Although allogeneic hematopoietic cell transplantation (allo-HCT) is currently the standard curative treatment of acute leukemia, relapse remains unacceptably high. Measurable (minimal) residual disease (MRD) after allo-HCT may be used as a predictor of impending relapse and should be part of routine follow-up for transplanted patients. Patients with MRD may respond to therapies aiming to unleash or enhance the graft-versus-leukemia effect. However, evidence-based recommendations on how to best implement MRD testing and MRD-directed therapy after allo-HCT are lacking. Here, I describe our institutional approach to MRD monitoring for preemptive MRD-triggered intervention, using patient scenarios to illustrate the discussion.

While >30 000 allogeneic hematopoietic cell transplantations (allo-HCTs) are performed annually worldwide, some 30% of the recipients are destined to relapse within 2 years of allo-HCT.1-3  Measurable (previously termed minimal) residual disease (MRD) monitoring in acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) has been introduced into clinical practice as a validated tool for early prediction of subsequent relapse and to improve outcomes by guiding subsequent therapy, including decisions regarding transplantation.4-6  MRD persistence at transplant has been identified as the strongest risk factor for posttransplant relapse,7,8  which may be at least partially overcome by additional intervention such as blinatumomab in B-cell ALL or augmented conditioning in AML.9,10  Still, there is a relative paucity of data regarding MRD and MRD-driven interventions following allo-HCT. MRD after transplant is associated with an increased incidence of relapse, but the clinical implications of MRD kinetics are not yet clearly defined. All the methodological considerations and limitations that complicate MRD testing in the nontransplant setting, such as a lack of leukemia-specific markers, nonstandardized techniques, poorly defined cutoffs and testing intervals, and MRD-target loss due to clonal/molecular evolution, also beleaguer posttransplant MRD monitoring.4  Furthermore, the clinical interpretation of MRD after allo-HCT is even more enigmatic (Table 1). Unlike chemotherapy, which induces an antileukemic effect of short duration, the GVL effect is prolonged, with unique and nonquantifiable dynamics in different individuals, and may require several months to eradicate any persisting tumor cells.11  The strong and protracted pressure of GVL may promote immune evasion in persisting leukemia cells, such as through genomic loss or downregulation of HLA genes, immune-checkpoint ligand overexpression, or generation of an anti-inflammatory milieu in the leukemic microenvironment, resulting in resistance to GVL.12-14  Patients with MRD may respond to discontinuation of immunosuppression and donor lymphocyte infusion (DLI), but convincing evidence that preemptive intervention strategies will improve outcome is lacking (Table 2).

Table 1.

Confounders in MRD monitoring after allo-HCT

MRD monitoringConfounding variables after allo-HCT
Donor chimerism Residual host signals, loss of heterozygosity in the HLA locus 
FC Regenerating hematogones 
Leukemia-specific markers Clearance depends on GVL dynamics, high clonal evolution rate 
DNMT3 Donor-derived clonal hematopoiesis 
FLT3-ITD Unstable, subclonal 
WT1 Overexpression in regenerating marrow 
Ig/TCR Comparable sequences in regenerating T and B cells 
NGS allelic burden High sequencing error rate 
MRD monitoringConfounding variables after allo-HCT
Donor chimerism Residual host signals, loss of heterozygosity in the HLA locus 
FC Regenerating hematogones 
Leukemia-specific markers Clearance depends on GVL dynamics, high clonal evolution rate 
DNMT3 Donor-derived clonal hematopoiesis 
FLT3-ITD Unstable, subclonal 
WT1 Overexpression in regenerating marrow 
Ig/TCR Comparable sequences in regenerating T and B cells 
NGS allelic burden High sequencing error rate 

FC, flow cytometry; GVL, graft-versus-leukemia; Ig/TCR, immunoglobulin and T-cell receptor; NGS, next-generation sequencing; WT1, Wilms tumor 1.

Table 2.

Studies of MRD-directed therapy after allo-HCT

Study typeDisease (n)MRD triggerMRD ITMRD+ with IT, nMRD+ with no IT, nMRD, nMRD response, %REL/DFS/OS in MRD+ with IT, %REL/DFS/OS in MRD+ with no IT, %REL/DFS/OS in MRD, %Reference
SC/retrospective AL, 80 (AML, 36; ALL, 44) WT1, Ig/TCR DLI 17 19 44 NA 6/NA/80 63/NA/26 16/NA/78 85  
SC/prospective AML, 85 WT1 (100 copies) DLI 23 59 96 76/NA/NA 67/NA/NA NA 86  
SC/prospective AML, 122 WT1 (180 copies) DLI 17 21 84 35 29/NA/NA 74/NA/NA NA 86  
SC/prospective AML/MDS, 59 LOC Aza 20 39 80 65/NA/40 NA NA 104  
MC/prospective AML/MDS, 141 LOC, LSM Aza 24 117 71 33*/NA/NA NA NA/86/89 105  
MC/retrospective Relapsed AML/MDS, 154 LOC, LSM Aza + DLI 19 NA NA NA NA/NA/69 NA NA 106  
MC/prospective t(8;21) AML, 92 RUNX1/RUNX1T1 mDLI 17 13 62 NA 24/64/NA 87/0/NA NA 30  
SC/prospective AL, 814 (AML, 529; ALL, 285) WT1, FC mDLI vs IL-2 56 49 709 NA 28/56/58 65/24/28 18/61/66 87  
SC/prospective AL, 129 (AML, 71; ALL, 58) LOC mDLI 24 23 82 NA 37/54/NA 74/22/NA 0/91/NA 43  
SC/retrospective AL, 20 (AML, 15; ALL, 5) WT1, Ig/TCR, LSM Haplo-DLI 20 NA NA 45 55/NA/43 NA NA 93  
Study typeDisease (n)MRD triggerMRD ITMRD+ with IT, nMRD+ with no IT, nMRD, nMRD response, %REL/DFS/OS in MRD+ with IT, %REL/DFS/OS in MRD+ with no IT, %REL/DFS/OS in MRD, %Reference
SC/retrospective AL, 80 (AML, 36; ALL, 44) WT1, Ig/TCR DLI 17 19 44 NA 6/NA/80 63/NA/26 16/NA/78 85  
SC/prospective AML, 85 WT1 (100 copies) DLI 23 59 96 76/NA/NA 67/NA/NA NA 86  
SC/prospective AML, 122 WT1 (180 copies) DLI 17 21 84 35 29/NA/NA 74/NA/NA NA 86  
SC/prospective AML/MDS, 59 LOC Aza 20 39 80 65/NA/40 NA NA 104  
MC/prospective AML/MDS, 141 LOC, LSM Aza 24 117 71 33*/NA/NA NA NA/86/89 105  
MC/retrospective Relapsed AML/MDS, 154 LOC, LSM Aza + DLI 19 NA NA NA NA/NA/69 NA NA 106  
MC/prospective t(8;21) AML, 92 RUNX1/RUNX1T1 mDLI 17 13 62 NA 24/64/NA 87/0/NA NA 30  
SC/prospective AL, 814 (AML, 529; ALL, 285) WT1, FC mDLI vs IL-2 56 49 709 NA 28/56/58 65/24/28 18/61/66 87  
SC/prospective AL, 129 (AML, 71; ALL, 58) LOC mDLI 24 23 82 NA 37/54/NA 74/22/NA 0/91/NA 43  
SC/retrospective AL, 20 (AML, 15; ALL, 5) WT1, Ig/TCR, LSM Haplo-DLI 20 NA NA 45 55/NA/43 NA NA 93  

AL, acute leukemia; Aza, 5-azacytidine; DFS, disease-free survival; haplo-DLI, DLI from a haploidentical donor; IL-2, interleukin-2; IT, immunological therapy; LOC, loss of donor chimerism; LSM, leukemia-specific marker; MC, multicenter; mDLI, modified DLI; MDS, myelodysplastic syndrome; NA, not available (or not specified); OS, overall survival; REL, cumulative incidence of relapse; SC, single center.

*

Follow-up at 13 months.

Follow-up at 12 months.

Low -dose IL-2 group.

How should we monitor MRD in acute leukemia after allo-HCT, when should we react, and how should we intervene? Published workshop guidelines,15-17  expert opinions,18,19  and consensus statements20  differ from evidence-based facts.21  The cases below illustrate our individualized, risk-adapted, but still somewhat intuitive approach in everyday clinical practice (Figure 1).

Figure 1.

How I monitor and treat MRD after allo-HCT. BiTEs, bi-specific T-cell engagers; HDAC, histone deacetylase; HMA, hypomethylating agents.

Figure 1.

How I monitor and treat MRD after allo-HCT. BiTEs, bi-specific T-cell engagers; HDAC, histone deacetylase; HMA, hypomethylating agents.

Close modal

Case 1

A 56-year-old woman with relapsed NPM1mut AML underwent myeloablative conditioning (MAC) allogeneic peripheral blood stem cell transplantation (PBSCT) from an 9/10 HLA-matched unrelated donor in her second complete remission (CR). Pretransplant bone marrow (BM) sampling showed 170 (1.7%) NPM1mut/104ABL1 copies by real-time quantitative polymerase chain reaction (RT-qPCR). Graft-versus-host disease (GVHD) prophylaxis consisted of low-dose alemtuzumab (10 mg) and cyclosporine, which was discontinued at day +122.22  At 3 months after allo-HCT, BM-MRD was positive with 45 NPM1mut/104ABL1 copies while 4-color FC was negative for leukemic blasts (sensitivity 1%). Repeated analyses 4 and 7 weeks after initial testing showed persistent BM-MRD (55 and 97 NPM1mut/104ABL1 copies, respectively). Chimerism evaluation with short tandem repeat (STR)-based PCR (sensitivity 1% to 5%) depicted complete donor chimerism (CC) in both BM and peripheral blood (PB) at all times.

NPM1mut monitoring

Ideally, pretransplant NPM1mut MRD-positive patients should become MRD negative early after transplant,23,24  though one study found no association between early MRD-positivity at day +28 post-allo-HCT and survival.25  In most studies, persistent NPM1mut transcripts after transplant were associated with a statistically significant increased incidence of relapse and NPM1mut-based MRD monitoring preceded other MRD markers in impending relapse.23,24,26,27  Levels >0.1% NPM1mut/ABL1 beyond day +60 post-allo-HCT are translated into increased frequency of relapses, whereas the most powerful independent prognostic cutoff level for reduced survival was 10% NPM1mut/ABL1.25,28  MRD persistence at 3 months post-allo-HCT evaluated by deep targeted sequencing (variant allele frequency [VAF] >0.02%) was significantly and independently associated with increased relapse rates, suggesting that conversion to MRD-negativity by day 100 after allo-HCT is a precondition for achievement of stable remission.23  Since NPM1mut relapses tend to occur late (>6 months) after allo-HCT, our current everyday clinical practice is close MRD monitoring by mutant-specific RT-qPCR (sensitivity 10−4 to 10−5) in BM every 3 months.3  Preemptive interventions are considered for patients with persistent MRD >0.1% in 3 consecutive measurements or MRD >1% when confirmed in a repeated analysis within 4 weeks. MRD >10% (1000 NPM1mut/104ABL1 copies) is an indication for an intermediate intervention, since these patient relapse within a short period of time.28 

Case 2

A 19-year-old male with a previously diagnosed t(8;21) core-binding factor (CBF) AML relapsed with 5567 RUNX1-RUNX1T1/104ABL1 copies in BM and underwent an HLA-matched sibling MAC-PBSCT in CR 2. BM at 3 months revealed 78 RUNX1-RUNX1T1/104ABL1 transcripts, while FC did not detect any leukemic cells. Cyclosporine was discontinued at day +145. BM at day +190 showed MRD persistence at a low level (4.3 RUNX1-RUNX1T1/104ABL1 copies). Chimerism assays of BM and PB indicated CC.

CBF fusion transcript monitoring

Though it is expected that CBF-positive nonleukemia cells should be cleared in the context of allo-HCT, CBF-fusion transcripts at low levels have been found to persist in long-term transplant survivors.29  A >3-log reduction in RUNX1-RUNX1T1 transcripts at any time point and >4-log reduction at 12 months post-allo-HCT are considered safe cutoff-levels for continuous CR. Increase of RUNX1-RUNX1T1 transcripts >1-log predicted relapse despite DLI, whereas patients with ≤1-log increase could be rescued with DLI.30,31  Similar MRD kinetics after transplant have been found in patients with CBFB-MYH11 AML.32,33  We quantify CBF-fusion transcripts by RT-qPCR in BM at 3 months posttransplant and adjust further MRD-monitoring intervals according to the MRD level detected. Preemptive interventions are considered for patients with persistent MRD >1% RUNX1RUNX1T1 or CBFB-MYH11/ABL1 in 2 consecutive measurements or with >0.5-log increase of the CBF fusions in repeated analyses.

Case 3

A 21-year-old female patient with a previous history of Ewing sarcoma at age of 17 developed a therapy-related t(11;19)(q23;p13.1) AML. She underwent a MAC-BM transplantation from her 4/8 HLA-matched haploidentical mother with posttransplant cyclophosphamide (PTCY). KMT2A (previously termed mixed lineage leukemia [MLL])-ELL fusion transcripts in BM pretransplant evaluated by nested PCR (sensitivity 10−4) were undetectable. BM-MRD for MLL-ELL transcripts at 3 months was negative but was found positive at routine analysis on day +203 and in a repeated analysis 3 weeks later. Donor chimerism fell to 90% in the MRD-positive BM samples, whereas FC did not detect residual leukemic blasts.

MLL monitoring

Liu et al found that any MLL expression (>0.0%) after allo-HCT was highly predictive for relapse in multivariate analysis (>90% patients relapsed, hazard ratio [HR], 18.643).34  Most MRD positivity emerged at 3 to 5 months posttransplant, and the median time to relapse after MLL detection was 109 days. In contrast, <10% of the patients with undetectable MLL fusion transcripts relapsed. We routinely screen BM every 3 months for patient-specific MLL rearrangements by nested PCR (sensitivity 10−4), and any MRD positivity confirmed by repeated analysis is considered as a trigger for a preemptive intervention.

Case 4

A 37-year-old man with a FLT3-ITDmut AML achieved CR after 2 cycles of induction chemotherapy and received 2 additional consolidation cycles combined with midostaurin. He underwent an HLA-matched sibling allogeneic MAC-PBSCT with cyclosporine and methotrexate. At that time, the randomized trial of gilteritinib maintenance was not yet recruiting at our institution (NCT02997202) and the patient’s medical insurance rejected coverage for “off-label” sorafenib prophylaxis. Routine chimerism studies by STR-based PCR at day +37 depicted CC in BM, PB, and circulating T cells (> 95%). However, PB donor chimerism progressively decreased to 70% (day +130), despite fast weaning of cyclosporine from day +70 onwards. BM sampling on day +95 and day +130 showed 80% and 70% donor chimerism in the mononuclear fractions and 70% in the magnetic-bead–isolated CD34+ cell subset. BM-FC was negative for leukemic blasts.

FLT3-ITDmut monitoring

FLT3-ITDmut is a poor MRD marker, as this mutation is typically subclonal.35  A recently described NGS-based FLT3-ITDmut MRD assay is currently being evaluated in its ability to predict relapse after allo-HCT in the gilteritinib randomized trial mentioned above.36 

Case 5

A 64-year-old male with a MDS with excess of blasts (18%), complex karyotype, and mutated DNMT3 gene received 4 cycles of 5-azacytidine and underwent a reduced-toxicity conditioning (fludarabine, carmustine, and melphalan) PBSCT from an 23-year-old unrelated 10/10 HLA-matched donor.37  At transplant, 4-color FC showed persistence of 4% CD34+/CD117+/HLA-DR+ blasts with aberrant expression of CD7+. Routine BM evaluation at day+32 and day+105 showed CC (>95%). PB T-cell chimerism was constantly mixed (80% to 90% donor). Cyclosporine was discontinued at day +158. Progressive thrombocytopenia occurred after day +230, in parallel with new-onset PB mixed chimerism (70% donor). The loss of donor chimerism was confirmed in subsequent analyses in BM and PB mononuclear and polymorphonuclear fractions. FC in BM detected a 1.1% CD34+/CD117+ but CD7 blast population.

Chimerism monitoring

Chimerism analysis detects host-derived genetic material that cannot be equated with persistence of leukemia cells. Low-level host DNA (<1%) can be found for a longer period after transplant (eg, due to recipient stromal cells),38  bringing into question the clinical utility of the highly sensitive variant-allele–specific RT-qPCR–based or droplet digital PCR (ddPCR) techniques (sensitivity 0.1% to 0.01%) as compared with the standard STR-based PCR (sensitivity 1% to 5% according to microsatellite marker).39-42  Fluctuating low-level mixed chimerism in PB at the early posttransplant phase may be due to viral infections and expansion of residual, recipient-derived, virus-specific T cells.41  Chimerism patterns, kinetics, and clinical interpretation are strongly related to the transplant practice (eg, MAC vs reduced-intensity conditioning [RIC], T-cell depletion vs none) and to the underlying disease (eg, malignant vs nonmalignant).43-45  Lineage-specific chimerism analysis may increase its specificity in predicting relapse.46,47  Mixed chimerism in T cells is common after T-cell–depleted or RIC transplants and is weakly associated with later disease relapse.45  A prospective study showed that the decrease of CD34+-specific donor chimerism to <80% had 100% sensitivity and 86% accuracy in predicting relapse. This contrasts with the 14% sensitivity and 46% accuracy of conventional chimerism testing.48  As a general rule, the speed and extent of the decrease of donor chimerism predicts relapse with a higher specificity than a static approach considering chimerism levels only at individual time points.41  We routinely monitor chimerism by STR-PCR in PB every 2 weeks within the first 3 months and at least monthly thereafter. We additionally check monthly chimerism in ficoll-isolated mononuclear and polymorphonuclear fractions and in T cells isolated by magnetic beads.39  BM-chimerism analysis is routinely performed on days +30, +100, +270, and +365. A continuous decrease of donor chimerism in the non–T-cell compartment or drop of donor chimerism to <80% in the magnetic-bead–isolated CD34+ subsets raises suspicion for impending relapse. Our decision making for a preemptive intervention encompasses other MRD markers and clinical parameters (eg, pretransplant risk assessment, new-onset cytopenia).

FC monitoring

FC is hampered by its low sensitivity (10−2 to 10−4) and the many factors that can cause false-negative results, such as number of events analyzed, sample processing, hemodilution, and immunophenotypic switch.4  Data analysis and interpretation requires considerable expertise and experience to differentiate “leukemia-associated immunophenotype” or “different-from-normal” populations from regenerating BM cells.49  In expert hands, FC-MRD can be very specific, as demonstrated by Jacobsohn et al, who sorted the FC-detected suspicious cells and proved that these were of recipient origin and contributed to later morphological relapse.50  By applying a highly sensitive (0.1%) 10-color/1-million-events FC-MRD protocol, Zhou et al found that early (day +30) FC-MRD evaluation after allo-HCT has a very low ability to capture leukemic clones that emerge at later time points.51  The existing studies indicate that a new-onset FC-MRD positivity identifies patients with a high relapse risk.52-56  However, there is considerable variation in the protocols used, which makes it difficult to compare results among different studies or implement the results in one’s own laboratory. Most routine laboratories, including ours, will be able to measure FC-MRD at a level of 1%. We categorize FC-MRD results as “suspicious” or “positive” depending on the detected abnormal blast population.

WT1 monitoring

WT1 is a nearly universal leukemia antigen that can be measured in PB but is also overexpressed in normal regenerating BM cells. Patients who do not clear their pretransplant high BM-WT1 transcripts (>250 copies) at 3 months post-allo-HCT or who show a continuous increase of PB-WT1 transcripts are at risk for relapse.57,58  Patients with sustained low WT1 levels after allo-HCT (BM <100, PB <50 copies) had excellent outcomes.58-60  We have recently commenced PB-WT1 testing using a commercially available European Leukemia Net–certified RT-qPCR kit as part of developing release criteria for administration of WT1-specific T cells within a forthcoming multicenter study.61,62 

DNMT3mut monitoring

The investigation of DTA mutations (ie, mutations in DNMT3, TET2, and ASXL1) after transplant may confront us with the engraftment of donor clonal hematopoiesis of indeterminate potential (CHIP).63 DNMT3-mutated host-derived CHIP is typically cleared (VAF <0.2%) at 3 months post-allo-HCT.64  Nakamura et al followed DNMT3 mutations (and other putative founder mutations) using a very sensitive (0.04%) ddPCR and found that increasing MRD positivity between 1 and 3 months post-allo-HCT was the most precise independent predictor of relapse (HR, 28.47; P < .0001).65  Of note, the DNMT3 mutations were found in the relapsed samples. Thus, in contrast to the nontransplant setting, where the persistence of DNMT3 mutations indicates remaining CHIP with no prognostic value,66  the above-mentioned study associates “nondonor” DNMT3 mutations posttransplant with the persistence of transformed DNMT3mut leukemic clones and increased relapse risk.65  The results from a similar prospective study are awaited to clarify this observation (NCT02872662). Another interesting finding of the Nakamura et al study is that the predictive utility of “noninvasive” MRD monitoring using serum DNA was comparable to that of MRD testing in BM.65 

NGS-based monitoring

A persistent VAF >0.2% mutational burden in BM at day +21 is associated with an increased relapse risk (HR, 4.75).64  However, in nearly half of the patients, the day +21–detected mutations were stepwise cleared by 6 months (potentially mediated by GVL), and these patients did not relapse. Thus, the positive predictive ability for relapse of early NGS-MRD remains poor and is also hampered by the high sequencing error rate. Interestingly, mutations detected by NGS at day +21 expanded at relapse in relapsed patients.64 

Case 6

A 34-year-old man with relapsed Philadelphia-chromosome–negative CD19+/CD10+ B- cell ALL was referred 16 months after initial diagnosis for a 9/10 HLA-matched unrelated MAC-PBSCT (12-Gy total body irradiation/etoposide) in CR 2. MRD monitoring was performed with Ig/TCR semi-quantitative PCR (sensitivity 10−4). At transplant, MRD was positive (10−3 to 10−4). FC in BM at day +41 revealed CD19+/CD10+/CD45dim low-side-scatter cells, well matched with regenerating precursors (hematogones). Ig/TCR-MRD in BM at 3 months was positive (10−3 to 10−4) and confirmed to be positive at the same level 2 and 4 weeks later. BM, PB, and T-cell fractions showed CC.

ALL monitoring

FC-based MRD monitoring in ALL post-allo-HCT is even more troublesome than in AML, since regenerating hematogones and ALL blasts share the same immunophenotype (CD19+/CD10+).67  Furthermore, amplification of comparable sequences in regenerated B/T cells may cause false-positive Ig/TCR results.68  IgH-V(D)J-NGS has better specificity and may also capture apparent clonal evolution.69,70  Early (day +30) Ig/TCR MRD positivity has low accuracy to predict a future relapse.71-73  At later time points, MRD levels >10−4 are associated with increased relapse rates but can be cleared by immunotherapy.71-74  In one series, patients with Ig/TCR MRD >10−3 or those who experienced >1-log MRD increase ultimately relapsed, regardless of immunosuppression discontinuation or DLI.72  Our clinical practice dictates that ALL patients should at least every other month monitored with Ig/TCR PCR (or NGS if feasible). Any MRD positivity beyond day +100 is a trigger for intervention. In case of >1-log increase of MRD or MRD level >10−2, we consider “off-label” blinatumomab or inotuzumab in line with phase 2 ongoing studies (eg, NCT03109093 and NCT04044560).

Withdrawal of immune suppression

The burden of immunosuppression administered peri- and posttransplant is associated with risk of relapse.75,76  Withdrawal of immunosuppression is routinely considered as the first therapeutic maneuver for patients with posttransplant MRD status and without GVHD. Different from chronic myeloid leukemia, where impressive responses have been seen, immunosuppression withdrawal by itself is unlikely to result in a clinically significant benefit.46,77 

DLIs

After withdrawal of immunosuppression, infusion of donor cells represents the standard approach for boosting the GVL effect.78  The efficacy of DLI has been demonstrated by their ability to prevent relapse or to treat overt relapse in some cases.79-82  In NPM1mut AML, DLIs have been shown to generate T cells with specificity against the mutant NPM1 protein.83,84  Published data on preemptive DLI for MRD positivity are limited to retrospective or nonrandomized prospective series in which the DLI-treated patients most likely represent a positively selected cohort (Table 2). MRD-WT1–triggered preemptive DLI could prevent relapse, especially when these were given at low (<100), but not higher (180), WT1 copies.85,86  The largest series of preemptive DLI was reported by a Chinese group, who prospectively monitored 814 patients (AML 70%) using FC- and WT1-based MRD. Administration of modified DLIs (see below) reduced 2.3-fold the relapse risk as compared with MRD-positive patients with no DLI.87  A preventive effect of DLI on relapse has also been suggested when the MRD trigger was chimerism loss or increased RUNX1-RUNX1T1 transcript levels.30,43 

The optimal administration schedule for DLI in the preemptive setting is uncertain. An important safety issue that emerged from studies in which DLIs were given prophylactically (mostly in T-cell–depleted allo-HCT) is that the patient should be free of GVHD for at least 1 month and that the starting dose should be 1-log lower (0.5-1 × 106 CD3+ cells/kg) than the one used in the relapse setting.88  Most groups will give repetitive, dose-escalated (by a 2 to 5-fold increment) unmanipulated donor cells in 4- to 12-week intervals, but data are insufficient for clear recommendation. Future research is needed to determine the dose intensity and total number infusions that are necessary to achieve long-term remission. Similarly, further work is necessary to confirm whether MRD negativity is a sufficient end point or whether additional therapy, potentially including a second transplantation, is needed to secure a cure. As a practical measure, we routinely freeze and use donor cells isolated from the initial granulocyte colony-stimulating factor–mobilized graft, which have been shown to possess similar activity as unstimulated DLI.89  We reevaluate MRD after 2 to 4 cycles of DLI, and we “consolidate” by MRD response with an additional 4 to 8 infusions, provided that the patient is free of GVHD.

Haplo-DLI

Haplo-DLI may pose an increased risk for GVHD due to the high donor-recipient HLA disparity. In T-cell–depleted/CD34+-selected haploidentical allo-HCT, the CD3+ threshold for GVHD has been set at 3 × 104 cells/kg.90  Zalmoxis and ATIR101 are suicide-gene–engineered and alloreactive T-cell–depleted haplo-DLI products, respectively given prophylactically to enhance immune reconstitution.91,92  For preemptive interventions after anti-thymocyte globulin (ATG)-based haploidentical allo-HCT, the Chinese groups use granulocyte colony-stimulating factor–mobilized haplo-DLI followed by short-term immunosuppression (termed modified-DLIs).20  After PTCY-based haploidentical allo-HCT, prophylactically or preemptively administered unmanipulated haplo-DLI with a starting dose of 1 × 104 to 1 × 105 CD3+ cells/kg (ie, 1-2 log lower than from HLA-matched donor) were relative safe.93-96  There are 25 cases of MRD-triggered haplo-DLI after PTCY haploidentical allo-HCT reported so far, of which nearly 50% responded (normalized WT1, negativity of leukemia-specific marker, increase of donor chimerism).93,94  With the increasing use of haploidentical transplants, it must be noticed that nearly one-third of relapses that occur after mismatched related transplants (and nearly 10% of relapses after unrelated allo-HCT) show genomic loss of the mismatched HLA haplotype in which the use of DLIs will probably have no effect.12,97  Thus, documentation of “HLA-loss” at posttransplant relapses has relevant therapeutic implications and can be performed in nonpurified samples by the use of the HLA-KMR qPCR (GenDx, Utrecht, The Netherlands).98 

Pharmacologic intervention with or without DLI after transplantation

Pharmacological intervention for posttransplant MRD aims to mediate a direct antileukemic effect and/or to enhance the alloreactive response. There is convincing evidence that immunomodulators alone cannot facilitate long-term survival, but this can be achieved when they are combined with DLI.81  HMAs (azacitidine and decitabine) may beneficially influence the balance between GVL and GVHD by enhancing the immunological visibility of leukemia cells (eg, through expression of silenced cancer/testis antigens and activation of interferon responses) while mitigating GVHD through expansion of regulatory T cells.99-101  However, it is also possible that HMAs may hamper GVL responses by inhibiting the function of natural killer cells or increasing the frequency of regulatory T cells in the BM.102,103  Two prospective studies in AML and MDS demonstrated that azacitidine post-allo-HCT could safely and effectively treat MRD (conversion of loss of CD34+ donor chimerism, NPM1mut or RUNX1-RUNX1T1 positivity), ultimately leading to prevention of relapse in approximately one-third of patients.104,105  Better results were found when azacitidine was given together with DLIs.106  Currently, a prospective randomized controlled study compares the safety and efficacy of MRD-triggered HMA+DLI versus DLI alone (NCT03662087). Our preemptive protocol for MRD positivity in AML and MDS entails 4 cycles of azacitidine (32-75 mg/m2, days 1-5) followed by a fixed dose of DLI at day +14. MRD responders are scheduled to receive 4 more cycles, and nonresponders receive escalated doses of DLI after each azacitidine cycle.

Interferon-α and interleukin-2 alone or together with DLIs have also been tested as immunomodulators in the MRD preemptive setting, but with doubtful effects and safety concerns.87,107  Lenalidomide given as maintenance therapy after allo-HCT has been associated with severe GVHD, but it can be more safely administered when combined with azacytidine.108,109  Extended azacitidine dosing using an oral formulation of the drug (CC-486) and panobinostat (deacetylase inhibitor) have shown promising results in prophylactic phase 1/2 studies.110,111  Case series reported the efficacy of immune checkpoint inhibitors (anti-CTLA-4 and anti-PD-1/PD-L1) in relapsed disease, but their use is associated with high rates of severe, often life-threatening GVHD and thus the administration of these drugs in the preemptive MRD setting is not justified outside a clinical trial.112,113 

Given the positive results in many retrospective and prospective studies, most FLT3-ITDmut AML patients today will receive maintenance therapy with an FLT3 inhibitor, either off-label or within a clinical trial. Given the possible synergism between FLT3 inhibitors and alloreactive donor T cells, we favor the combination of sorafenib with DLI in patients with FLT3-ITDmut AML needing preemptive therapy.114  The isocitrate dehydrogenase inhibitors (ivosidenib and enasidenib) are currently tested in isocitrate dehydrogenase–mutated AML as maintenance and salvage therapy after allo-HCT.

How did we intervene?

No preemptive intervention was undertaken for patient 2. Further BM aspirations were scheduled on days +270 and +365 after allo-HCT, which revealed 0.08% and 0.00% RUNX1-RUNX1T1/104ABL1 copies, respectively. We administrated DLI in the remaining patients. The starting DLI dose was 1 × 106 CD3+ cells/kg for patients 1, 5, and 6; 0.5 × 106 CD3+ cells/kg (<4 weeks after discontinuation of immunosuppression) for patient 4; and 1 × 104 CD3+ cells/kg (haploidentical donor) for patient 3. We combined DLI with sorafenib in case 4 (FLT3-ITDmut) and with azacitidine in case 5 (MDS). In patient 1 (NPM1mut AML), NPM1mut transcripts were no longer detectable in the BM after 3 DLI cycles. We gave in total 6 escalated DLIs (maximal dose, 5 × 106 CD3+ cells/kg) without complications, and the patient is now 4.5 years after transplantation and in good health. Patient 3 (MLL-ELL+ AML) relapsed after the third haplo-DLI (1 × 105 CD3+ cells/kg) and succumbed to her disease 371 days after transplant. Patient 4 (FLT3-ITDmut AML) received an initial total dose of 800 mg/day sorafenib, but this was reduced to 400 mg/day due to high serum amylase levels. After 3 months of sorafenib combined with DLI, the PB chimerism converted to completely donor. He received in total 6 DLIs and continues on sorafenib at 1.5 years after allo-HCT. Patient 5 (MDS) received in total 8 cycles of azacitidine (75 mg/m2 on days 1-5) followed by a fixed dose of DLI (1 × 106 CD3+ cells/ kg). The PB and BM mixed chimerism switched to CC after the fourth azacitidine/DLI cycle. He is now 17 months after the start of preemptive therapy with a good performance status. In patient 6 (ALL), the BM-MRD assessment was negative after the fourth DLI given at 5 × 106 CD3+ cells/kg. After the fifth DLI (1 × 107 CD3+ cells/kg), the patient developed a biopsy-proven liver GVHD (hepatitic variant), which was resolved after treatment with steroids and budesonide. The patient is now 3.2 years after transplantation with a good performance status. Preemptive MRD-triggered intervention, although not effective, was probably a correct decision in case 3. In cases 1, 4, 5, and 6, we cannot be sure whether we “overreacted.”

Dynamic MRD monitoring after allo-HCT may improve outcomes, but current existing data do not facilitate a clear recommendation for a standardized pathway for MRD testing and MRD-directed intervention after transplant. A significant challenge will be to perform well-designed prospective clinical trials in these relatively small patient populations. Although most relapses occur within the first year after transplantation, posttransplant surveillance should probably be continued for up to 2 years or beyond.3  This is especially true for haploidentical transplants in which the frequently observed “HLA-loss” relapses tend to occur at later time points.97  Technical aspects of MRD monitoring according to sample and target analyzed have been provided by the European Leukemia Net.4  BM-MRD is in general 1-log more sensitive than PB and should be routinely performed; however, in the context of allo-HCT, PB-chimerism studies can be used to adjust and better interpret other MRD test results (Figure 1).115  The MRD techniques continue to advance (eg, error-corrected NGS, sensitive ddPCR, and MRD from circulating DNA) and are expected to improve the accuracy of assessment of clonal and immunological changes in low-volume residual disease, thus enabling a more rational therapeutic intervention than is currently possible. In parallel, the landscape of cellular and targeted immunotherapy is evolving rapidly (eg, monoclonal antibodies, bispecific T-cell engagers, checkpoint inhibitors, antigen-specific T cells, chimeric antigen receptor and other genetically engineered T cells, and natural killer cells).116  Less progress has been made in monitoring the speed and quality of GVL reconstitution. Recent reports suggest that the increased frequency of regulatory T cells and exhausted leukemia-specific T cells in BM or the coexpression of inhibitory molecules on circulating T cells represents a dysfunctional GVL pattern that permits eventual relapse.103,117  Understanding the interplay between GVL and MRD remains a major challenge.

The author thanks Maria Gilleece and Panagiotis Tsirigotis for critical proofreading and editing of the manuscript and Maria Liga and Nikos Spyridis for their everyday collegial support.

Contribution: A.S. performed literature research and wrote the manuscript.

Conflict-of-interest disclosure: A.S. declares no competing financial interests.

Correspondence: Alexandros Spyridonidis, Department of Internal Medicine, BMT Unit, University Hospital of Patras, 26500 Rio, Patras, Greece; e-mail: spyridonidis@upatras.gr.

1.
Passweg
JR
,
Baldomero
H
,
Basak
GW
, et al;
European Society for Blood and Marrow Transplantation (EBMT)
.
The EBMT activity survey report 2017: a focus on allogeneic HCT for nonmalignant indications and on the use of non-HCT cell therapies
.
Bone Marrow Transplant
.
2019
;
54
(
10
):
1575
-
1585
.
2.
Horowitz
M
,
Schreiber
H
,
Elder
A
, et al
.
Epidemiology and biology of relapse after stem cell transplantation
.
Bone Marrow Transplant
.
2018
;
53
(
11
):
1379
-
1389
.
3.
Craddock
C
,
Versluis
J
,
Labopin
M
, et al;
Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation and HOVON-SAKK
.
Distinct factors determine the kinetics of disease relapse in adults transplanted for acute myeloid leukaemia
.
J Intern Med
.
2018
;
283
(
4
):
371
-
379
.
4.
Schuurhuis
GJ
,
Heuser
M
,
Freeman
S
, et al
.
Minimal/measurable residual disease in AML: a consensus document from the European LeukemiaNet MRD Working Party
.
Blood
.
2018
;
131
(
12
):
1275
-
1291
.
5.
Ivey
A
,
Hills
RK
,
Simpson
MA
, et al;
UK National Cancer Research Institute AML Working Group
.
Assessment of minimal residual disease in standard-risk AML
.
N Engl J Med
.
2016
;
374
(
5
):
422
-
433
.
6.
Venditti
A
,
Piciocchi
A
,
Candoni
A
, et al
.
GIMEMA AML1310 trial of risk-adapted, MRD-directed therapy for young adults with newly diagnosed acute myeloid leukemia
.
Blood
.
2019
;
134
(
12
):
935
-
945
.
7.
Buckley
SA
,
Wood
BL
,
Othus
M
, et al
.
Minimal residual disease prior to allogeneic hematopoietic cell transplantation in acute myeloid leukemia: a meta-analysis
.
Haematologica
.
2017
;
102
(
5
):
865
-
873
.
8.
Berry
DA
,
Zhou
S
,
Higley
H
, et al
.
Association of minimal residual disease with clinical outcome in pediatric and adult acute lymphoblastic leukemia: a meta-analysis
.
JAMA Oncol
.
2017
;
3
(
7
):
e170580
.
9.
Gökbuget
N
,
Dombret
H
,
Bonifacio
M
, et al
.
Blinatumomab for minimal residual disease in adults with B-cell precursor acute lymphoblastic leukemia
.
Blood
.
2018
;
131
(
14
):
1522
-
1531
.
10.
Hourigan
CS
,
Dillon
LW
,
Gui
G
, et al
.
Impact of conditioning intensity of allogeneic transplantation for acute myeloid leukemia with genomic evidence of residual disease [published online ahead of print 20 December 2019]
.
J Clin Oncol
.
doi:10.1200/JCO.19.03011
.
11.
Mellgren
K
,
Nierop
AFM
,
Abrahamsson
J
.
Use of multivariate immune reconstitution patterns to describe immune reconstitution after allogeneic stem cell transplantation in children
.
Biol Blood Marrow Transplant
.
2019
;
25
(
10
):
2045
-
2053
.
12.
Vago
L
,
Perna
SK
,
Zanussi
M
, et al
.
Loss of mismatched HLA in leukemia after stem-cell transplantation
.
N Engl J Med
.
2009
;
361
(
5
):
478
-
488
.
13.
Christopher
MJ
,
Petti
AA
,
Rettig
MP
, et al
.
Immune escape of relapsed AML cells after allogeneic transplantation
.
N Engl J Med
.
2018
;
379
(
24
):
2330
-
2341
.
14.
Toffalori
C
,
Zito
L
,
Gambacorta
V
, et al
.
Immune signature drives leukemia escape and relapse after hematopoietic cell transplantation
.
Nat Med
.
2019
;
25
(
4
):
603
-
611
.
15.
Pavletic
SZ
,
Kumar
S
,
Mohty
M
, et al
.
NCI First International Workshop on the Biology, Prevention, and Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation: report from the Committee on the Epidemiology and Natural History of Relapse following Allogeneic Cell Transplantation
.
Biol Blood Marrow Transplant
.
2010
;
16
(
7
):
871
-
890
.
16.
Kröger
N
,
Miyamura
K
,
Bishop
MR
.
Minimal residual disease following allogeneic hematopoietic stem cell transplantation
.
Biol Blood Marrow Transplant
.
2011
;
17
(
1 suppl
):
S94
-
S100
.
17.
Craddock
C
,
Hoelzer
D
,
Komanduri
KV
.
Current status and future clinical directions in the prevention and treatment of relapse following hematopoietic transplantation for acute myeloid and lymphoblastic leukemia
.
Bone Marrow Transplant
.
2019
;
54
(
1
):
6
-
16
.
18.
Tsirigotis
P
,
Byrne
M
,
Schmid
C
, et al
.
Relapse of AML after hematopoietic stem cell transplantation: methods of monitoring and preventive strategies. A review from the ALWP of the EBMT
.
Bone Marrow Transplant
.
2016
;
51
(
11
):
1431
-
1438
.
19.
Lee
CJ
,
Savani
BN
,
Mohty
M
, et al
.
Post-remission strategies for the prevention of relapse following allogeneic hematopoietic cell transplantation for high-risk acute myeloid leukemia: expert review from the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation
.
Bone Marrow Transplant
.
2019
;
54
(
4
):
519
-
530
.
20.
Wang
Y
,
Chen
H
,
Chen
J
, et al
.
The consensus on the monitoring, treatment, and prevention of leukemia relapse after allogeneic hematopoietic stem cell transplantation in China
.
Cancer Lett
.
2018
;
438
:
63
-
75
.
21.
Barosi
G
,
Gale
RP
.
Is there expert consensus on expert consensus?
Bone Marrow Transplant
.
2018
;
53
(
8
):
1055
-
1060
.
22.
Spyridonidis
A
,
Liga
M
,
Triantafyllou
E
, et al
.
Pharmacokinetics and clinical activity of very low-dose alemtuzumab in transplantation for acute leukemia
.
Bone Marrow Transplant
.
2011
;
46
(
10
):
1363
-
1368
.
23.
Delsing Malmberg
E
,
Johansson Alm
S
,
Nicklasson
M
, et al
.
Minimal residual disease assessed with deep sequencing of NPM1 mutations predicts relapse after allogeneic stem cell transplant in AML
.
Leuk Lymphoma
.
2019
;
60
(
2
):
409
-
417
.
24.
Zhou
Y
,
Othus
M
,
Walter
RB
,
Estey
EH
,
Wu
D
,
Wood
BL
.
Deep NPM1 sequencing following allogeneic hematopoietic cell transplantation improves risk assessment in adults with NPM1-Mutated AML
.
Biol Blood Marrow Transplant
.
2018
;
24
(
8
):
1615
-
1620
.
25.
Schnittger
S
,
Kern
W
,
Tschulik
C
, et al
.
Minimal residual disease levels assessed by NPM1 mutation-specific RQ-PCR provide important prognostic information in AML
.
Blood
.
2009
;
114
(
11
):
2220
-
2231
.
26.
Bacher
U
,
Badbaran
A
,
Fehse
B
,
Zabelina
T
,
Zander
AR
,
Kröger
N
.
Quantitative monitoring of NPM1 mutations provides a valid minimal residual disease parameter following allogeneic stem cell transplantation
.
Exp Hematol
.
2009
;
37
(
1
):
135
-
142
.
27.
Waterhouse
M
,
Pfeifer
D
,
Duque-Afonso
J
, et al
.
Droplet digital PCR for the simultaneous analysis of minimal residual disease and hematopoietic chimerism after allogeneic cell transplantation
.
Clin Chem Lab Med
.
2019
;
57
(
5
):
641
-
647
.
28.
Shayegi
N
,
Kramer
M
,
Bornhäuser
M
, et al;
Study Alliance Leukemia (SAL)
.
The level of residual disease based on mutant NPM1 is an independent prognostic factor for relapse and survival in AML
.
Blood
.
2013
;
122
(
1
):
83
-
92
.
29.
Jurlander
J
,
Caligiuri
MA
,
Ruutu
T
, et al
.
Persistence of the AML1/ETO fusion transcript in patients treated with allogeneic bone marrow transplantation for t(8;21) leukemia
.
Blood
.
1996
;
88
(
6
):
2183
-
2191
.
30.
Wang
Y
,
Wu
DP
,
Liu
QF
, et al
.
In adults with t(8;21)AML, posttransplant RUNX1/RUNX1T1-based MRD monitoring, rather than c-KIT mutations, allows further risk stratification
.
Blood
.
2014
;
124
(
12
):
1880
-
1886
.
31.
Qin
YZ
,
Wang
Y
,
Xu
LP
, et al
.
The dynamics of RUNX1-RUNX1T1 transcript levels after allogeneic hematopoietic stem cell transplantation predict relapse in patients with t(8;21) acute myeloid leukemia
.
J Hematol Oncol
.
2017
;
10
(
1
):
44
.
32.
Tang
FF
,
Xu
LP
,
Zhang
XH
, et al
.
Monitoring of post-transplant CBFB-MYH11 as minimal residual disease, rather than KIT mutations, can predict relapse after allogeneic haematopoietic cell transplantation in adults with inv(16) acute myeloid leukaemia
.
Br J Haematol
.
2018
;
180
(
3
):
448
-
451
.
33.
Elmaagacli
AH
,
Beelen
DW
,
Kroll
M
,
Trzensky
S
,
Stein
C
,
Schaefer
UW
.
Detection of CBFbeta/MYH11 fusion transcripts in patients with inv(16) acute myeloid leukemia after allogeneic bone marrow or peripheral blood progenitor cell transplantation
.
Bone Marrow Transplant
.
1998
;
21
(
2
):
159
-
166
.
34.
Liu
J
,
Wang
Y
,
Xu
LP
, et al
.
Monitoring mixed lineage leukemia expression may help identify patients with mixed lineage leukemia--rearranged acute leukemia who are at high risk of relapse after allogeneic hematopoietic stem cell transplantation
.
Biol Blood Marrow Transplant
.
2014
;
20
(
7
):
929
-
936
.
35.
Shih
LY
,
Huang
CF
,
Wu
JH
, et al
.
Internal tandem duplication of FLT3 in relapsed acute myeloid leukemia: a comparative analysis of bone marrow samples from 108 adult patients at diagnosis and relapse
.
Blood
.
2002
;
100
(
7
):
2387
-
2392
.
36.
Levis
MJ
,
Perl
AE
,
Altman
JK
, et al
.
A next-generation sequencing-based assay for minimal residual disease assessment in AML patients with FLT3-ITD mutations
.
Blood Adv
.
2018
;
2
(
8
):
825
-
831
.
37.
Spyridonidis
A
,
Bertz
H
,
Ihorst
G
,
Grüllich
C
,
Finke
J
.
Hematopoietic cell transplantation from unrelated donors as an effective therapy for older patients (> or = 60 years) with active myeloid malignancies
.
Blood
.
2005
;
105
(
10
):
4147
-
4148
.
38.
Spyridonidis
A
,
Küttler
T
,
Wäsch
R
, et al
.
Reduced intensity conditioning compared to standard conditioning preserves the in vitro growth capacity of bone marrow stroma, which remains of host origin
.
Stem Cells Dev
.
2005
;
14
(
2
):
213
-
222
.
39.
Spyridonidis
A
,
Zeiser
R
,
Wäsch
R
,
Bertz
H
,
Finke
J
.
Capillary electrophoresis for chimerism monitoring by PCR amplification of microsatellite markers after allogeneic hematopoietic cell transplantation
.
Clin Transplant
.
2005
;
19
(
3
):
350
-
356
.
40.
Valero-Garcia
J
,
González-Espinosa
MDC
,
Barrios
M
, et al
.
Earlier relapse detection after allogeneic haematopoietic stem cell transplantation by chimerism assays: Digital PCR versus quantitative real-time PCR of insertion/deletion polymorphisms [published correction appears in PLoS One. 2019;14(3):e0213966]
.
PLoS One
.
2019
;
14
(
2
):
e0212708
.
41.
Sellmann
L
,
Rabe
K
,
Bünting
I
, et al
.
Diagnostic value of highly-sensitive chimerism analysis after allogeneic stem cell transplantation
.
Bone Marrow Transplant
.
2018
;
53
(
11
):
1457
-
1465
.
42.
Stahl
T
,
Böhme
MU
,
Kröger
N
,
Fehse
B
.
Digital PCR to assess hematopoietic chimerism after allogeneic stem cell transplantation
.
Exp Hematol
.
2015
;
43
(
6
):
462
-
8.e1
.
43.
Qin
XY
,
Li
GX
,
Qin
YZ
, et al
.
Quantitative chimerism: an independent acute leukemia prognosis indicator following allogeneic hematopoietic SCT
.
Bone Marrow Transplant
.
2014
;
49
(
10
):
1269
-
1277
.
44.
McIlroy
G
,
Nikolousis
E
,
Abou-Zeid
N
, et al
.
Mixed T-cell chimerism at 3 months followed by donor lymphocyte infusion is independently associated with favorable outcomes in alemtuzumab-based reduced-intensity allogeneic hematopoietic stem cell transplantation
.
Leuk Lymphoma
.
2020
;
61
(
1
):
202
-
205
.
45.
Kinsella
FAM
,
Inman
CF
,
Gudger
A
, et al
.
Very early lineage-specific chimerism after reduced intensity stem cell transplantation is highly predictive of clinical outcome for patients with myeloid disease
.
Leuk Res
.
2019
;
83
:
106173
.
46.
Zeiser
R
,
Spyridonidis
A
,
Wäsch
R
, et al
.
Evaluation of immunomodulatory treatment based on conventional and lineage-specific chimerism analysis in patients with myeloid malignancies after myeloablative allogeneic hematopoietic cell transplantation
.
Leukemia
.
2005
;
19
(
5
):
814
-
821
.
47.
Bornhäuser
M
,
Oelschlaegel
U
,
Platzbecker
U
, et al
.
Monitoring of donor chimerism in sorted CD34+ peripheral blood cells allows the sensitive detection of imminent relapse after allogeneic stem cell transplantation
.
Haematologica
.
2009
;
94
(
11
):
1613
-
1617
.
48.
Unnikrishnan
A
,
Meacham
AM
,
Goldstein
SS
, et al
.
CD34+ chimerism analysis for minimal residual disease monitoring after allogeneic hematopoietic cell transplantation
.
Leuk Res
.
2018
;
74
:
110
-
112
.
49.
Rossi
G
,
Nomdedéu Guinot
JF
,
Fontana
A
,
Minervini
MM
,
García-Dabrio
MC
,
Cascavilla
N
.
CD117-CD15 in acute myeloid leukemia: no role as LAIP in the study of minimal residual disease
.
Eur J Haematol
.
2013
;
90
(
2
):
171
-
174
.
50.
Jacobsohn
DA
,
Loken
MR
,
Fei
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 Transplant
.
2018
;
24
(
10
):
2040
-
2046
.
51.
Zhou
Y
,
Othus
M
,
Araki
D
, et al
.
Pre- and post-transplant quantification of measurable (“minimal”) residual disease via multiparameter flow cytometry in adult acute myeloid leukemia
.
Leukemia
.
2016
;
30
(
7
):
1456
-
1464
.
52.
Bastos-Oreiro
M
,
Perez-Corral
A
,
Martínez-Laperche
C
, et al
.
Prognostic impact of minimal residual disease analysis by flow cytometry in patients with acute myeloid leukemia before and after allogeneic hemopoietic stem cell transplantation
.
Eur J Haematol
.
2014
;
93
(
3
):
239
-
246
.
53.
Shah
MV
,
Jorgensen
JL
,
Saliba
RM
, et al
.
Early post-transplant minimal residual disease assessment improves risk stratification in acute myeloid leukemia
.
Biol Blood Marrow Transplant
.
2018
;
24
(
7
):
1514
-
1520
.
54.
Bernal
T
,
Diez-Campelo
M
,
Godoy
V
, et al
.
Role of minimal residual disease and chimerism after reduced-intensity and myeloablative allo-transplantation in acute myeloid leukemia and high-risk myelodysplastic syndrome
.
Leuk Res
.
2014
;
38
(
5
):
551
-
556
.
55.
Rossi
G
,
Carella
AM
,
Minervini
MM
, et al
.
Optimal time-points for minimal residual disease monitoring change on the basis of the method used in patients with acute myeloid leukemia who underwent allogeneic stem cell transplantation: a comparison between multiparameter flow cytometry and Wilms’ tumor 1 expression
.
Leuk Res
.
2015
;
39
(
2
):
138
-
143
.
56.
Zhao
XS
,
Yan
CH
,
Liu
DH
, et al
.
Combined use of WT1 and flow cytometry monitoring can promote sensitivity of predicting relapse after allogeneic HSCT without affecting specificity
.
Ann Hematol
.
2013
;
92
(
8
):
1111
-
1119
.
57.
Cho
BS
,
Min
GJ
,
Park
SS
, et al
.
WT1 measurable residual disease assay in patients with acute myeloid leukemia who underwent allogeneic hematopoietic stem cell transplantation: optimal time points, thresholds, and candidates
.
Biol Blood Marrow Transplant
.
2019
;
25
(
10
):
1925
-
1932
.
58.
Rautenberg
C
,
Pechtel
S
,
Hildebrandt
B
, et al
.
Wilms’ tumor 1 gene expression using a standardized European LeukemiaNet-certified assay compared to other methods for detection of minimal residual disease in myelodysplastic syndrome and acute myelogenous leukemia after allogeneic blood stem cell transplantation
.
Biol Blood Marrow Transplant
.
2018
;
24
(
11
):
2337
-
2343
.
59.
Candoni
A
,
Tiribelli
M
,
Toffoletti
E
, et al
.
Quantitative assessment of WT1 gene expression after allogeneic stem cell transplantation is a useful tool for monitoring minimal residual disease in acute myeloid leukemia
.
Eur J Haematol
.
2009
;
82
(
1
):
61
-
68
.
60.
Nomdedéu
JF
,
Esquirol
A
,
Carricondo
M
, et al
.
Bone marrow WT1 levels in allogeneic hematopoietic stem cell transplantation for acute myelogenous leukemia and myelodysplasia: clinically relevant time points and 100 copies threshold value
.
Biol Blood Marrow Transplant
.
2018
;
24
(
1
):
55
-
63
.
61.
Rautenberg
C
,
Pechtel
S
,
Hildebrandt
B
, et al
.
Longitudinal monitoring of Wilms’Tumor 1 (WT1) expression by a standardized European Leukemia Net (ELN)-certified assay for detection of minimal residual disease (MRD) in MDS patients undergoing allogeneic blood stem cell transplantation
.
Oncol Res Treat
.
2016
;
39
:
141
-
141
.
62.
Koukoulias
K
,
Papadopoulou
A
,
Deliyanni
I
, et al
.
Clinical scale production of leukemia specific t-cells from non-transplantable cord blood units
.
Bone Marrow Transplant
.
2017
;
52
:
S151
-
S151
.
63.
Frick
M
,
Chan
W
,
Arends
CM
, et al
.
Role of donor clonal hematopoiesis in allogeneic hematopoietic stem-cell transplantation
.
J Clin Oncol
.
2019
;
37
(
5
):
375
-
385
.
64.
Kim
T
,
Moon
JH
,
Ahn
JS
, et al
.
Next-generation sequencing-based posttransplant monitoring of acute myeloid leukemia identifies patients at high risk of relapse
.
Blood
.
2018
;
132
(
15
):
1604
-
1613
.
65.
Nakamura
S
,
Yokoyama
K
,
Shimizu
E
, et al
.
Prognostic impact of circulating tumor DNA status post-allogeneic hematopoietic stem cell transplantation in AML and MDS
.
Blood
.
2019
;
133
(
25
):
2682
-
2695
.
66.
Jongen-Lavrencic
M
,
Grob
T
,
Hanekamp
D
, et al
.
Molecular minimal residual disease in acute myeloid leukemia
.
N Engl J Med
.
2018
;
378
(
13
):
1189
-
1199
.
67.
Ishio
T
,
Sugita
J
,
Tateno
T
, et al
.
Hematogones predict better outcome in allogeneic hematopoietic stem cell transplantation irrespective of graft sources
.
Biol Blood Marrow Transplant
.
2018
;
24
(
10
):
1990
-
1996
.
68.
Fronkova
E
,
Muzikova
K
,
Mejstrikova
E
, et al
.
B-cell reconstitution after allogeneic SCT impairs minimal residual disease monitoring in children with ALL
.
Bone Marrow Transplant
.
2008
;
42
(
3
):
187
-
196
.
69.
Kotrova
M
,
van der Velden
VHJ
,
van Dongen
JJM
, et al
.
Next-generation sequencing indicates false-positive MRD results and better predicts prognosis after SCT in patients with childhood ALL
.
Bone Marrow Transplant
.
2017
;
52
(
7
):
962
-
968
.
70.
Pulsipher
MA
,
Carlson
C
,
Langholz
B
, 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
(
22
):
3501
-
3508
.
71.
Bader
P
,
Kreyenberg
H
,
von Stackelberg
A
, 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
(
11
):
1275
-
1284
.
72.
Balduzzi
A
,
Di Maio
L
,
Silvestri
D
, et al
.
Minimal residual disease before and after transplantation for childhood acute lymphoblastic leukaemia: is there any room for intervention?
Br J Haematol
.
2014
;
164
(
3
):
396
-
408
.
73.
Miglino
M
,
Berisso
G
,
Grasso
R
, et al
.
Allogeneic bone marrow transplantation (BMT) for adults with acute lymphoblastic leukemia (ALL): predictive role of minimal residual disease monitoring on relapse
.
Bone Marrow Transplant
.
2002
;
30
(
9
):
579
-
585
.
74.
Terwey
TH
,
Hemmati
PG
,
Nagy
M
, et al
.
Comparison of chimerism and minimal residual disease monitoring for relapse prediction after allogeneic stem cell transplantation for adult acute lymphoblastic leukemia
.
Biol Blood Marrow Transplant
.
2014
;
20
(
10
):
1522
-
1529
.
75.
Craddock
C
,
Nagra
S
,
Peniket
A
, et al
.
Factors predicting long-term survival after T-cell depleted reduced intensity allogeneic stem cell transplantation for acute myeloid leukemia
.
Haematologica
.
2010
;
95
(
6
):
989
-
995
.
76.
Spyridonidis
A
.
How much immunosuppression do we need?
Blood
.
2017
;
129
(
10
):
1241
-
1243
.
77.
Oran
B
,
Giralt
S
,
Couriel
D
, et al
.
Treatment of AML and MDS relapsing after reduced-intensity conditioning and allogeneic hematopoietic stem cell transplantation
.
Leukemia
.
2007
;
21
(
12
):
2540
-
2544
.
78.
Spyridonidis
A
,
Liga
M
.
A long road of T-cells to cure cancer: from adoptive immunotherapy with unspecific cellular products to donor lymphocyte infusions and transfer of engineered tumor-specific T-cells
.
Am J Blood Res
.
2012
;
2
(
2
):
98
-
104
.
79.
Tsirigotis
P
,
Liga
M
,
Gkirkas
K
, et al
.
Low-dose alemtuzumab for GvHD prevention followed by prophylactic donor lymphocyte infusions in high-risk leukemia
.
Bone Marrow Transplant
.
2017
;
52
(
3
):
445
-
451
.
80.
Schmid
C
,
Labopin
M
,
Schaap
N
, et al;
EBMT Acute Leukaemia Working Party
.
Prophylactic donor lymphocyte infusion after allogeneic stem cell transplantation in acute leukaemia - a matched pair analysis by the Acute Leukaemia Working Party of EBMT
.
Br J Haematol
.
2019
;
184
(
5
):
782
-
787
.
81.
Schmid
C
,
Labopin
M
,
Nagler
A
, et al;
EBMT Acute Leukemia Working Party
.
Donor lymphocyte infusion in the treatment of first hematological relapse after allogeneic stem-cell transplantation in adults with acute myeloid leukemia: a retrospective risk factors analysis and comparison with other strategies by the EBMT Acute Leukemia Working Party
.
J Clin Oncol
.
2007
;
25
(
31
):
4938
-
4945
.
82.
Zeiser
R
,
Bertz
H
,
Spyridonidis
A
,
Houet
L
,
Finke
J
.
Donor lymphocyte infusions for multiple myeloma: clinical results and novel perspectives
.
Bone Marrow Transplant
.
2004
;
34
(
11
):
923
-
928
.
83.
Hofmann
S
,
Götz
M
,
Schneider
V
, et al
.
Donor lymphocyte infusion induces polyspecific CD8(+) T-cell responses with concurrent molecular remission in acute myeloid leukemia with NPM1 mutation
.
J Clin Oncol
.
2013
;
31
(
3
):
e44
-
e47
.
84.
Greiner
J
,
Schneider
V
,
Schmitt
M
, et al
.
Immune responses against the mutated region of cytoplasmatic NPM1 might contribute to the favorable clinical outcome of AML patients with NPM1 mutations (NPM1mut)
.
Blood
.
2013
;
122
(
6
):
1087
-
1088
.
85.
Dominietto
A
,
Pozzi
S
,
Miglino
M
, et al
.
Donor lymphocyte infusions for the treatment of minimal residual disease in acute leukemia
.
Blood
.
2007
;
109
(
11
):
5063
-
5064
.
86.
Di Grazia
C
,
Pozzi
S
,
Geroldi
S
, et al
.
Wilms tumor 1 expression and pre-emptive immunotherapy in patients with acute myeloid leukemia undergoing an allogeneic hemopoietic stem cell transplantation
.
Biol Blood Marrow Transplant
.
2016
;
22
(
7
):
1242
-
1246
.
87.
Yan
CH
,
Liu
DH
,
Liu
KY
, et al
.
Risk stratification-directed donor lymphocyte infusion could reduce relapse of standard-risk acute leukemia patients after allogeneic hematopoietic stem cell transplantation
.
Blood
.
2012
;
119
(
14
):
3256
-
3262
.
88.
Liga
M
,
Triantafyllou
E
,
Tiniakou
M
, et al
.
High alloreactivity of low-dose prophylactic donor lymphocyte infusion in patients with acute leukemia undergoing allogeneic hematopoietic cell transplantation with an alemtuzumab-containing conditioning regimen
.
Biol Blood Marrow Transplant
.
2013
;
19
(
1
):
75
-
81
.
89.
Hasskarl
J
,
Zerweck
A
,
Wäsch
R
,
Ihorst
G
,
Bertz
H
,
Finke
J
.
Induction of graft versus malignancy effect after unrelated allogeneic PBSCT using donor lymphocyte infusions derived from frozen aliquots of the original graft
.
Bone Marrow Transplant
.
2012
;
47
(
2
):
277
-
282
.
90.
Martelli
MF
,
Di Ianni
M
,
Ruggeri
L
, et al
.
HLA-haploidentical transplantation with regulatory and conventional T-cell adoptive immunotherapy prevents acute leukemia relapse
.
Blood
.
2014
;
124
(
4
):
638
-
644
.
91.
Ciceri
F
,
Bonini
C
,
Stanghellini
MT
, 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
(
5
):
489
-
500
.
92.
Roy
DC
,
Lachance
S
,
Cohen
S
, et al
.
Allodepleted T-cell immunotherapy after haploidentical haematopoietic stem cell transplantation without severe acute graft-versus-host disease (GVHD) in the absence of GVHD prophylaxis
.
Br J Haematol
.
2019
;
186
(
5
):
754
-
766
.
93.
Ghiso
A
,
Raiola
AM
,
Gualandi
F
, et al
.
DLI after haploidentical BMT with post-transplant CY
.
Bone Marrow Transplant
.
2015
;
50
(
1
):
56
-
61
.
94.
Zeidan
AM
,
Forde
PM
,
Symons
H
, et al
.
HLA-haploidentical donor lymphocyte infusions for patients with relapsed hematologic malignancies after related HLA-haploidentical bone marrow transplantation
.
Biol Blood Marrow Transplant
.
2014
;
20
(
3
):
314
-
318
.
95.
Goldsmith
SR
,
Slade
M
,
DiPersio
JF
, et al
.
Donor-lymphocyte infusion following haploidentical hematopoietic cell transplantation with peripheral blood stem cell grafts and PTCy
.
Bone Marrow Transplant
.
2017
;
52
(
12
):
1623
-
1628
.
96.
Cauchois
R
,
Castagna
L
,
Pagliardini
T
, et al
.
Prophylactic donor lymphocyte infusions after haploidentical haematopoietic stem cell transplantation for high risk haematological malignancies: a retrospective bicentric analysis of serial infusions of increasing doses of CD3+ cells
.
Br J Haematol
.
2019
;
185
(
3
):
570
-
573
.
97.
Crucitti
L
,
Crocchiolo
R
,
Toffalori
C
, et al
.
Incidence, risk factors and clinical outcome of leukemia relapses with loss of the mismatched HLA after partially incompatible hematopoietic stem cell transplantation
.
Leukemia
.
2015
;
29
(
5
):
1143
-
1152
.
98.
Ahci
M
,
Toffalori
C
,
Bouwmans
E
, et al
.
A new tool for rapid and reliable diagnosis of HLA loss relapses after HSCT
.
Blood
.
2017
;
130
(
10
):
1270
-
1273
.
99.
Almstedt
M
,
Blagitko-Dorfs
N
,
Duque-Afonso
J
, et al
.
The DNA demethylating agent 5-aza-2′-deoxycytidine induces expression of NY-ESO-1 and other cancer/testis antigens in myeloid leukemia cells
.
Leuk Res
.
2010
;
34
(
7
):
899
-
905
.
100.
Sánchez-Abarca
LI
,
Gutierrez-Cosio
S
,
Santamaría
C
, et al
.
Immunomodulatory effect of 5-azacytidine (5-azaC): potential role in the transplantation setting
.
Blood
.
2010
;
115
(
1
):
107
-
121
.
101.
Goodyear
OC
,
Dennis
M
,
Jilani
NY
, et al
.
Azacitidine augments expansion of regulatory T cells after allogeneic stem cell transplantation in patients with acute myeloid leukemia (AML)
.
Blood
.
2012
;
119
(
14
):
3361
-
3369
.
102.
Schönefeldt
C
,
Sockel
K
,
Wehner
R
, et al
.
Azacytidine impairs NK cell activity in AML and MDS patients undergoing MRD-based pre-emptive treatment after allogeneic stem cell transplantation
.
Blood Cancer J
.
2013
;
3
(
8
):
e136
.
103.
Noviello
M
,
Manfredi
F
,
Ruggiero
E
, et al
.
Bone marrow central memory and memory stem T-cell exhaustion in AML patients relapsing after HSCT
.
Nat Commun
.
2019
;
10
(
1
):
1065
.
104.
Platzbecker
U
,
Wermke
M
,
Radke
J
, et al
.
Azacitidine for treatment of imminent relapse in MDS or AML patients after allogeneic HSCT: results of the RELAZA trial
.
Leukemia
.
2012
;
26
(
3
):
381
-
389
.
105.
Platzbecker
U
,
Middeke
JM
,
Sockel
K
, et al
.
Measurable residual disease-guided treatment with azacitidine to prevent haematological relapse in patients with myelodysplastic syndrome and acute myeloid leukaemia (RELAZA2): an open-label, multicentre, phase 2 trial
.
Lancet Oncol
.
2018
;
19
(
12
):
1668
-
1679
.
106.
Schroeder
T
,
Rachlis
E
,
Bug
G
, 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 Transplant
.
2015
;
21
(
4
):
653
-
660
.
107.
Lin
XJ
,
Dai
HP
,
Wang
AJ
, et al
.
Effects of preemptive interferon-α monotherapy in acute leukemia patients with relapse tendency after allogeneic hematopoietic stem cell transplantation: a case-control study
.
Ann Hematol
.
2018
;
97
(
11
):
2195
-
2204
.
108.
Kneppers
E
,
van der Holt
B
,
Kersten
MJ
, et al
.
Lenalidomide maintenance after nonmyeloablative allogeneic stem cell transplantation in multiple myeloma is not feasible: results of the HOVON 76 Trial
.
Blood
.
2011
;
118
(
9
):
2413
-
2419
.
109.
Craddock
C
,
Slade
D
,
De Santo
C
, et al
.
Combination lenalidomide and azacitidine: a novel salvage therapy in patients who relapse after allogeneic stem-cell transplantation for acute myeloid leukemia
.
J Clin Oncol
.
2019
;
37
(
7
):
580
-
588
.
110.
Bug
G
,
Burchert
A
,
Wagner
EM
, et al
.
Phase I/II study of the deacetylase inhibitor panobinostat after allogeneic stem cell transplantation in patients with high-risk MDS or AML (PANOBEST trial)
.
Leukemia
.
2017
;
31
(
11
):
2523
-
2525
.
111.
de Lima
M
,
Oran
B
,
Champlin
RE
, et al
.
CC-486 maintenance after stem cell transplantation in patients with acute myeloid leukemia or myelodysplastic syndromes
.
Biol Blood Marrow Transplant
.
2018
;
24
(
10
):
2017
-
2024
.
112.
Davids
MS
,
Kim
HT
,
Bachireddy
P
, et al;
Leukemia and Lymphoma Society Blood Cancer Research Partnership
.
Ipilimumab for patients with relapse after allogeneic transplantation
.
N Engl J Med
.
2016
;
375
(
2
):
143
-
153
.
113.
Haverkos
BM
,
Abbott
D
,
Hamadani
M
, et al
.
PD-1 blockade for relapsed lymphoma post-allogeneic hematopoietic cell transplant: high response rate but frequent GVHD
.
Blood
.
2017
;
130
(
2
):
221
-
228
.
114.
Mathew
NR
,
Baumgartner
F
,
Braun
L
, et al
.
Sorafenib promotes graft-versus-leukemia activity in mice and humans through IL-15 production in FLT3-ITD-mutant leukemia cells [published correction appears in Nat Med. 2018;24(4):526]
.
Nat Med
.
2018
;
24
(
3
):
282
-
291
.
115.
Stahl
T
,
Badbaran
A
,
Kröger
N
, et al
.
Minimal residual disease diagnostics in patients with acute myeloid leukemia in the post-transplant period: comparison of peripheral blood and bone marrow analysis
.
Leuk Lymphoma
.
2010
;
51
(
10
):
1837
-
1843
.
116.
Falkenburg
F
,
Ruggiero
E
,
Bonini
C
, et al
.
Prevention and treatment of relapse after stem cell transplantation by cellular therapies
.
Bone Marrow Transplant
.
2019
;
54
(
1
):
26
-
34
.
117.
Hutten
TJA
,
Norde
WJ
,
Woestenenk
R
, et al
.
Increased coexpression of PD-1, TIGIT, and KLRG-1 on tumor-reactive CD8+ T cells during relapse after allogeneic stem cell transplantation
.
Biol Blood Marrow Transplant
.
2018
;
24
(
4
):
666
-
677
.
Sign in via your Institution