg, A.; Locatelli, F.; Zugmaier, G.; Handgretinger, R.; Trippett, T.M.; Rizzari, C.; Bader, P.; O’Brien, M.M.; Brethon, B.; Bhojwani, D.; et al. Phase I/Phase II Study of Blinatumomab in Pediatric Patients with Relapsed/Refractory Acute Lymphoblastic Leukemia. J. Clin. Oncol. 2016, 34, 4381–4389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Locatelli, F.; Zugmaier, G.; Mergen, N.; Bader, P.; Jeha, S.; Schlegel, P.G.; Bourquin, J.P.; Handgretinger, R.; Brethon, B.; Rossig, C.; et al. Blinatumomab in pediatric patients with relapsed/refractory acute lymphoblastic leukemia: Results of the RIALTO trial, an expanded access study. Blood Cancer J. 2020, 10, 77. [Google Scholar] [CrossRef]
  139. Locatelli, F.; Zugmaier, G.; Rizzari, C.; Morris, J.D.; Gruhn, B.; Klingebiel, T.; Parasole, R.; Linderkamp, C.; Flotho, C.; Petit, A.; et al. Effect of Blinatumomab vs. Chemotherapy on Event-Free Survival among Children with High-risk First-Relapse B-Cell Acute Lymphoblastic Leukemia: A Randomized Clinical Trial. JAMA 2021, 325, 843–854. [Google Scholar] [CrossRef] [PubMed]
  140. Brown, P.A.; Ji, L.; Xu, X.; Devidas, M.; Hogan, L.E.; Borowitz, M.J.; Raetz, E.A.; Zugmaier, G.; Sharon, E.; Bernhardt, M.B.; et al. Effect of Postreinduction Therapy Consolidation with Blinatumomab vs. Chemotherapy on Disease-Free Survival in Children, Adolescents, and Young Adults with First Relapse of B-Cell Acute Lymphoblastic Leukemia: A Randomized Clinical Trial. JAMA 2021, 325, 833–842. [Google Scholar] [CrossRef]
  141. Brivio, E.; Locatelli, F.; Lopez-Yurda, M.; Malone, A.; Diaz-de-Heredia, C.; Bielorai, B.; Rossig, C.; van der Velden, V.H.J.; Ammerlaan, A.C.J.; Thano, A.; et al. A phase 1 study of inotuzumab ozogamicin in pediatric relapsed/refractory acute lymphoblastic leukemia (ITCC-059 study). Blood 2021, 137, 1582–1590. [Google Scholar] [CrossRef]
  142. Bhojwani, D.; Sposto, R.; Shah, N.N.; Rodriguez, V.; Yuan, C.; Stetler-Stevenson, M.; O’Brien, M.M.; McNeer, J.L.; Quereshi, A.; Cabannes, A.; et al. Inotuzumab ozogamicin in pediatric patients with relapsed/refractory acute lymphoblastic leukemia. Leukemia 2019, 33, 884–892. [Google Scholar] [CrossRef] [PubMed]
  143. Shah, N.N.; Lee, D.W.; Yates, B.; Yuan, C.M.; Shalabi, H.; Martin, S.; Wolters, P.L.; Steinberg, S.M.; Baker, E.H.; Delbrook, C.P.; et al. Long-Term Follow-Up of CD19-CAR T-Cell Therapy in Children and Young Adults With B-ALL. J. Clin. Oncol. 2021, 39, 1650–1659. [Google Scholar] [CrossRef] [PubMed]
  144. Gardner, R.A.; Finney, O.; Annesley, C.; Brakke, H.; Summers, C.; Leger, K.; Bleakley, M.; Brown, C.; Mgebroff, S.; Kelly-Spratt, K.S.; et al. Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults. Blood 2017, 129, 3322–3331. [Google Scholar] [CrossRef]
  145. Summers, C.; Annesley, C.; Bleakley, M.; Dahlberg, A.; Jensen, M.C.; Gardner, R. Long Term Follow-up after SCRI-CAR19v1 Reveals Late Recurrences as Well as a Survival Advantage to Consolidation with HCT after CAR T Cell Induced Remission. Blood 2018, 132, 967. [Google Scholar] [CrossRef]
  146. Shah, N.N.; Highfill, S.L.; Shalabi, H.; Yates, B.; Jin, J.; Wolters, P.L.; Ombrello, A.; Steinberg, S.M.; Martin, S.; Delbrook, C.; et al. CD4/CD8 T-Cell Selection Affects Chimeric Antigen Receptor (CAR) T-Cell Potency and Toxicity: Updated Results from a Phase I Anti-CD22 CAR T-Cell Trial. J. Clin. Oncol. 2020, 38, 1938–1950. [Google Scholar] [CrossRef] [PubMed]
  147. Pulsipher, M.A.; Han, X.; Quigley, M.; Kari, G.; Rives, S.; Laetsch, T.W.; Myers, G.D.; Hiramatsu, H.; Yanik, G.A.; Qayed, M.; et al. Molecular Detection of Minimal Residual Disease Precedes Morphological Relapse and Could be Used to Identify Relapse in Pediatric and Young Adult B-Cell Acute Lymphoblastic Leukemia Patients Treated with Tisagenlecleucel. Blood 2018, 132, 1551. [Google Scholar] [CrossRef]
  148. Gamis, A.S.; Alonzo, T.A.; Meshinchi, S.; Sung, L.; Gerbing, R.B.; Raimondi, S.C.; Hirsch, B.A.; Kahwash, S.B.; Heerema-McKenney, A.; Winter, L.; et al. Gemtuzumab ozogamicin in children and adolescents with de novo acute myeloid leukemia improves event-free survival by reducing relapse risk: Results from the randomized phase III Children’s Oncology Group trial AAML0531. J. Clin. Oncol. 2014, 32, 3021–3032. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Lamba, J.K.; Chauhan, L.; Shin, M.; Loken, M.R.; Pollard, J.A.; Wang, Y.C.; Ries, R.E.; Aplenc, R.; Hirsch, B.A.; Raimondi, S.C.; et al. CD33 Splicing Polymorphism Determines Gemtuzumab Ozogamicin Response in De Novo Acute Myeloid Leukemia: Report from Randomized Phase III Children’s Oncology Group Trial AAML0531. J. Clin. Oncol. 2017, 35, 2674–2682. [Google Scholar] [CrossRef] [PubMed]
  150. Pollard, J.A.; Loken, M.; Gerbing, R.B.; Raimondi, S.C.; Hirsch, B.A.; Aplenc, R.; Bernstein, I.D.; Gamis, A.S.; Alonzo, T.A.; Meshinchi, S. CD33 Expression and Its Association with Gemtuzumab Ozogamicin Response: Results From the Randomized Phase III Children’s Oncology Group Trial AAML0531. J. Clin. Oncol. 2016, 34, 747–755. [Google Scholar] [CrossRef]
  151. Tarlock, K.; Alonzo, T.A.; Gerbing, R.B.; Raimondi, S.C.; Hirsch, B.A.; Sung, L.; Pollard, J.A.; Aplenc, R.; Loken, M.R.; Gamis, A.S.; et al. Gemtuzumab Ozogamicin Reduces Relapse Risk in FLT3/ITD Acute Myeloid Leukemia: A Report from the Children’s Oncology Group. Clin. Cancer Res. 2016, 22, 1951–1957. [Google Scholar] [CrossRef] [Green Version]
  152. Pollard, J.A.; Guest, E.; Alonzo, T.A.; Gerbing, R.B.; Loken, M.R.; Brodersen, L.E.; Kolb, E.A.; Aplenc, R.; Meshinchi, S.; Raimondi, S.C.; et al. Gemtuzumab Ozogamicin Improves Event-Free Survival and Reduces Relapse in Pediatric KMT2A-Rearranged AML: Results from the Phase III Children’s Oncology Group Trial AAML0531. J. Clin. Oncol. 2021, JCO2003048. [Google Scholar] [CrossRef]
  153. Rafiee, R.; Chauhan, L.; Alonzo, T.A.; Wang, Y.C.; Elmasry, A.; Loken, M.R.; Pollard, J.; Aplenc, R.; Raimondi, S.; Hirsch, B.A.; et al. ABCB1 SNP predicts outcome in patients with acute myeloid leukemia treated with Gemtuzumab ozogamicin: A report from Children’s Oncology Group AAML0531 Trial. Blood Cancer J. 2019, 9, 51. [Google Scholar] [CrossRef] [PubMed]
  154. Daver, N.; Alotaibi, A.S.; Bucklein, V.; Subklewe, M. T-cell-based immunotherapy of acute myeloid leukemia: Current concepts and future developments. Leukemia 2021, 35, 1843–1863. [Google Scholar] [CrossRef]
  155. Lamble, A.J.; Tasian, S.K. Opportunities for immunotherapy in childhood acute myeloid leukemia. Blood Adv. 2019, 3, 3750–3758. [Google Scholar] [CrossRef]
  156. Vogiatzi, F.; Winterberg, D.; Lenk, L.; Buchmann, S.; Cario, G.; Schrappe, M.; Peipp, M.; Richter-Pechanska, P.; Kulozik, A.E.; Lentes, J.; et al. Daratumumab eradicates minimal residual disease in a preclinical model of pediatric T-cell acute lymphoblastic leukemia. Blood 2019, 134, 713–716. [Google Scholar] [CrossRef] [PubMed]
  157. Ofran, Y.; Ringelstein-Harlev, S.; Slouzkey, I.; Zuckerman, T.; Yehudai-Ofir, D.; Henig, I.; Beyar-Katz, O.; Hayun, M.; Frisch, A. Daratumumab for eradication of minimal residual disease in high-risk advanced relapse of T-cell/CD19/CD22-negative acute lymphoblastic leukemia. Leukemia 2020, 34, 293–295. [Google Scholar] [CrossRef] [PubMed]
  158. Lamble, A.J.; Gardner, R. CAR T cells for other pediatric non-B-cell hematologic malignancies. Hematol. Am. Soc. Hematol. Educ. Program 2020, 2020, 494–500. [Google Scholar] [CrossRef]
  159. Gomes-Silva, D.; Srinivasan, M.; Sharma, S.; Lee, C.M.; Wagner, D.L.; Davis, T.H.; Rouce, R.H.; Bao, G.; Brenner, M.K.; Mamonkin, M. CD7-edited T cells expressing a CD7-specific CAR for the therapy of T-cell malignancies. Blood 2017, 130, 285–296. [Google Scholar] [CrossRef]
  160. Mamonkin, M.; Rouce, R.H.; Tashiro, H.; Brenner, M.K. A T-cell-directed chimeric antigen receptor for the selective treatment of T-cell malignancies. Blood 2015, 126, 983–992. [Google Scholar] [CrossRef] [Green Version]
  161. Sanchez-Martinez, D.; Baroni, M.L.; Gutierrez-Aguera, F.; Roca-Ho, H.; Blanch-Lombarte, O.; Gonzalez-Garcia, S.; Torrebadell, M.; Junca, J.; Ramirez-Orellana, M.; Velasco-Hernandez, T.; et al. Fratricide-resistant CD1a-specific CAR T cells for the treatment of cortical T-cell acute lymphoblastic leukemia. Blood 2019, 133, 2291–2304. [Google Scholar] [CrossRef] [PubMed]
  162. Georgiadis, C.; Rasaiyaah, J.; Gkazi, S.A.; Preece, R.; Etuk, A.; Christi, A.; Qasim, W. Base-edited CAR T cells for combinational therapy against T cell malignancies. Leukemia 2021. [Google Scholar] [CrossRef] [PubMed]
  163. Cooper, M.L.; Choi, J.; Staser, K.; Ritchey, J.K.; Devenport, J.M.; Eckardt, K.; Rettig, M.P.; Wang, B.; Eissenberg, L.G.; Ghobadi, A.; et al. An “off-the-shelf” fratricide-resistant CAR-T for the treatment of T cell hematologic malignancies. Leukemia 2018, 32, 1970–1983. [Google Scholar] [CrossRef] [PubMed]
  164. Bader, P.; Kreyenberg, H.; von Stackelberg, A.; Eckert, C.; Salzmann-Manrique, E.; Meisel, R.; Poetschger, U.; Stachel, D.; Schrappe, M.; Alten, J.; et al. Monitoring of minimal residual disease after allogeneic stem-cell transplantation in relapsed childhood acute lymphoblastic leukemia allows for the identification of impending relapse: Results of the ALL-BFM-SCT 2003 trial. J. Clin. Oncol. 2015, 33, 1275–1284. [Google Scholar] [CrossRef]
  165. Balduzzi, A.; Di Maio, L.; Silvestri, D.; Songia, S.; Bonanomi, S.; Rovelli, A.; Conter, V.; Biondi, A.; Cazzaniga, G.; Valsecchi, M.G. Minimal residual disease before and after transplantation for childhood acute lymphoblastic leukaemia: Is there any room for intervention? Br. J. Haematol. 2014, 164, 396–408. [Google Scholar] [CrossRef] [PubMed]
  166. Pulsipher, M.A.; Langholz, B.; Wall, D.A.; Schultz, K.R.; Bunin, N.; Carroll, W.; Raetz, E.; Gardner, S.; Goyal, R.K.; Gastier-Foster, J.; et al. Risk factors and timing of relapse after allogeneic transplantation in pediatric ALL: For whom and when should interventions be tested? Bone Marrow Transpl. 2015, 50, 1173–1179. [Google Scholar] [CrossRef]
  167. Zecca, M.; Prete, A.; Rondelli, R.; Lanino, E.; Balduzzi, A.; Messina, C.; Fagioli, F.; Porta, F.; Favre, C.; Pession, A.; et al. Chronic graft-versus-host disease in children: Incidence, risk factors, and impact on outcome. Blood 2002, 100, 1192–1200. [Google Scholar] [CrossRef] [Green Version]
  168. Bader, P.; Kreyenberg, H.; Hoelle, W.; Dueckers, G.; Handgretinger, R.; Lang, P.; Kremens, B.; Dilloo, D.; Sykora, K.W.; Schrappe, M.; et al. Increasing mixed chimerism is an important prognostic factor for unfavorable outcome in children with acute lymphoblastic leukemia after allogeneic stem-cell transplantation: Possible role for pre-emptive immunotherapy? J. Clin. Oncol. 2004, 22, 1696–1705. [Google Scholar] [CrossRef]
  169. Horn, B.; Petrovic, A.; Wahlstrom, J.; Dvorak, C.C.; Kong, D.; Hwang, J.; Expose-Spencer, J.; Gates, M.; Cowan, M.J. Chimerism-based pre-emptive immunotherapy with fast withdrawal of immunosuppression and donor lymphocyte infusions after allogeneic stem cell transplantation for pediatric hematologic malignancies. Biol. Blood Marrow Transpl. 2015, 21, 729–737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Rettinger, E.; Merker, M.; Salzmann-Manrique, E.; Kreyenberg, H.; Krenn, T.; Durken, M.; Faber, J.; Huenecke, S.; Cappel, C.; Bremm, M.; et al. Pre-Emptive Immunotherapy for Clearance of Molecular Disease in Childhood Acute Lymphoblastic Leukemia after Transplantation. Biol. Blood Marrow Transpl. 2017, 23, 87–95. [Google Scholar] [CrossRef] [Green Version]
  171. Locatelli, F.; Zecca, M.; Rondelli, R.; Bonetti, F.; Dini, G.; Prete, A.; Messina, C.; Uderzo, C.; Ripaldi, M.; Porta, F.; et al. Graft versus host disease prophylaxis with low-dose cyclosporine-A reduces the risk of relapse in children with acute leukemia given HLA-identical sibling bone marrow transplantation: Results of a randomized trial. Blood 2000, 95, 1572–1579. [Google Scholar] [CrossRef]
  172. Abraham, R.; Szer, J.; Bardy, P.; Grigg, A. Early cyclosporine taper in high-risk sibling allogeneic bone marrow transplants. Bone Marrow Transpl. 1997, 20, 773–777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Gandemer, V.; Pochon, C.; Oger, E.; Dalle, J.H.; Michel, G.; Schmitt, C.; de Berranger, E.; Galambrun, C.; Cave, H.; Cayuela, J.M.; et al. Clinical value of pre-transplant minimal residual disease in childhood lymphoblastic leukaemia: The results of the French minimal residual disease-guided protocol. Br. J. Haematol. 2014, 165, 392–401. [Google Scholar] [CrossRef] [PubMed]
  174. Mehta, J.; Powles, R.; Singhal, S.; Tait, D.; Swansbury, J.; Treleaven, J. Cytokine-mediated immunotherapy with or without donor leukocytes for poor-risk acute myeloid leukemia relapsing after allogeneic bone marrow transplantation. Bone Marrow Transpl. 1995, 16, 133–137. [Google Scholar]
  175. Rettinger, E.; Huenecke, S.; Bonig, H.; Merker, M.; Jarisch, A.; Soerensen, J.; Willasch, A.; Bug, G.; Schulz, A.; Klingebiel, T.; et al. Interleukin-15-activated cytokine-induced killer cells may sustain remission in leukemia patients after allogeneic stem cell transplantation: Feasibility, safety and first insights on efficacy. Haematologica 2016, 101, e153–e156. [Google Scholar] [CrossRef] [Green Version]
  176. Carpenter, P.A.; Snyder, D.S.; Flowers, M.E.; Sanders, J.E.; Gooley, T.A.; Martin, P.J.; Appelbaum, F.R.; Radich, J.P. Prophylactic administration of imatinib after hematopoietic cell transplantation for high-risk Philadelphia chromosome-positive leukemia. Blood 2007, 109, 2791–2793. [Google Scholar] [CrossRef] [Green Version]
  177. Tarlock, K.; Chang, B.; Cooper, T.; Gross, T.; Gupta, S.; Neudorf, S.; Adlard, K.; Ho, P.A.; McGoldrick, S.; Watt, T.; et al. Sorafenib treatment following hematopoietic stem cell transplant in pediatric FLT3/ITD acute myeloid leukemia. Pediatr. Blood Cancer 2015, 62, 1048–1054. [Google Scholar] [CrossRef]
  178. Gagelmann, N.; Wolschke, C.; Klyuchnikov, E.; Christopeit, M.; Ayuk, F.; Kroger, N. TKI Maintenance After Stem-Cell Transplantation for FLT3-ITD Positive Acute Myeloid Leukemia: A Systematic Review and Meta-Analysis. Front. Immunol 2021, 12, 630429. [Google Scholar] [CrossRef] [PubMed]
  179. Schroeder, T.; Rachlis, E.; Bug, G.; Stelljes, M.; Klein, S.; Steckel, N.K.; Wolf, D.; Ringhoffer, M.; Czibere, A.; Nachtkamp, K.; et al. Treatment of acute myeloid leukemia or myelodysplastic syndrome relapse after allogeneic stem cell transplantation with azacitidine and donor lymphocyte infusions—A retrospective multicenter analysis from the German Cooperative Transplant Study Group. Biol. Blood Marrow Transpl. 2015, 21, 653–660. [Google Scholar] [CrossRef] [Green Version]
  180. Barrett, A.J.; Horowitz, M.M.; Pollock, B.H.; Zhang, M.J.; Bortin, M.M.; Buchanan, G.R.; Camitta, B.M.; Ochs, J.; Graham-Pole, J.; Rowlings, P.A.; et al. Bone marrow transplants from HLA-identical siblings as compared with chemotherapy for children with acute lymphoblastic leukemia in a second remission. N. Engl. J. Med. 1994, 331, 1253–1258. [Google Scholar] [CrossRef] [PubMed]
  181. Horan, J.T.; Alonzo, T.A.; Lyman, G.H.; Gerbing, R.B.; Lange, B.J.; Ravindranath, Y.; Becton, D.; Smith, F.O.; Woods, W.G.; Children’s Oncology, G. Impact of disease risk on efficacy of matched related bone marrow transplantation for pediatric acute myeloid leukemia: The Children’s Oncology Group. J. Clin. Oncol. 2008, 26, 5797–5801. [Google Scholar] [CrossRef] [Green Version]
  182. Locatelli, F.; Zecca, M.; Messina, C.; Rondelli, R.; Lanino, E.; Sacchi, N.; Uderzo, C.; Fagioli, F.; Conter, V.; Bonetti, F.; et al. Improvement over time in outcome for children with acute lymphoblastic leukemia in second remission given hematopoietic stem cell transplantation from unrelated donors. Leukemia 2002, 16, 2228–2237. [Google Scholar] [CrossRef] [Green Version]
  183. Dini, G.; Zecca, M.; Balduzzi, A.; Messina, C.; Masetti, R.; Fagioli, F.; Favre, C.; Rabusin, M.; Porta, F.; Biral, E.; et al. No difference in outcome between children and adolescents transplanted for acute lymphoblastic leukemia in second remission. Blood 2011, 118, 6683–6690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Fagioli, F.; Quarello, P.; Zecca, M.; Lanino, E.; Rognoni, C.; Balduzzi, A.; Messina, C.; Favre, C.; Foa, R.; Ripaldi, M.; et al. Hematopoietic stem cell transplantation for children with high-risk acute lymphoblastic leukemia in first complete remission: A report from the AIEOP registry. Haematologica 2013, 98, 1273–1281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Saarinen-Pihkala, U.M.; Gustafsson, G.; Ringden, O.; Heilmann, C.; Glomstein, A.; Lonnerholm, G.; Abrahamsson, J.; Bekassy, A.N.; Schroeder, H.; Mellander, L.; et al. No disadvantage in outcome of using matched unrelated donors as compared with matched sibling donors for bone marrow transplantation in children with acute lymphoblastic leukemia in second remission. J. Clin. Oncol. 2001, 19, 3406–3414. [Google Scholar] [CrossRef] [PubMed]
  186. Shaw, P.J.; Kan, F.; Woo Ahn, K.; Spellman, S.R.; Aljurf, M.; Ayas, M.; Burke, M.; Cairo, M.S.; Chen, A.R.; Davies, S.M.; et al. Outcomes of pediatric bone marrow transplantation for leukemia and myelodysplasia using matched sibling, mismatched related, or matched unrelated donors. Blood 2010, 116, 4007–4015. [Google Scholar] [CrossRef] [PubMed]
  187. Leung, W.; Campana, D.; Yang, J.; Pei, D.; Coustan-Smith, E.; Gan, K.; Rubnitz, J.E.; Sandlund, J.T.; Ribeiro, R.C.; Srinivasan, A.; et al. High success rate of hematopoietic cell transplantation regardless of donor source in children with very high-risk leukemia. Blood 2011, 118, 223–230. [Google Scholar] [CrossRef]
  188. Dalle, J.H.; Balduzzi, A.; Bader, P.; Lankester, A.; Yaniv, I.; Wachowiak, J.; Pieczonka, A.; Bierings, M.; Yesilipek, A.; Sedlacek, P.; et al. Allogeneic Stem Cell Transplantation from HLA-Mismatched Donors for Pediatric Patients with Acute Lymphoblastic Leukemia Treated According to the 2003 BFM and 2007 International BFM Studies: Impact of Disease Risk on Outcomes. Biol. Blood Marrow Transpl. 2018, 24, 1848–1855. [Google Scholar] [CrossRef] [Green Version]
  189. Rocha, V.; Cornish, J.; Sievers, E.L.; Filipovich, A.; Locatelli, F.; Peters, C.; Remberger, M.; Michel, G.; Arcese, W.; Dallorso, S.; et al. Comparison of outcomes of unrelated bone marrow and umbilical cord blood transplants in children with acute leukemia. Blood 2001, 97, 2962–2971. [Google Scholar] [CrossRef]
  190. Eapen, M.; Rubinstein, P.; Zhang, M.J.; Stevens, C.; Kurtzberg, J.; Scaradavou, A.; Loberiza, F.R.; Champlin, R.E.; Klein, J.P.; Horowitz, M.M.; et al. Outcomes of transplantation of unrelated donor umbilical cord blood and bone marrow in children with acute leukaemia: A comparison study. Lancet 2007, 369, 1947–1954. [Google Scholar] [CrossRef]
  191. Ruggeri, A.; Michel, G.; Dalle, J.H.; Caniglia, M.; Locatelli, F.; Campos, A.; de Heredia, C.D.; Mohty, M.; Hurtado, J.M.; Bierings, M.; et al. Impact of pretransplant minimal residual disease after cord blood transplantation for childhood acute lymphoblastic leukemia in remission: An Eurocord, PDWP-EBMT analysis. Leukemia 2012, 26, 2455–2461. [Google Scholar] [CrossRef] [Green Version]
  192. Michel, G.; Rocha, V.; Chevret, S.; Arcese, W.; Chan, K.W.; Filipovich, A.; Takahashi, T.A.; Vowels, M.; Ortega, J.; Bordigoni, P.; et al. Unrelated cord blood transplantation for childhood acute myeloid leukemia: A Eurocord Group analysis. Blood 2003, 102, 4290–4297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Eapen, M.; Klein, J.P.; Sanz, G.F.; Spellman, S.; Ruggeri, A.; Anasetti, C.; Brown, M.; Champlin, R.E.; Garcia-Lopez, J.; Hattersely, G.; et al. Effect of donor-recipient HLA matching at HLA A, B, C, and DRB1 on outcomes after umbilical-cord blood transplantation for leukaemia and myelodysplastic syndrome: A retrospective analysis. Lancet Oncol. 2011, 12, 1214–1221. [Google Scholar] [CrossRef] [Green Version]
  194. Eapen, M.; Klein, J.P.; Ruggeri, A.; Spellman, S.; Lee, S.J.; Anasetti, C.; Arcese, W.; Barker, J.N.; Baxter-Lowe, L.A.; Brown, M.; et al. Impact of allele-level HLA matching on outcomes after myeloablative single unit umbilical cord blood transplantation for hematologic malignancy. Blood 2014, 123, 133–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Wagner, J.E., Jr.; Eapen, M.; Carter, S.; Wang, Y.; Schultz, K.R.; Wall, D.A.; Bunin, N.; Delaney, C.; Haut, P.; Margolis, D.; et al. One-unit versus two-unit cord-blood transplantation for hematologic cancers. N. Engl. J. Med. 2014, 371, 1685–1694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Michel, G.; Galambrun, C.; Sirvent, A.; Pochon, C.; Bruno, B.; Jubert, C.; Loundou, A.; Yakoub-Agha, I.; Milpied, N.; Lutz, P.; et al. Single- vs. double-unit cord blood transplantation for children and young adults with acute leukemia or myelodysplastic syndrome. Blood 2016, 127, 3450–3457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Milano, F.; Gooley, T.; Wood, B.; Woolfrey, A.; Flowers, M.E.; Doney, K.; Witherspoon, R.; Mielcarek, M.; Deeg, J.H.; Sorror, M.; et al. Cord-Blood Transplantation in Patients with Minimal Residual Disease. N. Engl. J. Med. 2016, 375, 944–953. [Google Scholar] [CrossRef] [PubMed]
  198. Balligand, L.; Galambrun, C.; Sirvent, A.; Roux, C.; Pochon, C.; Bruno, B.; Jubert, C.; Loundou, A.; Esmiol, S.; Yakoub-Agha, I.; et al. Single-Unit versus Double-Unit Umbilical Cord Blood Transplantation in Children and Young Adults with Residual Leukemic Disease. Biol. Blood Marrow Transpl. 2019, 25, 734–742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Algeri, M.; Gaspari, S.; Locatelli, F. Cord blood transplantation for acute leukemia. Expert Opin. Biol. Ther. 2020, 20, 1223–1236. [Google Scholar] [CrossRef]
  200. Horwitz, M.E.; Wease, S.; Blackwell, B.; Valcarcel, D.; Frassoni, F.; Boelens, J.J.; Nierkens, S.; Jagasia, M.; Wagner, J.E.; Kuball, J.; et al. Phase I/II Study of Stem-Cell Transplantation Using a Single Cord Blood Unit Expanded Ex Vivo with Nicotinamide. J. Clin. Oncol. 2019, 37, 367–374. [Google Scholar] [CrossRef]
  201. Cohen, S.; Roy, J.; Lachance, S.; Delisle, J.S.; Marinier, A.; Busque, L.; Roy, D.C.; Barabe, F.; Ahmad, I.; Bambace, N.; et al. Hematopoietic stem cell transplantation using single UM171-expanded cord blood: A single-arm, phase 1-2 safety and feasibility study. Lancet Haematol. 2020, 7, e134–e145. [Google Scholar] [CrossRef]
  202. Passweg, J.R.; Baldomero, H.; Chabannon, C.; Basak, G.W.; de la Camara, R.; Corbacioglu, S.; Dolstra, H.; Duarte, R.; Glass, B.; Greco, R.; et al. Hematopoietic cell transplantation and cellular therapy survey of the EBMT: Monitoring of activities and trends over 30 years. Bone Marrow Transpl. 2021, 56, 1651–1664. [Google Scholar] [CrossRef]
  203. 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] [Green Version]
  204. Bertaina, A.; Pitisci, A.; Sinibaldi, M.; Algeri, M. T Cell-Depleted and T Cell-Replete HLA-Haploidentical Stem Cell Transplantation for Non-malignant Disorders. Curr. Hematol. Malig. Rep. 2017, 12, 68–78. [Google Scholar] [CrossRef]
  205. Handgretinger, R.; Chen, X.; Pfeiffer, M.; Mueller, I.; Feuchtinger, T.; Hale, G.A.; Lang, P. Feasibility and outcome of reduced-intensity conditioning in haploidentical transplantation. Ann. N. Y. Acad. Sci. 2007, 1106, 279–289. [Google Scholar] [CrossRef]
  206. Lang, P.; Teltschik, H.M.; Feuchtinger, T.; Muller, I.; Pfeiffer, M.; Schumm, M.; Ebinger, M.; Schwarze, C.P.; Gruhn, B.; Schrauder, A.; et al. Transplantation of CD3/CD19 depleted allografts from haploidentical family donors in paediatric leukaemia. Br. J. Haematol. 2014, 165, 688–698. [Google Scholar] [CrossRef]
  207. Bertaina, A.; Merli, P.; Rutella, S.; Pagliara, D.; Bernardo, M.E.; Masetti, R.; Pende, D.; Falco, M.; Handgretinger, R.; Moretta, F.; et al. HLA-haploidentical stem cell transplantation after removal of alphabeta+ T and B cells in children with nonmalignant disorders. Blood 2014, 124, 822–826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Locatelli, F.; Bauquet, A.; Palumbo, G.; Moretta, F.; Bertaina, A. Negative depletion of alpha/beta+ T cells and of CD19+ B lymphocytes: A novel frontier to optimize the effect of innate immunity in HLA-mismatched hematopoietic stem cell transplantation. Immunol. Lett. 2013, 155, 21–23. [Google Scholar] [CrossRef] [PubMed]
  209. Li Pira, G.; Malaspina, D.; Girolami, E.; Biagini, S.; Cicchetti, E.; Conflitti, G.; Broglia, M.; Ceccarelli, S.; Lazzaro, S.; Pagliara, D.; et al. Selective Depletion of alphabeta T Cells and B Cells for Human Leukocyte Antigen-Haploidentical Hematopoietic Stem Cell Transplantation. A Three-Year Follow-Up of Procedure Efficiency. Biol. Blood Marrow Transpl. 2016, 22, 2056–2064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Locatelli, F.; Merli, P.; Pagliara, D.; Li Pira, G.; Falco, M.; Pende, D.; Rondelli, R.; Lucarelli, B.; Brescia, L.P.; Masetti, R.; et al. Outcome of children with acute leukemia given HLA-haploidentical HSCT after alphabeta T-cell and B-cell depletion. Blood 2017, 130, 677–685. [Google Scholar] [CrossRef] [PubMed]
  211. Moretta, L.; Locatelli, F.; Pende, D.; Marcenaro, E.; Mingari, M.C.; Moretta, A. Killer Ig-like receptor-mediated control of natural killer cell alloreactivity in haploidentical hematopoietic stem cell transplantation. Blood 2011, 117, 764–771. [Google Scholar] [CrossRef] [Green Version]
  212. Ciurea, S.O.; Al Malki, M.M.; Kongtim, P.; Fuchs, E.J.; Luznik, L.; Huang, X.J.; Ciceri, F.; Locatelli, F.; Aversa, F.; Castagna, L.; et al. The European Society for Blood and Marrow Transplantation (EBMT) consensus recommendations for donor selection in haploidentical hematopoietic cell transplantation. Bone Marrow Transpl. 2020, 55, 12–24. [Google Scholar] [CrossRef]
  213. Ruggeri, L.; Capanni, M.; Urbani, E.; Perruccio, K.; Shlomchik, W.D.; Tosti, A.; Posati, S.; Rogaia, D.; Frassoni, F.; Aversa, F.; et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 2002, 295, 2097–2100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Pende, D.; Marcenaro, S.; Falco, M.; Martini, S.; Bernardo, M.E.; Montagna, D.; Romeo, E.; Cognet, C.; Martinetti, M.; Maccario, R.; et al. Anti-leukemia activity of alloreactive NK cells in KIR ligand-mismatched haploidentical HSCT for pediatric patients: Evaluation of the functional role of activating KIR and redefinition of inhibitory KIR specificity. Blood 2009, 113, 3119–3129. [Google Scholar] [CrossRef] [Green Version]
  215. Ruggeri, L.; Mancusi, A.; Capanni, M.; Urbani, E.; Carotti, A.; Aloisi, T.; Stern, M.; Pende, D.; Perruccio, K.; Burchielli, E.; et al. Donor natural killer cell allorecognition of missing self in haploidentical hematopoietic transplantation for acute myeloid leukemia: Challenging its predictive value. Blood 2007, 110, 433–440. [Google Scholar] [CrossRef] [Green Version]
  216. Oevermann, L.; Michaelis, S.U.; Mezger, M.; Lang, P.; Toporski, J.; Bertaina, A.; Zecca, M.; Moretta, L.; Locatelli, F.; Handgretinger, R. KIR B haplotype donors confer a reduced risk for relapse after haploidentical transplantation in children with ALL. Blood 2014, 124, 2744–2747. [Google Scholar] [CrossRef] [PubMed]
  217. Locatelli, F.; Merli, P.; Rutella, S. At the Bedside: Innate immunity as an immunotherapy tool for hematological malignancies. J. Leukoc. Biol. 2013, 94, 1141–1157. [Google Scholar] [CrossRef] [PubMed]
  218. Perez-Martinez, A.; Ferreras, C.; Pascual, A.; Gonzalez-Vicent, M.; Alonso, L.; Badell, I.; Fernandez Navarro, J.M.; Regueiro, A.; Plaza, M.; Perez Hurtado, J.M.; et al. Haploidentical transplantation in high-risk pediatric leukemia: A retrospective comparative analysis on behalf of the Spanish working Group for bone marrow transplantation in children (GETMON) and the Spanish Grupo for hematopoietic transplantation (GETH). Am. J. Hematol. 2020, 95, 28–37. [Google Scholar] [CrossRef]
  219. Airoldi, I.; Bertaina, A.; Prigione, I.; Zorzoli, A.; Pagliara, D.; Cocco, C.; Meazza, R.; Loiacono, F.; Lucarelli, B.; Bernardo, M.E.; et al. gammadelta T-cell reconstitution after HLA-haploidentical hematopoietic transplantation depleted of TCR-alphabeta+/CD19+ lymphocytes. Blood 2015, 125, 2349–2358. [Google Scholar] [CrossRef] [PubMed]
  220. Bertaina, A.; Zorzoli, A.; Petretto, A.; Barbarito, G.; Inglese, E.; Merli, P.; Lavarello, C.; Brescia, L.P.; De Angelis, B.; Tripodi, G.; et al. Zoledronic acid boosts gammadelta T-cell activity in children receiving alphabeta(+) T and CD19(+) cell-depleted grafts from an HLA-haplo-identical donor. Oncoimmunology 2017, 6, e1216291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  221. Merli, P.; Algeri, M.; Galaverna, F.; Milano, G.M.; Bertaina, V.; Biagini, S.; Girolami, E.; Palumbo, G.; Sinibaldi, M.; Becilli, M.; et al. Immune Modulation Properties of Zoledronic Acid on TcRgammadelta T-Lymphocytes After TcRalphabeta/CD19-Depleted Haploidentical Stem Cell Transplantation: An analysis on 46 Pediatric Patients Affected by Acute Leukemia. Front. Immunol. 2020, 11, 699. [Google Scholar] [CrossRef]
  222. Lucarelli, B.; Merli, P.; Bertaina, V.; Locatelli, F. Strategies to accelerate immune recovery after allogeneic hematopoietic stem cell transplantation. Expert Rev. Clin. Immunol. 2016, 12, 343–358. [Google Scholar] [CrossRef]
  223. Ciceri, F.; Bonini, C.; Stanghellini, M.T.; Bondanza, A.; Traversari, C.; Salomoni, M.; Turchetto, L.; Colombi, S.; Bernardi, M.; Peccatori, J.; et al. Infusion of suicide-gene-engineered donor lymphocytes after family haploidentical haemopoietic stem-cell transplantation for leukaemia (the TK007 trial): A non-randomised phase I-II study. Lancet Oncol. 2009, 10, 489–500. [Google Scholar] [CrossRef]
  224. Greco, R.; Oliveira, G.; Stanghellini, M.T.; Vago, L.; Bondanza, A.; Peccatori, J.; Cieri, N.; Marktel, S.; Mastaglio, S.; Bordignon, C.; et al. Improving the safety of cell therapy with the TK-suicide gene. Front. Pharm. 2015, 6, 95. [Google Scholar] [CrossRef]
  225. Di Stasi, A.; Tey, S.K.; Dotti, G.; Fujita, Y.; Kennedy-Nasser, A.; Martinez, C.; Straathof, K.; Liu, E.; Durett, A.G.; Grilley, B.; et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N. Engl. J. Med. 2011, 365, 1673–1683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Zhou, X.; Di Stasi, A.; Tey, S.K.; Krance, R.A.; Martinez, C.; Leung, K.S.; Durett, A.G.; Wu, M.F.; Liu, H.; Leen, A.M.; et al. Long-term outcome after haploidentical stem cell transplant and infusion of T cells expressing the inducible caspase 9 safety transgene. Blood 2014, 123, 3895–3905. [Google Scholar] [CrossRef] [Green Version]
  227. Locatelli, F.; Ruggeri, A.; Merli, P.; Naik, S.; Agarwal, R.; Aquino, V.; Jacobsohn, D.A.; Qasim, W.; Nemecek, E.R.; Krishnamurti, L.; et al. Administration of BPX-501 Cells Following Aβ T and B-Cell-Depleted HLA Haploidentical HSCT (haplo-HSCT) in Children with Acute Leukemias. Blood 2018, 132, 307. [Google Scholar] [CrossRef]
  228. Ruggeri, A.; Merli, P.; Algeri, M.; Zecca, M.; Fagioli, F.; Li Pira, G.; Bertaina, V.; Prete, A.; Montanari, M.; Del Bufalo, F.; et al. Comparative Analysis of Alpha-Beta T-Cell and B-Cell Depleted (abTCD) HLA-Haploidentical Hematopoietic Stem Cell Transplantation (haplo-HSCT) Versus Abtcd Haplo-HSCT with T-Cell Add-Back of Rivogenlecleucel Cell [Donor T Cells Transduced with the Inducible Caspase 9 (iC9) Gene Safety Switch] in Children with High-Risk Acute Leukemia (AL) in Remission. Blood 2019, 134, 145. [Google Scholar] [CrossRef]
  229. Dunaikina, M.; Zhekhovtsova, Z.; Shelikhova, L.; Glushkova, S.; Nikolaev, R.; Blagov, S.; Khismatullina, R.; Balashov, D.; Kurnikova, E.; Pershin, D.; et al. Safety and efficacy of the low-dose memory (CD45RA-depleted) donor lymphocyte infusion in recipients of alphabeta T cell-depleted haploidentical grafts: Results of a prospective randomized trial in high-risk childhood leukemia. Bone Marrow Transpl. 2021, 56, 1614–1624. [Google Scholar] [CrossRef]
  230. Anderson, B.E.; McNiff, J.; Yan, J.; Doyle, H.; Mamula, M.; Shlomchik, M.J.; Shlomchik, W.D. Memory CD4+ T cells do not induce graft-versus-host disease. J. Clin. Investig. 2003, 112, 101–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  231. Mamcarz, E.; Madden, R.; Qudeimat, A.; Srinivasan, A.; Talleur, A.; Sharma, A.; Suliman, A.; Maron, G.; Sunkara, A.; Kang, G.; et al. Improved survival rate in T-cell depleted haploidentical hematopoietic cell transplantation over the last 15 years at a single institution. Bone Marrow Transpl. 2020, 55, 929–938. [Google Scholar] [CrossRef]
  232. Luznik, L.; O’Donnell, P.V.; Symons, H.J.; Chen, A.R.; Leffell, M.S.; Zahurak, M.; Gooley, T.A.; Piantadosi, S.; Kaup, M.; Ambinder, R.F.; et al. HLA-haploidentical bone marrow transplantation for hematologic malignancies using nonmyeloablative conditioning and high-dose, posttransplantation cyclophosphamide. Biol. Blood Marrow Transpl. 2008, 14, 641–650. [Google Scholar] [CrossRef] [Green Version]
  233. Bashey, A.; Zhang, M.J.; McCurdy, S.R.; St. Martin, A.; Argall, T.; Anasetti, C.; Ciurea, S.O.; Fasan, O.; Gaballa, S.; Hamadani, M.; et al. Mobilized Peripheral Blood Stem Cells Versus Unstimulated Bone Marrow as a Graft Source for T-Cell-Replete Haploidentical Donor Transplantation Using Post-Transplant Cyclophosphamide. J. Clin. Oncol. 2017, 35, 3002–3009. [Google Scholar] [CrossRef] [PubMed]
  234. Lorentino, F.; Labopin, M.; Bernardi, M.; Ciceri, F.; Socie, G.; Cornelissen, J.J.; Esteve, J.; Ruggeri, A.; Volin, L.; Yacoub-Agha, I.; et al. Comparable outcomes of haploidentical, 10/10 and 9/10 unrelated donor transplantation in adverse karyotype AML in first complete remission. Am. J. Hematol. 2018, 93, 1236–1244. [Google Scholar] [CrossRef] [Green Version]
  235. Saglio, F.; Berger, M.; Spadea, M.; Pessolano, R.; Carraro, F.; Barone, M.; Quarello, P.; Vassallo, E.; Fagioli, F. Haploidentical HSCT with post transplantation cyclophosphamide versus unrelated donor HSCT in pediatric patients affected by acute leukemia. Bone Marrow Transpl. 2021, 56, 586–595. [Google Scholar] [CrossRef] [PubMed]
  236. Symons, H.J.; Zahurak, M.; Cao, Y.; Chen, A.; Cooke, K.; Gamper, C.; Klein, O.; Llosa, N.; Zambidis, E.T.; Ambinder, R.; et al. Myeloablative haploidentical BMT with posttransplant cyclophosphamide for hematologic malignancies in children and adults. Blood Adv. 2020, 4, 3913–3925. [Google Scholar] [CrossRef] [PubMed]
  237. Ruggeri, A.; Galimard, J.E.; Paina, O.; Fagioli, F.; Tbakhi, A.; Yesilipek, A.; Navarro, J.M.F.; Faraci, M.; Hamladji, R.M.; Skorobogatova, E.; et al. Outcomes of Unmanipulated Haploidentical Transplantation Using Post-Transplant Cyclophosphamide (PT-Cy) in Pediatric Patients with Acute Lymphoblastic Leukemia. Transpl. Cell Ther. 2021, 27, 424.e1–424.e9. [Google Scholar] [CrossRef]
  238. 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] [PubMed]
  239. Gragert, L.; Eapen, M.; Williams, E.; Freeman, J.; Spellman, S.; Baitty, R.; Hartzman, R.; Rizzo, J.D.; Horowitz, M.; Confer, D.; et al. HLA match likelihoods for hematopoietic stem-cell grafts in the U.S. registry. N. Engl. J. Med. 2014, 371, 339–348. [Google Scholar] [CrossRef] [Green Version]
  240. Bakhtiar, S.; Salzmann-Manrique, E.; Hutter, M.; Krenn, T.; Duerken, M.; Faber, J.; Reinhard, H.; Kreyenberg, H.; Huenecke, S.; Cappel, C.; et al. AlloHSCT in paediatric ALL and AML in complete remission: Improvement over time impacted by accreditation? Bone Marrow Transpl. 2019, 54, 737–745. [Google Scholar] [CrossRef]
  241. Qayed, M.; Ahn, K.W.; Kitko, C.L.; Johnson, M.H.; Shah, N.N.; Dvorak, C.; Mellgren, K.; Friend, B.D.; Verneris, M.R.; Leung, W.; et al. A validated pediatric disease risk index for allogeneic hematopoietic cell transplantation. Blood 2021, 137, 983–993. [Google Scholar] [CrossRef] [PubMed]
Table 1. HSCT indications in pediatric patients with ALL (adapted from AIEOP-BFM ALL 2017 and IntReALL 2010 protocol).
Table 1. HSCT indications in pediatric patients with ALL (adapted from AIEOP-BFM ALL 2017 and IntReALL 2010 protocol).
CR1TCF3-HLF, irrespective of MRD results
Positive MRD at TP1 or TP2 (irrespective of MRD value) and:
-
No CR at d33
-
KMT2A-AFF1
-
hypodiploidy <44 chr. or DNA index <0.8
-
IKZF1plus and intermediate-/high-risk MRD results
-
T-ALL + poor-prednisone response and/or MFC-MRD d15 >10%
MRD TP2 ≥5 × 10−4
Ph+ positive ALL and MRD ≥5 × 10−4 at end Induction IB
Infants (age < 1 year) with KMT2A-rearrangments and:
-
age < 6 months and initial WBC > 300,000/μL
-
age < 6 months and Prednisone Poor-Response
-
No CR at d33
-
MRD TP2 ≥5 × 10−4
CR2HRVery early isolated extramedullary relapse of BCP or T-ALL
Any bone marrow relapse of T-ALL (irrespectively of the time elapsing between diagnosis and relapse)
Very early BCP-ALL bone marrow relapse
Early BCP-ALL bone marrow relapse
Very early BCP-ALL combined bone marrow relapse
SRLate isolated or combined bone marrow relapse of BCP-ALL and poor MRD response at the end of induction therapy *
Early combined bone marrow relapse and poor MRD response at the end of induction therapy *
Early isolated extramedullary relapse of BCP or T-ALL
>CR3All patients
TCF3-HLF: TCF3-HLF positive acute lymphoblastic leukemia; MRD: minimal residual disease; CR: complete remission; MFC-MRD: flow-citometry MRD; WBC: white blood cells; BCP: B-cell precursor ALL (acute lymphoblastic leukemia). T-ALL: T-cell ALL; TP1: time point 1; TP2: time point 2. Time point of relapse: Very early, <18 months after primary diagnosis; early, ≥18 months after primary diagnosis and <6 after completion of primary; late, ≥6 months after completion of primary therapy. SR: standard risk; HR: high risk. * cut-off is defined by the specific treatment arm.
Table 2. HSCT indications in childhood AML.
Table 2. HSCT indications in childhood AML.
CR1GENETIC RISK CRITERIA
Complex karyotype (≥3 aberrations including at least one structural aberration)
Monosomal karyotype (−7, −5, del 5q)
11q23/KMT2A rearrangements, involving:
-
t(10;11)(p12;q23)/KMT2A-AF10
-
t(10;11)(p11.2;q23)/KMT2A-ABI1
-
t(6;11)(q27;q23)/KMT2A-MLLT4
-
t(4;11)(q21;q23.3)/KMT2A-MLLT2
t(11;12)(p15;p13)/NUP98-KDM5A
t(7;11)(p15.4;p15)/NUP98-HOXA9
t(5;11)(q35;p15)/NUP98-NSD1
t(6;9)(p23;q34)/DEK-NUP214
t(16;21)(q24;q22)/RUNX1-CBFA2T3
t(7;12)(q36;p13)/MNX1-ETV6
t(3;21)(26.2;q22)/RUNX1-MECOM
t(16;21)(p11.2;q22.2)/FUS-ERG
FLT3-ITD with AR ≥0.5 without NPM1 mutations
inv(3)(q21.3q26.2)/t(3;3)(q21.3q26.2)/RPN1-MECOM
inv(16)(p13.3q24.3)/CBFA2T3-GLIS2
12p abnormalities
RESPONSE RISK CRITERIA
MRD ≥ 1% after the first induction course
MRD ≥ 0.1% after the second induction course
Primary Induction Failure [i.e. patients with ≥25% blasts after the first induction course and ≥5% blasts after the second induction course]
SECONDARY AML
Therapy-related AML
AML evolving from myelodysplastic syndrome (MDS)
≥CR2All patients
HSCT: hematopoietic stem cell transplantation; NPM1: nucleophosmin1.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Algeri, M.; Merli, P.; Locatelli, F.; Pagliara, D. The Role of Allogeneic Hematopoietic Stem Cell Transplantation in Pediatric Leukemia. J. Clin. Med. 2021, 10, 3790. https://doi.org/10.3390/jcm10173790

AMA Style

Algeri M, Merli P, Locatelli F, Pagliara D. The Role of Allogeneic Hematopoietic Stem Cell Transplantation in Pediatric Leukemia. Journal of Clinical Medicine. 2021; 10(17):3790. https://doi.org/10.3390/jcm10173790

Chicago/Turabian Style

Algeri, Mattia, Pietro Merli, Franco Locatelli, and Daria Pagliara. 2021. "The Role of Allogeneic Hematopoietic Stem Cell Transplantation in Pediatric Leukemia" Journal of Clinical Medicine 10, no. 17: 3790. https://doi.org/10.3390/jcm10173790

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