Abstract
Allogeneic haematopoietic stem cell transplantation (HSCT) remains the only potential cure for intermediate to high-risk acute myeloid leukemia (AML). The therapeutic effect of HSCT is largely dependent on the powerful donor-derived immune response against recipient leukaemia cells, known as graft-versus-leukaemia effect (GvL). However, the donor-derived immune system can also cause acute or chronic damage to normal recipient organs and tissues, in a process known as graft-versus-host disease (GvHD). GvHD is a leading cause of non-relapse mortality in HSCT recipients. There are many similarities and cross talk between the immune pathways of GvL and GvHD. Studies have demonstrated that both processes require the presence of mismatched alloantigens between the donor and recipient, and activation of immune responses centered around donor T-cells, which can be further modulated by various recipient or donor factors. To dissect GvL from GvHD for more effective GvHD prevention and enhanced GvL has been the holy grail of HSCT research. In this review, we focused on the key factors that contribute to the immune responses of GvL and GvHD, the effect on GvL with different GvHD prophylactic strategies, and the potential impact of various AML relapse prevention therapy or treatments on GvHD.
Keywords: graft-versus-leukaemia, graft-versus-host disease, allogeneic haematolpoietic stem cell transplantation, acute myeloid leukaemia
Introduction
The application of allogeneic haematopoietic stem cell transplantation (HSCT) in human was first reported in 1957 based on the discovery in mice that allogeneic immune cells could exert powerful anti-leukemic effects, i.e., graft-versus-leukaemia (GvL) [1, 2]. Since then, major discoveries in human immune system and leukaemia biology have greatly improved transplant outcomes and molded the modern-day practice of HSCT [3, 4]. To date, HSCT remains the only potentially curative treatment for eligible patients with acute myeloid leukaemia (AML) that harbors adverse clinical, cytogenetic or molecular risk factors [5, 6]. AML is the most common indication for HSCT in the US and worldwide [7, 8]. Still, post-transplant relapse occurs in 15–60% of patients and carries a dismal prognosis [9]. To prevent relapse, it is crucial to maintain or enhance GvL early after HSCT.
Graft-versus-host disease (GvHD) is a unique side effect of HSCT. In GvHD, the donor immune system elicits an immune response towards the allogeneic donor tissue, causing a myriad of symptoms such as acute rash, diarrhea, and liver injury weeks to months post-HSCT (acute GvHD, aGvHD), and/or chronic malabsorption, sclerotic skin, sicca, and obstructive lung disease months to years later (chronic GvHD, cGvHD) [2, 10–12]. Even with declining incidence and better treatment, GvHD remains the leading cause of non-relapse mortality (NRM) post-HSCT [13–19]. Although both GvL and GvHD are dependent on sustained immune reconstitution of the donor immune system, the presence of GvHD does not always translate into stronger GvL and reduced relapse incidence (RI), suggesting differences in the immune responses which could be targeted to GvL without inducing GvHD, or reducing GvHD without dampening GvL [20, 21]. The immune responses that are implicated in GvL and GvHD are yet to be fully elucidated. Here we summarize the key components of GvL/GvHD immune responses and the mechanisms of action of selected GvHD prophylactic therapy and relapse-prevention/treatment regimens with a focus on AML.
Histocompatibility antigens
Human leukocyte antigens
The human leukocyte antigen (HLA) system comprises the main antigens involved in GvHD and GvL. HLAs are highly polymorphic and presented on all nucleated cells (class I) or APCs (class II), triggering CD8+ T-cell and CD4+ T-cell-dependent immune response, respectively [22]. Recognition of mismatched recipient alloantigens on leukaemia cells and the subsequent immune response centered around donor mature CD8+ cytotoxic T-cells is the key to GvL [23]. By losing the mismatched HLA, AML can evade GvL and relapse [24]. Alloantigens also participate in GvHD. Unlike pathogen-specific immunity, GvHD immune response is characterized by ubiquitously present alloantigen, does not require antigen presenting cells (APCs) maturation, and the immune response is constant as the alloantigens cannot be eliminated [25]. Higher disparity of HLA epitope between the donor and recipient theoretically enhances donor-derived immune reaction that leads to both GvL and GvHD.
Clinically, the effect of HLA-disparities on transplant outcome can be modulated by many other donor/recipient factors, conditioning and GvHD prophylaxis. As an example, haploidentical related donor (HRD) HSCT, once associated with prohibitive risks of severe GvHD due to high level HLA disparity, is widely used now owing to the development of modern GvHD prophylaxis strategies [26, 27]. Various studies examined the effect of HLA-matching on the outcome of HSCT in AML with some conflicting results. A multicenter study from China compared the outcome of HRD (n = 231) with HLA-identical matched sibling donor (MSD, n = 219) HSCT in patients with AML in 1st complete remission (CR1), and found no significant difference in DFS, OS, incidence of relapse and non-relapse mortality (NRM) between the two, but the incidence of aGvHD grade II-IV/III-IV and cGvHD was much higher with HRD [28]. The EBMT conducted several large retrospective studies comparing donor type in different scenarios. In high-risk AML in first relapse, MUD HSCT was associated with better LFS and lower relapse incidence compared with MSD [29]. In AML in CR1 with previous induction failure, 10/10 MUD, HRD and MSD achieved similar RI, LFS and OS, but GvHD-free relapse-free survival (GRFS) was worse in 9/10 mismatched MUD HSCT [30]. In high-cytogenetic risk AML in CR1, HRD was associated with a higher incidence of GvHD, lower RI, and similar LFS, OS and GRFS compared with MSD, but in intermediate-risk AML MSD outperformed HRD owing to higher GvHD-NRM in the latter [31].
A few studies on optimal donor choice were controlled for GvHD prophylactic regimen. A CIBMTR study compared the outcome of 336 post-transplant cyclophosphamide (PtCy)-based HRD vs 869 calcineurin-inhibitor based MSD HSCT in AML in CR1, showing significantly lower incidence of cGvHD in the former (hazard ratio, HR 0.38), while OS, LFS, NRM, RI, incidence of grade II-IV aGvHD were similar [32]. When PtCy is used homogenously, another EBMT study showed that HRD HSCT (n = 789) carried a significantly higher risk of grade II-IV aGvHD (HR 1.6) and NRM (HR 2.6), lower RI (HR 0.7), similar LFS (HR 1.1) and GRFS (HR 1.0) compared with MSD (n = 215) and MUD (n = 235) HSCT for AML in CR1 [33]. A Spanish multicenter retrospective study where PtCy was uniformly used, however, demonstrated lower rates of aGvHD in MSD/MUD than HRD for patients with AML undergoing HSCT, while RI, NRM, OS, GRFS were similar [34]. Overall, the outcome of HRD is comparable to MSD and 10/10 MUD for high-risk AML patients.
Besides the degree of HLA mismatching, the effect of different HLA antigens on GvHD or GvL varies. For example, mismatch in class I antigens such as HLA-A or HLA-C is a strong risk factor for aGvHD, while class II antigen mismatch has a less prominent role in aGvHD [35]. Mismatches in HLA-A, -B, -C, and DPB1 were associated with grade III–IV aGvHD, mismatches in HLA-C with cGvHD, and both HLA-C and -DPB1 mismatches were associated with reduced RI [36]. The direction of HLA mismatch also matters. In HRD HSCT, HLA-DRB1 mismatches in the GvH direction are associated with lower NRM, and HLA-DPB1 non-permissive mismatches are associated with lower RI comparing to permissive mismatches. Further, mismatch in certain HLA single nucleotide polymorphisms (SNPs) may induce stronger T-cell alloimmunity and GvHD [37]. Deeper HLA sequencing to the SNP level may therefore reduce GvHD and improve disease-free survival (DFS) [38, 39]. Future endeavors in HLA research may direct towards finding more GvL-inducing and GvHD-reducing HLA-combinations to improve donor selection for optimal HSCT outcomes.
Non-HLA antigens
The most studied non-HLA antigens are the minor histocompatibility antigens (miHAs) [40]. The number of miHA in matched unrelated donor (MUD) is roughly twice as many as matched sibling donor (MSD) transplants, speaking to the greater genetic disparity of the former [41]. MiHA-mismatch is crucial to maintain CD8+ T-cell alloreactivity [42]. When pre-sensitized with donor antigen, CD4+ T-cells could develop immunotolerance across miHA barriers but CD8+ T-cells do not [43]. Infusion of miHA-specific CD8+ alloreactive T-cell clone could cause GvL, acute and chronic GvHD [44, 45]. Mismatch in non-HLA antigens, such as the SNP of Paired-immunoglobulin type 2-like receptor β, has been associated with increased transplant-related mortality (TRM) due to increased GvHD [46].
The ability of miHAs to induce GvHD and GvL depends not on the total number of miHA mismatches, but on the immunogenicity and the location of different miHAs [41, 47, 48]. As an example, miHA encoded by the Y-chromosome are associated with higher incidence of GvHD due to wider tissue distribution, while autosomal-encoded miHAs are associated with prolonged relapse-free survival (RFS) due to their more restricted distribution on haematopoietic cells [49, 50]. Immunodominance is an important phenomenon in which certain miHAs, such as HA-1, dominate alloreactivity, stimulate miHA-specific T-cell responses, and drive the presence of GvL or GvHD [50–52]. Variations in the presentation of miHAs by different MHC types also contribute to the clinical presentation of GvHD [53]. Identifying the haematopoietic tissue-specific miHA and the less immunogenic miHA or HLA-miHA couple could potentially alleviate GvHD and augment GvL. One such example is GRK4, a miHA highly expressed in AML and several other cancers, but has little to no expression in other normal tissues except the testis [41].
Proinflammatory T-cells
Sustained reconstitution of mature donor αβ T-cells is pivotal for both GvHD and GvL, demonstrated by a plethora of preclinical and clinical studies [43, 54–56]. Donor T-cell immune reconstitution is also essential for infection prevention and engraftment. Different T-cell subtypes exert distinctive effects in GvHD and GvL [54, 56–58]. The dose of CD3+ T-cells, especially the CD8+ subset, is predictive of the severity of aGvHD [59]. Among the CD4+ T-cells, naïve T-cells are more potent than memory T-cells for GvHD. In murine models, naïve T-cells induced a more severe acute GvHD than alloantigen-primed memory T-cells and had more vigorous homing activity to the colonic tissue and mesenteric lymph nodes [60]. Naïve T-cells induce the differentiation of miHA-specific alloreactive CD8+ T cells, whereas memory T-cells are more likely to generate pathogen-specific immune responses [61]. Unprimed memory T-cells lack the ability to induce GvHD due to an abortive immune response to alloantigen [62].
T-helper (Th)1 and Th17 cells are potent GvHD inducers and are derived from naïve T-cells upon stimulation by exposed alloantigens on tissues and altered intestinal microbiota signals [54, 63]. They produce inflammatory cytokines such as interferon(IFN)-γ, interleukin(IL)-17 and tumor necrosing factor (TNF)α and activate cytotoxic T-cells [64]. Murine studies showed that inhibiting IFN-γ could reduce gastrointestinal GvHD but increase Th17/IL-17 production and pulmonary damage, while IL-17 blockade would lead to Th1 proliferation, suggesting a reciprocal differentiation of Th1 and Th17 cells post-HSCT [65, 66]. Blocking the differentiation of both Th1 and Th17 cells could effectively prevent GvHD without affecting GvL [67]. Th22 cells are a recently defined Th subtype associated with inflammatory skin conditions such as scleroderma [68]. Th22 and its product IL-22 have been associated with the pathogenesis of cutaneous chronic GvHD [69]. Down regulation of IL-22 could attenuate skin GvHD while sparing the GvL effect [70]. Paradoxically, IL22 is protective in intestinal GvHD, highlighting the complexity of GvHD pathogenesis [71–73].
Targeting proinflammatory T-cells to prevent GvHD in AML
GvHD prophylactic regimens evolve around how to eliminate or reduce the function of proinflammatory T-cells. A T-cell replete peripheral blood (PB) graft carries a higher volume of mature donor T-cells which confers stronger GvL and GvHD effects. Studies across donor types and GvHD prophylaxis regimens invariably demonstrated faster engraftment, less infection, reduced RI, improved DFS, but higher risk of aGvHD and cGvHD using T-cell replete PB than bone marrow (BM) graft [74–77]. Non-selective ex vivo T-cell depletion (TCD) can significantly reduce the risk of GvHD but at the cost of significantly higher RI, risk of infection, graft failure, and post-transplant lymphoproliferative disease [78–80]. Delayed regeneration of T-cell dependent functions and reduced diversity of T-cell receptor (TCR) repertoire were observed in TCD BM HSCT [81, 82]. However, high intensity conditioning (myeloablative chemotherapy plus myeloablative total body irradiation) seemed to ameliorate the detrimental effect of TCD on relapse while preserving its anti-GvHD effect [83]. Selective TCD by ex vivo αβ TCD to less than 1×105/kg αβ T-cells, frequently in combination with CD19+ B-cell depletion, has been studied mainly in pediatric patients [84]. The risk of aGvHD II-IV was seen in 3–39% and cGvHD in 0–30%. Relapse occurred in 22–58% of patients and was the main cause of death [84]. Naïve TCD has also been studied. In a total of 138 patients with acute leukaemia, naïve TCD lead to low incidence of grade III–IV aGvHD (4%) and moderate to severe cGvHD (1%), without apparent increase of RI or NRM [85].
Pharmacologic immunosuppressive therapy (IST) such as methotrexate (MTX), calcineurin inhibitors and mycophenolate mofetil (MMF) reduces T cell activation, proliferation and differentiation. ISTs have been widely used for GvHD prophylaxis since the inception of HSCT, with some studies suggesting higher RI with IST [86, 87]. Therefore, timely discontinuation of IST is important after HSCT for AML, especially those transplanted with measurable residual disease (MRD) or for those who represent MRD features after prior clearance. There is no consensus regarding the optimal timing of IST discontinuation or whether IST should be tapered, though most practices stop or taper IST around 60–100 days post-HSCT in the absence of GvHD.
Anti-thymocyte globulins (ATG) are polyclonal antibodies extracted from horse or rabbit sera, immunized with human thymic cells or T lymphoblastoid cell line [88]. ATG has been used for ex vivo and in vivo TCD. When used in vivo, both host T-cells and incoming allograft T-cells can be effectively reduced due to ATG’s long half-life [89]. ATG reduces GvHD by reducing proinflammatory T-cells, increasing regulatory T-cells, selective enhancement of NK-cells, inducing B-cell apoptosis and reducing APC function [90–93]. Among different ATG formulations, rabbit ATG is more potent than horse ATG in preventing GvHD, likely due to stronger T-cell depleting ability [94, 95]. ATG has been widely studied in MUD HSCT, and in HRD HSCT in China. Randomized and non-randomized studies have consistently showed less incidence and severity of acute or chronic GvHD, and no major change in RI in patients with haematologic malignancies receiving ATG for GvHD prophylaxis [89, 96–101].
PtCy has demonstrated a powerful ability to reduce GvHD in murine studies and was soon applied to human HRD HSCT [102–104]. PtCy not only reduces the function, proliferation and differentiation of alloreactive effector T-cells like IST, but also promotes the preferential recovery of CD4+ regulatory T-cells [105, 106]. In addition, PtCy restores early post-transplant B-cell homeostasis favoring naïve B-cells and suppresses activation of B-cells at peripheral lymph nodes, adding additional protection against cGvHD [107]. With PtCy, HRD HSCT now has comparable outcomes to non-PtCy based MSD HSCT, greatly enlarging the donor pool [32, 108]. Besides the HRD setting, PtCy-based GvHD prophylaxis has been increasingly applied to HLA-matched HSCT [109–112] ref.
Whether the use of PtCy affects GvL remains inconclusive. The occurrence of aGvHD or cGvHD after PtCy has not been associated with reduced RI, suggesting a dissociation between GvL and GvHD in this setting [113, 114]. The higher RI after PtCy observed in some studies may well be explained by the difference in conditioning intensity and not the PtCy [115]. When controlled for conditioning intensity, PtCy-based GvHD prophylaxis significantly lowered the incidence of both aGvHD and cGvHD compared with non-PtCy, without change in RI [110]. Patients who relapse after PtCy-based HSCT showed different T- and NK-cell activation signatures than those who did not relapse [116]. Several large, randomized studies compared PtCy-based GvHD prophylaxis with other GvHD prophylaxis methods. The BMTCTN 1301 study compared CD34-selected PB (n = 114) vs single agent PtCy BM (n = 109) vs tacrolimus/MTX BM HLA-matched HCT (n = 114), with around 60% of patients in each group transplanted for AML [111]. The PtCy group had the lowest RI compared to the other two groups (HR 0.52, p = 0.037), comparable OS to tacrolimus/methotrexate and better OS than CD34-selection [111]. The BMTCTN 1703 compared PtCy/tacrolimus/MMF vs tacrolimus/MTX for MSD, MUD, or 7/8 mismatched unrelated donor reduced intensity conditioning HSCT, in which about 50% of patients in each group had AML [112]. The RI and OS at 1-year were similar between the groups, but the PtCy group had better GRFS (HR 0.64, p = 0.001) due to lower incidence of aGvHD and cGvHD [112].
Post-transplant Bendamustine (PtB) has been studied as an alternative to PtCy as murine studies provided evidence that PtB was as effective as PtCy in mitigating GvHD, less myelosuppressive, and preserved GvL likely by increasing myeloid-derived suppressor cells [117]. Katsanis et al. reported a phase 1 study substituting day +4 PtCy with PtB. The PtCy/PtB combination was well-tolerated, had faster engraftment and lower incidence of CMV reactivation compared with contemporaneous controls using PtCy [118]. Moiseev et al. reported a dose-finding prospective clinical trial studying PtB in patients with refractory leukaemia (22 AML and 5 ALL) receiving myeloablative conditioning [119]. Compared with historical PtCy data, PtB was associated with a faster and more successful engraftment even in refractory leukaemia. As a single agent, PtB prevented aGvHD well but not cGvHD. Unexpectedly, PtB was associated with cytokine release syndrome that mimicked macrophage activation syndrome, which was not seen in pre-clinical models [119]. Ongoing studies on immunomodulatory GvHD prevention are summarized in Table 1.
Table 1.
Ongoing investigational immunomodulatory approaches in AML HSCT.
Immune target | Clinicaltrials.gov identifier # | Clinical Setting, if applicable | Country |
---|---|---|---|
miHA | |||
MiHA-loaded PD-L-silenced DC Vaccination | NCT02528682 | Netherlands | |
Ex vivo T-cell depletion | |||
alpha-beta T-cell depletion, with CD19+ B cell depletion | NCT04337515, NCT02323867, NCT05794880 (with targeted ATG dosing), NCT04099966 | United States | |
TCRαβ-depleted Progenitor Cell Graft with Additional Memory T-cell DLI | NCT03849651 | United States | |
Naïve T cell depletion | NCT03779854 | Pediatric | United States |
Allogeneic CD34+-enriched and CD45RA-depleted PBSCs | NCT03970096 | United States | |
Ex Vivo T-Cell Depletion of Mobilized PBSCs via CD34-Selection | NCT01189786 | United States | |
Ex Vivo Expanded/Activated Gamma Delta T-cell Infusion | NCT03533816 | After HRD with PtCy | United States |
Orca-T: T-cell-Depleted Graft with Additional Infusion of Conventional T Cells and Regulatory T Cells | NCT05316701 | United States | |
PT-Cy | |||
PTCy + ATG | NCT04202835, NCT03357159, NCT03608059 | Canada, Australia, Israel, China | |
PT-Cy vs. ATG | NCT04888741, NCT05153226 | United Kingdom, Germany | |
PT-Cy + methotrexate | NCT04622956 | Brazil | |
PT-Cy + PT-Bendamustine or PT-Bendamustine alone | NCT02996773, NCT04022239 | United States | |
PT-Cy + Bortezomib | NCT03850366 | United States | |
PT-Cy, Bortezomib + Abatacept | NCT05289167 | MSD or ≥7/8 MUD | United States |
PT-Cy, Abatacept + Tacrolimus | NCT05621759 | HRD | United States |
PT-Cy + uhCG/EGF | NCT04886726 | MMUD | United States |
PT-Cy + Itacitinib | NCT05364762, NCT05823571 | 8/8 MRD or MUD HRD | United States |
PT-Cy + Ruxolitinib | NCT05622318 | United States | |
PT-Cy + total marrow and lymphoid irradiation (TMLI) | NCT03467386 | 10/10 MRD or MUD | United States |
PT-Cy, Abatacept, Vedolizumab + CNI | NCT05515029 | Pediatric/AYA | Russia |
PT-Cy dose de-escalation | NCT05436418, NCT04959175, NCT03983850, NCT05158608 | United States Russia | |
DLI | |||
T-cell Receptor α/β Depleted Donor Lymphocyte Infusions | NCT05350163 | United States | |
Siremadlin Alone and in Combination with DLI | NCT05447663 | United States, Israel, Italy, Spain, Germany | |
Regulatory T-cells | |||
Treg-enriched donor cell infusion |
NCT05095649
NCT04678401 |
SR-CGVHD Peri-HRD transplant | Spain United States |
Purified regulatory T-cells (Treg) plus CD34+ HSPC | NCT05088356 | United States | |
T-allo10 cells: T regulatory type 1 (Tr1) cells | NCT03198234 | MMRD, MMUD | United States |
NK Cells | |||
Ex Vivo Expanded Allogeneic NK Cells Infusion | NCT05250362 | China | |
Cytokine Induced Memorylike NK Cell Adoptive Therapy | NCT02782546 | After HRD | United States |
Expanded Natural Killer Cells | NCT03300492 | Switzerland | |
Other Adoptive Cell Therapies | |||
TSC-100, TSC-101 (donorderived T cells targeting HA-1 and HA-2, respectively) | NCT05473910 | HLA A*02:01 positive patients, HRD | United States |
Interferon γ-Primed Mesenchymal Stromal Cells | NCT04328714 | United States | |
Immunomodulators for GVHD prophylaxis | |||
Vorinostat | NCT03842696 | Pediatric, AYA | United States |
Tocilizumab | NCT03434730, NCT04688021 | United States China | |
Itacitinib | NCT03755414 | United States | |
Ponatinib | NCT03690115 | France | |
Baricitinib | NCT04131738 | United States | |
Sitagliptin | NCT05149365 | China | |
Belimumab | NCT03207958 | United States | |
RGI-2001 | NCT04014790 | United States | |
Alpha-1 Antitrypsin (AAT) | NCT03805789 | United States, Australia, Germany, Spain, United Kingdom | |
High Dose Vitamin A | NCT03719092 | United States | |
Tildrakizumab | NCT04112810 | United States | |
Ustekinumab | NCT04572815 | United States | |
Anti-CD40L Domain Antibody (BMS-986004) | NCT03605927 | United States | |
Microbiota | |||
Prebiotic Galactooligosaccharide | NCT04373057 | United States | |
Oral Pooled Fecal Microbiotherapy (MaaT033) | NCT05762211 | France | |
Sodium butyrate | NCT05808985 | Korea | |
Fecal Microbiota Transplantation (FMT) | NCT04935684 | France | |
Post-HSCT Maintenance/ Relapse Prevention | |||
Low Dose Azacitidine | NCT01995578 | United States | |
Glasdegib | NCT04168502 | Italy | |
Tagraxofusp | NCT05233618 | CD123+ AML | United States |
Magrolimab + Azacitidine | NCT05823480 | United States | |
Venetoclax + Azacitidine vs decitabine/cedazuridine | NCT03613532 | After ven + conditioning | United States |
Decitabine | NCT05528354 | After ven + decitabine + conditioning | China |
Azacitidine + Chidamide | NCT05270200 | China |
Regulatory T-cells
Regulatory T-cells (Treg) are important negative modulators of GvHD. They are crucial for immune tolerance and can alleviate GvHD by cytolysis and metabolic suppression of effector T-cells, modulating APC maturation and function, and producing immunosuppressive cytokines [120]. The number of Treg in the graft has been inversely associated with aGvHD preclinically and clinically [121]. Thymic injury that occurs due to peri-HSCT events including aGvHD, will impair negative selection, corrupt APC function, leading to Treg failure and ultimately cGvHD [53, 121]. Tregs have higher affinity to IL-2 than conventional CD4+ T-cells. In vivo expansion of Treg using IL-2 can restore Treg and alleviate cGvHD symptoms in small phase 1/2 clinical trials for patients with GvHD [122–124]. However, higher leukaemia relapse incidence was seen in small clinical trials using low dose IL-2 as GvHD prophylaxis [125, 126]. Treg infusion trials have not shown convincing efficacy in preventing GvHD [127, 128]. The disappointing results of Treg-enhancing therapy could be explained by the plasticity of late-stage Treg such as Treg to Th-17 conversion triggered by APCs [129]. Belumosudil, a ROCK2 inhibitor FDA-approved for refractory cGvHD, exerts its therapeutic effect partly through shifting the Th17/Treg balance towards Treg [130].
Antigen-presenting cells
Host APCs participate in T-cell-dependent aGvHD [131]. Host APCs are estimated to be 100–1000 times more potent than donor APCs in inducing aGvHD via HLA class II. Among host APCs, the tissue-resident non-haematopoietic APCs such as intestinal epithelial cells are more potent than professional APCs in the lymphoid organs [132, 133]. Conditioning-induced tissue damage and microbiota disruption are important activators of APCs [134, 135]. In mice, depletion of host macrophages and dendritic cells (DC) from the liver and spleen could reduce recruitment of donor CD8+ T-cells to the liver and inhibit liver aGvHD [136]. In the gut, the propagation of aGvHD is dependent on a profound feed-forward loop between host DC and naïve donor DC [137]. The antigen presentation by donor DC subsequently triggers the migration of proinflammatory Th1/Th17 to the gut to cause GvHD [138]. Despite the prominent role of host APC in GvHD, depletion of host APC cannot prevent or ameliorate GvHD, given the fact that all host tissues harbor alloantigens and the existing redundancy of antigen-presentation in GvHD immune response [139].
Donor APCs intensify GvHD but are not essential for GvL [140]. Donor DC creates a feed-forward cascade by antigen presentation and cytokine release in GvHD [137]. The level CD123+ plasmacytoid DC in the graft is a strong predictor of aGvHD [59]. Depletion of donor CD11b- DC from the BM graft led to significant expansion of donor CD4+ memory T-cells, enhanced GvL without producing GvHD in a murine BMT model [141]. Another way to exploit donor DC in the setting of AML is to selectively enrich the leukaemia antigen-specific DC, which was shown to enhance GvL without causing GvHD in a postallograft relapse animal model [142].
Humoral immunity
B-cells are involved in both acute and chronic GvHD [107, 143]. Higher B-cell content in the graft is associated with higher risk of aGvHD [59]. In cGvHD, B-cells often express exhaustion markers such as CD22 [144]. Patients with cGvHD often have elevated immunoglobulin and auto/alloantibody levels, but the high immunoglobin levels do not translate into effective antimicrobial response [145]. Several B-cell markers have been proposed as biomarkers for cGvHD, such as plasma B-cell activating factor (BAFF) level, BAFF/CD19+ ratio, and level of CD19+CD21low transitional B-cells [146–148]. Various B-cell targeting agents have been applied to the treatment of cGvHD, highlighting the importance of B-cells in the pathogenesis [149–151]. Although B-cells may participate in GvL by producing anti-miHA antibodies, the role is less prominent than in GvHD [152]. Treating cGvHD with B-cell targeting agents such as ibrutinib and fostamatinib did not increase the incidence leukaemia relapse [150, 151].
Innate immune cells
Innate immune cells are promising therapeutics for AML due to their unique properties of inducing GvL without exacerbating GvHD, or even reducing GvHD. Natural killer (NK)-cells are the first donor-derived lymphocytes to recover after HSCT [153]. Timely NK-cell recovery is associated with decreased incidence of leukaemia relapse, viral reactivation and GvHD [154]. In a landmark study by Ruggeri et al., HSCT with NK-cell alloreactivity, compared with those without, had significantly lower incidence of AML relapse (0 vs 75%, p < 0.0008), graft rejection (0 vs 15.5%, p ≤ 0.01) and aGvHD grade ≥ 2 (0 vs 13.7%, p ≤ 0.01) [155]. In patients receiving PtCy, timely NK-cell recovery is predictive of improved OS, lower OS and RI [116]. Donor memory-like NK cells have potent anti-AML effects when given to patients with post-transplant relapse and did not induce GvHD [156, 157]. Both donor and recipient NK-cells can down-regulate GvHD. Donor NK-cells can eliminate recipient APCs and prevent donor T-cell activation upon recognition of non-self HLA ligands by surface killer immunoglobulin-like receptors (KIRs) [155, 158]. Donor NK-cells suppress the number and activity of alloreactive T-cells via perforin- and Fas ligand-dependent pathways [159]. Delayed NK-cell recovery, especially the CD56-high CD16-dim naïve NK-cells, is associated with increased incidence and severity of aGvHD [160, 161]. Recipient NK-cells, on the other hand, can reduce alloreactive T-cell expansion in target organs. Pre-HSCT conditioning may promote GvHD by eliminating recipient NK-cells and disrupting the intestinal barrier [162].
Γδ T-cells are an integral part of both the innate and adaptive immune system [163]. They comprise the majority of residential T-cells in the skin and mucosa [164]. Unlike αβ T-cells, γδ T-cells usually do not express the CD4 or CD8 coreceptors and undergo activation in an HLA-unrestricted manner [163]. As a result, γδ T-cells kill leukaemia cells via direct recognition of surface antigens independent of HLA [165]. Many studies have demonstrated that γδ T-cells are crucial for GvL and anti-microbial immune responses [164, 166, 167]. In one study of 153 patients with acute leukaemia, patients with high vs low/normal post-HSCT γδ T-cells had better 5-year LFS (54.4 vs 19.1%, p<0.0003) and OS (70.8 vs 19.6, p<0.0001), there were no difference in GvHD risk [168, 169]. In AML patients with pre-transplant MRD, early γδ T-cell recovery reduced the risk of relapse by 60% and significantly improved LFS and OS [170]. The beneficial effect of high γδ T-cell levels on survival may also be attributed to reduced incidence of infection as a result of more robust immune reconstitution [171]. Several studies have suggested a limited role of γδ T-cells in GvHD pathogenesis. A meta-analysis of 11 studies (919 patients) showed that high γδ T-cell levels post-HSCT was associated with less relapse, fewer viral infections, higher overall and disease-free survivals, and no association with aGvHD [172]. Several clinical trials that sought to increase γδ T-cell levels post-HSCT by in vivo expansion or infusion of activated γδ T-cells are currently underway, with promising preliminary results showing prolonged survival and low incidence of GvHD [165, 173].
Donor clonal haematopoiesis
Several retrospective studies demonstrated the possible connection between donor clonal hematopoiesis (CH), relapse, and the development of acute or chronic GvHD [174–177]. Specifically, mutations in DNMT3A, TET2, and ASXL1 are associated with a 2-fold and 2 to 8-fold increase in the incidence of grade II–IV and grade III–IV aGvHD, respectively, likely due to enhanced production of proinflammatory cytokines and Th17 in the presence of these mutations [174]. Donor DNMT3A-CH is also associated with reduced RI and increased cGvHD likely by augmenting both GvL and GvHD through proinflammatory T-cell cytokines [176]. Donor CH carries the risk of donor-derived myeloid malignancies [176, 178]. These data suggest a potential benefit to screen for donor CH prior to transplant especially in older donors where CH is more prevalent.
Microbiome
The fact that organs/tissues with microbiota are mostly affected by GvHD is not a mere coincidence but suggestive of the importance of dysbiosis in the pathogenesis of GvHD. There is also growing evidence that microbiota are associated with anti-leukaemia immunity. Disrupted gut microbiota may reduce anti-leukaemia immunity by altered secretion of bacterial metabolites [179]. A phase 3 clinical trial demonstrated that gut-decontamination with oral metronidazole could lower the risk of grade II–IV aGvHD [180]. A small phase 2 study showed that gut decontamination with oral vancomycin-polymycin B did not significantly change the gut microbiome diversity, T-cell or B-cell subset reconstitution, and clinical outcomes, but reduced the risk of blood stream infection [181]. The diversity of gut microbiome is inversely correlated with TRM and the risk of GvHD [182]. Specifically, elimination of certain Clostrida species is associated with the development of GvHD [183]. The abundance of certain species, such as Blautia, has been associated with lower GvHD-related mortality [182]. Small case series demonstrated that restoration of gut microbiome diversity and symbiosis by fecal microbiota transplantation may be useful in severe intestinal SR-GvHD [184]. Similarly, decreased skin microbiome diversity has been observed in cutaneous GvHD [185]. Various peri-HSCT events can affect the health of human microbiome, such as pre-transplant chemotherapy, conditioning regimens, and antibiotics. Abnormal microbiome can cause reduced production of molecules such as short chain fatty acids, decrease bile acid clearance, thereby impairing endothelial repair [134]. Bacterial antigens could trigger DC activation and subsequent recruitment of proinflammatory T-cells, eventually causing GvHD [186]. Besides bacteria, fungal elements may also recruit proinflammatory T-cells and play a role in GvHD [187]. It is plausible that better preservation of microbiome homeostasis during AML treatment would improve immune reconstitution, leading to lower GvHD and relapse risk.
In summary, the mechanism of GvHD is a complicated process and has frequent overlap with GvL (Figure 1). The mismatched donor alloantigens and other antigens such as altered microbial elements, activate GvHD immune response centered around pro-inflammatory donor T-cells. The immune response attacks recipient tissues and organs, which are already vulnerable after chemotherapy, radiation, infection and other insults, leading to GvHD. Therefore, the prevention of GvHD should aim at reducing recipient tissue damage, antigen mismatching, and immune reactivation especially proinflammatory donor T-cells, while preserving the strong anti-leukaemia effect exerted by the graft.
Figure 1.
Key factors that intersect with the immunopathogenesis of GvHD and GvL in alloHSCT for AML
Post-transplant therapy and GvHD
Post-HSCT relapse of AML remains one of the main causes of treatment failure and is associated with an extremely poor prognosis, with 1-year OS after relapse of 20–25% [188]. Multiple evasion mechanisms have been identified in AML that help the leukaemia cells escape immune surveillance by donor-derived immune system [189]. To prevent relapse, various post-transplant therapies are being studied retrospectively or in a clinical trial setting (Table 1). The goal of post-HSCT therapy is to suppress residual AML, prevent immune evasion, while avoiding the induction of GvHD.
Preemptive and prophylactic DLI
In patients with mixed chimerism, positive MRD, or at high risk of leukaemia relapse, donor lymphocyte infusions (DLI) given preemptively (preDLI) or prophylactically (proDLI) may be employed to prevent recurrent disease. The former refers to DLI given to patients with MRD, molecular relapse, or mixed chimerism (MC); the latter refers to DLI given to patients with high-risk disease or T-cell depleted grafts [190]. A series of murine studies demonstrated that after non-myeloablative transplant, preDLI could eliminate mixed chimerism without causing clinically significant GvHD [191, 192].
A German two-center retrospective study showed that proDLI doubled the 7-year OS of high-risk AML patients (DLI vs no DLI, 67% vs 31%, p < 0.001) [193]. Similarly, a multicenter study from China demonstrated that proDLI significantly improving the 3-year OS and LFS in patients with high-risk acute leukaemia [194]. According to a retrospective EBMT study of 105 patients with AML, pre/proDLI was associated with improved 5-year OS in AML patients with unfavorable cytogenetics or transplant beyond CR1 (compared with controls, 69.8% vs 40.2%, p = 0.027) [195]. In another EBMT study of 318 patients (AML, n = 249), the 5-year RI of preDLI for MRD, pre-DLI for MC and proDLI were 44%, 28% and 28%, and 5-year OS were 51%, 63%, and 68%, suggesting the detrimental effect of MRD on relapse and survival. Patients with MRD/mixed chimerism who responded to preDLI had better 5-year OS than non-responders (63%/76% vs 30%) [190].
GvHD is a major contributor to post-DLI NRM, but it remains unclear whether pre/proDLI truly increases the risk of GvHD. One EBMT study suggested that the increase in GvHD risk associated with DLI was not significant [195]. In contrast, the aforementioned Chinese study showed that prophylactic DLI increased the incidence of cGvHD (DLI vs control, 2-year incidence of cGvHD 38% vs 17%, p=0.021), but not aGvHD (incidence of grade II–IV aGvHD, 17% vs 23%, p=0.35) [194]. In the pre/proDLI setting, DLI-induced GvHD is not associated with reduced RI but clearly associated with higher NRM and poorer OS [190]. Risk factors for post-DLI GvHD included age > 60 years (p=0.046), transplantation beyond CR1 (p=0.003), interval from HSCT to DLI < 6 months (p=0.018), and a history of grade II–IV aGvHD before DLI (p=0.036) [190]. Careful selection of patients for pre/proDLI may be necessary to avoid the risk of GvHD and preserve the benefit of GvL.
Maintenance therapy
Post-HSCT pharmacologic maintenance therapy should optimally reduce the incidence of relapse but should do so without exerting hematological side effects or detrimental drug interactions with other post-transplant medications. Hypomethylating agents (HMAs) are effective therapy for AML and have been studied as post-HSCT maintenance. In the post-transplant setting, preclinical studies suggested that HMAs could epigenetically modulate donor-derived immunity, leading to Treg expansion, induce cytotoxic T-cell response to upregulated tumor antigens, and enhance NK-cell function by inducing KIR expression [196–198]. Therefore, HMAs could theoretically enhance GvL and reduce GvHD [199]. Moreover, HMAs are less myelosuppressive than conventional chemotherapy, making them desirable agents to test in the post-HSCT maintenance setting. In clinical trials, however, there are conflicting results whether post-HSCT HMA maintenance could reduce RI or improvement in RFS [200–204]. In a phase 3 study randomizing 187 patients with high-risk myeloid malignancy (around 60% high-risk AML) to azacitidine vs no azacitidine post-HSCT, the two groups had no significant difference in OS (HR 0.84, p = 0.43) or RFS (HR 0.73, p = 0.14) [204]. However, less than 30% patients were able to complete the planned total 12 cycles of therapy, raising concern about the tolerability and feasibility of maintenance azacitidine [204]. The addition of granulocyte colony-stimulating factor (G-CSF) may reduce hematologic toxicity of HMA and enable delivery of therapy. A randomized phase II study from China studied the G-CSF/decitabine maintenance vs no maintenance in 204 high-risk AML patients post-transplant and found significantly lower 2-year cumulative RI in the maintenance group (15.0% vs 38.3%, p < 0.01) and no significant increase in the rate of cGvHD [205].
Venetoclax (VEN) has been studied off-label in the post-HSCT maintenance setting. In single center studies, the main adverse events of venetoclax included cytopenia and diarrhea, but it did not increase the risk of GvHD beyond historic controls [206, 207]. A small study tested the combination of low dose decitabine with venetoclax as post-HSCT maintenance in 20 patients (AML, 17), showing a 2-year OS of 85.2%, aGvHD (any grade) in 55% and cGvHD in 20% of patients [207]. A randomized, phase 3 study of azacitidine/venetoclax as post-HSCT maintenance (VIALE-T) is currently being conducted [208].
Targeted agents against leukaemia specific mutations have been applied to post-HSCT maintenance. Sorafenib is a FLT3 inhibitor that has on-target effect against FLT3-mutated AML, furthermore, it may enhance GvL through autocrine cytokine production [209]. Two randomized multicenter studies in Europe and China, respectively, showed that sorafenib was associated with reduced RI, improved RFS and OS [210, 211]. However, difficulty in recruiting and premature discontinuation of treatment have been observed in these trials, questioning the feasibility and tolerability of sorafenib. Moreover, sorafenib was associated with increased risk of aGvHD and cGvHD [211]. Another FLT3 inhibitor, midostaurin, was assessed in the randomized RADIUS trial as post-HSCT maintenance. The addition of midostaurin achieved a 40% relative reduction in the risk of relapse but did not significantly improve RFS or OS [212]. Although midostaurin was not associated with higher risk of GvHD, it significantly increased gastrointestinal adverse events [212]. Gilteritinib, a newer generation FLT3 inhibitor, demonstrated promising effect in preventing AML relapse in small retrospective studies [213]. However, preliminary results from the randomized phase 3 MORPHO trial (BMTCTN 1506) showed no improvement in RFS with gilteritinib maintenance [214]. IDH1 and IDH2 inhibitors have also been tested in small phase 1 studies as post-HSCT maintenance for patients with respective mutations. Enasidenib appeared safe post-HSCT [215]. Results from the ivosidenib maintenance trial is still pending (NCT03564821).
Adoptive NK-cell therapy
The efficacy and risk of adoptive NK-cell therapy to prevent AML relapse has been studied in small, early phase clinical trials [216]. The NK-cell donor may be different from the stem cell donor (third party) and selected based on KIR ligand mismatch and KIR B motif number [217, 218]. Infusing NK-cells from a third party may have lower risk of GvHD [218]. Prophylactic NK-cell therapy may be performed before or after HSCT. A phase 1 clinical trial delivered expanded NK-cell transfer from a third party haploidentical donor prior to HSCT for high-risk myeloid malignancy, and demonstrated a positive correlation between survival and the NK-cell dose [218]. In another phase 1 clinical trial, infusion of ex vivo expanded, purified NK-cells before and after (day −2, +7 and +28) HRD HSCT for high-risk myeloid malignancies was safe, with lower incidence of relapse, and no increase in GvHD observed in the 13 patients enrolled [219]. A phase 1/2 study studied the efficacy and safety of IL-15 and IL-21 expanded HSCT donor-derived NK-cell delivered at 2-week and 3-week post-HSCT in patients with high-risk acute leukaemia (AML, 78%, n = 32), showing significant reduction in relapse with NK-cell infusion, and no increase in GvHD [220]. However, another phase 2 study from the same group showed that additional NK-cell infusion at day +6 and +9 did not further reduce relapse but was associated with higher toxicity from cytokine release syndrome [221]. Although adoptive NK-cell therapy showed promising results in prevention post-HSCT relapse, ex vivo expansion and in vivo persistence of the transferred NK-cells remain the major limitations. The optimal timing and dosage of NK-cell infusion also need to be defined [222].
GvHD and the Treatment for post-HSCT Relapse
There is a lack of consensus on the best treatment for post-HSCT relapse. A combination strategy of tapering immunosuppressants, chemotherapy, DLI, and/or second HSCT is commonly deployed. The complexity of treatment often makes it difficult to distinguish the effect on GvHD from each individual approach.
In medically-fit patients with post-SCT relapse, conventional intensive chemotherapy may be associated with better overall response rates and OS compared with hypomethylating agents (HMA) [223]. In different studies, CR rates after conventional chemotherapy were 40–70%, and around 10% to HMA [223–227]. HMA in combination with VEN has been used in post-HSCT relapse. There is no clear evidence that HMA/VEN triggers or exacerbates GvHD. In a small case series of four patients, one developed limited skin GvHD after HMA/VEN while also receiving four sessions of DLI [228]. In one multicenter retrospective study, 32 patients received HMA/VEN for post-transplant relapse, none developed new aGvHD, one developed cGvHD in the setting of DLI[229].
DLI is a commonly used treatment for post-HSCT relapse. In a German single center study of 51 patients with post-HSCT AML relapse who were given DLI, median 2-year and 5-year OS were 25.5 and 9.8%, respectively [230]. In an EBMT multicenter retrospective study of adult patients with first relapse post-HSCT, the 2-year OS was 21% vs 9% in DLI-recipients (n = 171) vs no DLI (n = 228, p < 0.0001) [231]. DLI improves the CR rates and survival in both chemotherapy- and HMA-treated patients with relapsed AML [223]. Like prophylactic DLI, therapeutic DLI is also associated with significantly increased, and even higher, risks of acute and chronic GvHD. Schmid et al, showed that the incidence of post-DLI grade II–IV aGvHD approached 40% and was associated with inferior survival (relative risk, RR, 0.54, p = 0.003), whereas the risk of cGvHD was 46% and associated with a better outcome among survivors more than 100 days after DLI (RR, 2.29, p = 0.004) [231].
Post-DLI GvHD and GvL are both dose-dependent, but some patients could enjoy a potent GvL effect without clinical GvHD [232, 233]. The risk of GvHD after DLI for relapsed AML is similar to pre/proDLI [230]. Therefore, a DLI dose escalation schedule was widely accepted [234, 235]. Initial CD3+ cell dose higher than 100 million/kg was associated with higher incidence of GvHD, and did not improve the RI or survival [236]. Pre-DLI lymphodepletion with fludarabine and cyclophosphamide was also associated with significantly higher incidence of aGvHD due to increased T-cell proliferation. Risks of grade II-IV aGvHD and grade III-IV aGvHD were 60% and 47% in patients receiving lymphodepletion and DLI, vs 24% and 14% in DLI alone (both p = 0.01) [237]. Immunomodulating agents can potentially ameliorate the pro-GvHD effect of DLI. In murine studies, post-mismatched DLI cyclophosphamide could eradicate alloreactive T-cells, prevent lethal GvHD while preserving a potent GvL effect [238].
Second HSCT can be considered in medically fit patients especially those who relapsed ≥ 6 months post-first HSCT. The choice of donor may be the same donor, a different matched donor, or a different HRD. The EBMT compared the outcomes of 2nd HSCT in AML using different donors, and found similar LFS, incidence of GvHD and relapse among the groups, but the risk of NRM doubled in HRD [239].
Relapsed AML has increased expression of immune evasion associated proteins (e.g., PD-L1) and/or HLA class II silencing [240]. Targeting the PD-1/PD-L1 pathway has yielded conflicting results in small phase 1/2 clinical trials for relapsed/refractory AML [241]. Pre-transplant PD-1/PD-L1 blockade is associated with exacerbated GvHD, which can be ameliorated by PtCy [242–244]. PD-1/PD-L1 blockade has not been studied in post-HSCT settings. Other novel checkpoint inhibitors, such as magrolimab, sabatolimab, have not been studied for post-transplant relapse either [245, 246]. Adoptive cellular therapies are in early development for relapsed AML. A phase 1 study showed poor feasibility of anti-HA-1 T-cells in post-HSCT relapse [247]. Off-the-shelf CD33 CAR-NK cells are safe in relapsed/refractory AML, but the feasibility and efficacy of CAR-NK warrants further testing in larger clinical trials [248].
Summary
Despite frequent cross-talks between the immune mechanism of GvL and GvHD, several key differences have been suggested by pre-clinical or clinical studies, such as hematologic tissue-restricted vs more ubiquitously expressed alloantigens, different T-cell differentiation profiles and the higher dependence on humoral immunity by GvHD. Innate immune cells such as NK-cells and γδ T-cells may have potent antileukemic effects while remaining suppressive or neutral on GvHD. Deeper understanding of the differences in GvL and GvHD immunopathogenesis will likely inspire better GvHD prevention, boost the GvL effect, and lead to more-effective anti-relapse therapy, ultimately to fully harness the power of HSCT in AML.
Practice Points.
HSCT remains the only potential curative therapeutic approach for patients with intermediate or high-risk AML.
The main causes of failure of HSCT are AML relapse and GvHD.
Detailed understanding of the immunopathogenesis of GvHD has enabled the employment of more effective GvHD prevention strategies.
Research Agenda.
GvHD remains the leading cause of NRM for patients with AML undergoing HSCT.
Deeper understanding of the differences between the mechanisms of GvHD and GvL immune responses could facilitate the development of better HSCT platforms to suppress the former and enhance the latter.
Treatment of post-HSCT AML relapse should avoid triggering or exacerbating GvHD.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of Interest
The authors declare no conflicts of interest.
References
- 1.Thomas ED, Lochte HL, Lu WC, Ferrebee JW. Intravenous Infusion of Bone Marrow in Patients Receiving Radiation and Chemotherapy. New England Journal of Medicine. 1957;257(11):491–6. [DOI] [PubMed] [Google Scholar]
- 2.Barnes DWH, Corp MJ, Loutit JF, Neal FE. Treatment of Murine Leukaemia with X Rays and Homologous Bone Marrow. BMJ. 1956;2(4993):626–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Dausset J The major histocompatibility complex in man. Science. 1981;213(4515):1469–74. [DOI] [PubMed] [Google Scholar]
- 4.Cui L, Liu Y, Pang Y, Qian T, Quan L, Cheng Z, et al. Emerging agents and regimens for treatment of relapsed and refractory acute myeloid leukemia. Cancer Gene Therapy. 2020;27(1–2):1–14. [DOI] [PubMed] [Google Scholar]
- 5.Snowden JA, Sánchez-Ortega I, Corbacioglu S, Basak GW, Chabannon C, De La Camara R, et al. Indications for haematopoietic cell transplantation for haematological diseases, solid tumours and immune disorders: current practice in Europe, 2022. Bone Marrow Transplantation. 2022;57(8):1217–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Loke J, Buka R, Craddock C. Allogeneic Stem Cell Transplantation for Acute Myeloid Leukemia: Who, When, and How? Front Immunol. 2021;12:659595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Auletta J, Kou J, Chen M, Shaw B. Current use and outcome of hematopoietic stem cell transplantation: CIBMTR US summary slides. Center for International Blood and Marrow Transplant Research. 2021. [Google Scholar]
- 8.Niederwieser D, Baldomero H, Bazuaye N, Bupp C, Chaudhri N, Corbacioglu S, et al. One and a half million hematopoietic stem cell transplants: continuous and differential improvement in worldwide access with the use of non-identical family donors. Haematologica. 2021;107(5):1045–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Abou Dalle I, Atoui A, Bazarbachi A. The Elephant in The Room: AML Relapse Post Allogeneic Hematopoietic Cell Transplantation. Frontiers in Oncology. 2022;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Vriesendorp HM, Heidt PJ. History of graft-versus-host disease. Experimental Hematology. 2016;44(8):674–88. [DOI] [PubMed] [Google Scholar]
- 11.MATHE G, AMIEL JL, SCHWARZENBERG L, CATTAN A, SCHNEIDER M, DEVRIES MJ, et al. SUCCESSFUL ALLOGENIC BONE MARROW TRANSPLANTATION IN MAN: CHIMERISM, INDUCED SPECIFIC TOLERANCE AND POSSIBLE ANTI-LEUKEMIC EFFECTS. Blood. 1965;25:179–96. [PubMed] [Google Scholar]
- 12.Shulman HM, Sale GE, Lerner KG, Barker EA, Weiden PL, Sullivan K, et al. Chronic cutaneous graft-versus-host disease in man. Am J Pathol. 1978;91(3):545–70. [PMC free article] [PubMed] [Google Scholar]
- 13.Weiden PL, Flournoy N, Thomas ED, Prentice R, Fefer A, Buckner CD, et al. Antileukemic Effect of Graft-versus-Host Disease in Human Recipients of Allogeneic-Marrow Grafts. New England Journal of Medicine. 1979;300(19):1068–73. [DOI] [PubMed] [Google Scholar]
- 14.Wingard JR, Majhail NS, Brazauskas R, Wang Z, Sobocinski KA, Jacobsohn D, et al. Long-Term Survival and Late Deaths After Allogeneic Hematopoietic Cell Transplantation. Journal of Clinical Oncology. 2011;29(16):2230–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Lee SJ, Kim HT, Ho VT, Cutler C, Alyea EP, Soiffer RJ, et al. Quality of life associated with acute and chronic graft-versus-host disease. Bone Marrow Transplant. 2006;38(4):305–10. [DOI] [PubMed] [Google Scholar]
- 16.DeFilipp Z, Alousi AM, Pidala JA, Carpenter PA, Onstad LE, Arai S, et al. Nonrelapse mortality among patients diagnosed with chronic GVHD: an updated analysis from the Chronic GVHD Consortium. Blood Advances. 2021;5(20):4278–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Arai S, Arora M, Wang T, Spellman SR, He W, Couriel DR, et al. Increasing Incidence of Chronic Graft-versus-Host Disease in Allogeneic Transplantation: A Report from the Center for International Blood and Marrow Transplant Research. Biology of Blood and Marrow Transplantation. 2015;21(2):266–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Hahn T, McCarthy PL, Hassebroek A, Bredeson C, Gajewski JL, Hale GA, et al. Significant Improvement in Survival After Allogeneic Hematopoietic Cell Transplantation During a Period of Significantly Increased Use, Older Recipient Age, and Use of Unrelated Donors. Journal of Clinical Oncology. 2013;31(19):2437–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Greinix HT, Eikema D-J, Koster L, Penack O, Yakoub-Agha I, Montoto S, et al. Improved outcome of patients with graft-versus-host disease after allogeneic hematopoietic cell transplantation for hematologic malignancies over time: an EBMT mega-file study. Haematologica. 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Thomas ED, Buckner CD, Banaji M, Clift RA, Fefer A, Flournoy N, et al. One hundred patients with acute leukemia treated by chemotherapy, total body irradiation, and allogeneic marrow transplantation. Blood. 1977;49(4):511–33. [PubMed] [Google Scholar]
- 21.Boyiadzis M, Arora M, Klein JP, Hassebroek A, Hemmer M, Urbano-Ispizua A, et al. Impact of Chronic Graft-versus-Host Disease on Late Relapse and Survival on 7,489 Patients after Myeloablative Allogeneic Hematopoietic Cell Transplantation for Leukemia. Clin Cancer Res. 2015;21(9):2020–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Choo SY. The HLA System: Genetics, Immunology, Clinical Testing, and Clinical Implications. Yonsei Medical Journal. 2007;48(1):11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Bleakley M, Riddell SR. Molecules and mechanisms of the graft-versus-leukaemia effect. Nature Reviews Cancer. 2004;4(5):371–80. [DOI] [PubMed] [Google Scholar]
- 24.McCurdy SR, Iglehart BS, Batista DA, Gocke CD, Ning Y, Knaus HA, et al. Loss of the mismatched human leukocyte antigen haplotype in two acute myelogenous leukemia relapses after haploidentical bone marrow transplantation with post-transplantation cyclophosphamide. Leukemia. 2016;30(10):2102–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Shlomchik WD. Graft-versus-host disease. Nature Reviews Immunology. 2007;7(5):340–52. [DOI] [PubMed] [Google Scholar]
- 26.Anasetti C, Amos D, Beatty PG, Appelbaum FR, Bensinger W, Buckner CD, et al. Effect of HLA compatibility on engraftment of bone marrow transplants in patients with leukemia or lymphoma. New England Journal of Medicine. 1989;320(4):197–204. [DOI] [PubMed] [Google Scholar]
- 27.Anasetti C, Beatty PG, Storb R, Martin PJ, Mori M, Sanders JE, et al. Effect of HLA incompatibility on graft-versus-host disease, relapse, and survival after marrow transplantation for patients with leukemia or lymphoma. Human immunology. 1990;29(2):79–91. [DOI] [PubMed] [Google Scholar]
- 28.Wang Y, Liu Q-F, Xu L-P, Liu K-Y, Zhang X-H, Ma X, et al. Haploidentical vs identical-sibling transplant for AML in remission: a multicenter, prospective study. Blood. 2015;125(25):3956–62. [DOI] [PubMed] [Google Scholar]
- 29.Ruggeri A, Battipaglia G, Labopin M, Ehninger G, Beelen D, Tischer J, et al. Unrelated donor versus matched sibling donor in adults with acute myeloid leukemia in first relapse: an ALWP-EBMT study. J Hematol Oncol. 2016;9(1):89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Nagler A, Labopin M, Mielke S, Passweg J, Blaise D, Gedde-Dahl T, et al. Matched related versus unrelated versus haploidentical donors for allogeneic transplantation in AML patients achieving first complete remission after two induction courses: a study from the ALWP/EBMT. Bone Marrow Transplant. 2023. [DOI] [PubMed] [Google Scholar]
- 31.Salvatore D, Labopin M, Ruggeri A, Battipaglia G, Ghavamzadeh A, Ciceri F, et al. Outcomes of hematopoietic stem cell transplantation from unmanipulated haploidentical versus matched sibling donor in patients with acute myeloid leukemia in first complete remission with intermediate or high-risk cytogenetics: a study from the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation. Haematologica. 2018;103(8):1317–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Rashidi A, Hamadani M, Zhang M-J, Wang H-L, Abdel-Azim H, Aljurf M, et al. Outcomes of haploidentical vs matched sibling transplantation for acute myeloid leukemia in first complete remission. Blood advances. 2019;3(12):1826–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Sanz J, Galimard JE, Labopin M, Afanasyev B, Angelucci E, Ciceri F, et al. Post-transplant cyclophosphamide after matched sibling, unrelated and haploidentical donor transplants in patients with acute myeloid leukemia: a comparative study of the ALWP EBMT. J Hematol Oncol. 2020;13(1):46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Bailén R, Pascual-Cascón MJ, Guerreiro M, López-Corral L, Chinea A, Bermúdez A, et al. Post-Transplantation Cyclophosphamide After HLA Identical Compared to Haploidentical Donor Transplant in Acute Myeloid Leukemia: A Study on Behalf of GETH-TC. Transplantation and Cellular Therapy. 2022;28(4):204.e1–e10. [DOI] [PubMed] [Google Scholar]
- 35.Sasazuki T, Juji T, Morishima Y, Kinukawa N, Kashiwabara H, Inoko H, et al. Effect of Matching of Class I HLA Alleles on Clinical Outcome after Transplantation of Hematopoietic Stem Cells from an Unrelated Donor. New England Journal of Medicine. 1998;339(17):1177–85. [DOI] [PubMed] [Google Scholar]
- 36.Morishima Y, Kashiwase K, Matsuo K, Azuma F, Morishima S, Onizuka M, et al. Biological significance of HLA locus matching in unrelated donor bone marrow transplantation. Blood. 2015;125(7):1189–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Petersdorf EW, Carrington M, O’HUigin C, Bengtsson M, De Santis D, Dubois V, et al. Role of HLA-B exon 1 in graft-versus-host disease after unrelated haemopoietic cell transplantation: a retrospective cohort study. Lancet Haematol. 2020;7(1):e50–e60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Petersdorf EW, Malkki M, Gooley TA, Spellman SR, Haagenson MD, Horowitz MM, et al. MHC-Resident Variation Affects Risks After Unrelated Donor Hematopoietic Cell Transplantation. Science Translational Medicine. 2012;4(144):144ra01–ra01. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Jeanmougin M, Noirel J, Coulonges C, Zagury JF. HLA-check: evaluating HLA data from SNP information. BMC Bioinformatics. 2017;18(1):334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.den Haan JMM, Sherman NE, Blokland E, Huczko E, Koning F, Drijfhout JW, et al. Identification of a Graft Versus Host Disease-Associated Human Minor Histocompatibility Antigen. Science. 1995;268(5216):1476–80. [DOI] [PubMed] [Google Scholar]
- 41.Lansford JL, Dharmasiri U, Chai S, Hunsucker SA, Bortone DS, Keating JE, et al. Computational modeling and confirmation of leukemia-associated minor histocompatibility antigens. Blood Advances. 2018;2(16):2052–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Choksi S, Kim JC, Whitaker-Menezes D, Murphy GF, Friedman TM, Korngold R. A CD8 DE loop peptide analog prevents graft-versus-host disease in a multiple minor histocompatibility antigen-mismatched bone marrow transplantation model. Biol Blood Marrow Transplant. 2004;10(10):669–80. [DOI] [PubMed] [Google Scholar]
- 43.Korngold R, Sprent J. Variable capacity of L3T4+ T cells to cause lethal graft-versus-host disease across minor histocompatibility barriers in mice. Journal of Experimental Medicine. 1987;165(6):155–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kircher B, Stevanovic S, Urbanek M, Mitterschiffthaler A, Rammensee H-G, Grünewald K, et al. Induction of HA-1-specific cytotoxic T-cell clones parallels the therapeutic effect of donor lymphocyte infusion. British Journal of Haematology. 2002;117(4):935–9. [DOI] [PubMed] [Google Scholar]
- 45.Marijt WAE, Heemskerk MHM, Kloosterboer FM, Goulmy E, Kester MGD, Van Der Hoorn MAWG, et al. Hematopoiesis-restricted minor histocompatibility antigens HA-1- or HA-2-specific T cells can induce complete remissions of relapsed leukemia. Proceedings of the National Academy of Sciences. 2003;100(5):2742–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Hahn T, Wang J, Preus LM, Karaesmen E, Rizvi A, Clay-Gilmour AI, et al. Novel genetic variants associated with mortality after unrelated donor allogeneic hematopoietic cell transplantation. EClinicalMedicine. 2021;40:101093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Jones SC, Murphy GF, Friedman TM, Korngold R. Importance of minor histocompatibility antigen expression by nonhematopoietic tissues in a CD4+ T cell–mediated graft-versus-host disease model. Journal of Clinical Investigation. 2003;112(12):1880–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Broen K, Greupink-Draaisma A, Woestenenk R, Schaap N, Brickner AG, Dolstra H. Concurrent Detection of Circulating Minor Histocompatibility Antigen-Specific CD8+ T Cells in SCT Recipients by Combinatorial Encoding MHC Multimers. PLoS ONE. 2011;6(6):e21266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Dzierzak-Mietla M, Markiewicz M, Siekiera U, Mizia S, Koclega A, Zielinska P, et al. Occurrence and Impact of Minor Histocompatibility Antigens’ Disparities on Outcomes of Hematopoietic Stem Cell Transplantation from HLA-Matched Sibling Donors. Bone Marrow Research. 2012;2012:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Hobo W, Broen K, Van Der Velden WJFM, Greupink-Draaisma A, Adisty N, Wouters Y, et al. Association of Disparities in Known Minor Histocompatibility Antigens with Relapse-Free Survival and Graft-versus-Host Disease after Allogeneic Stem Cell Transplantation. Biology of Blood and Marrow Transplantation. 2013;19(2):274–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kaplan DH, Anderson BE, McNiff JM, Jain D, Shlomchik MJ, Shlomchik WD. Target antigens determine graft-versus-host disease phenotype. J Immunol. 2004;173(9):5467–75. [DOI] [PubMed] [Google Scholar]
- 52.Hombrink P, Hadrup SR, Bakker A, Kester MGD, Falkenburg JHF, Von Dem Borne PA, et al. High-Throughput Identification of Potential Minor Histocompatibility Antigens by MHC Tetramer-Based Screening: Feasibility and Limitations. PLoS ONE. 2011;6(8):e22523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Müller AMS, Min D, Wernig G, Levy RB, Perez VL, Herretes S, et al. Modeling Chronic Graft-versus-Host Disease in MHC-Matched Mouse Strains: Genetics, Graft Composition, and Tissue Targets. Biology of Blood and Marrow Transplantation. 2019;25(12):2338–49. [DOI] [PubMed] [Google Scholar]
- 54.Jiang H, Fu D, Bidgoli A, Paczesny S. T Cell Subsets in Graft Versus Host Disease and Graft Versus Tumor. Frontiers in Immunology. 2021;12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Korngold R, Sprent J. Lethal graft-versus-host disease after bone marrow transplantation across minor histocompatibility barriers in mice. Prevention by removing mature T cells from marrow. Journal of Experimental Medicine. 1978;148(6):1687–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Korngold R, Sprent J. Features of T cells causing H-2-restricted lethal graft-vs.-host disease across minor histocompatibility barriers. Journal of Experimental Medicine. 1982;155(3):872–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Truitt R, Shih C-Y, Lefever A, Tempelis L, Andreani M, Bortin M. Characterization of alloimmunization-induced T lymphocytes reactive against AKR leukemia in vitro and correlation with graft-vs-leukemia activity in vivo. The Journal of Immunology. 1983;131(4):2050–8. [PubMed] [Google Scholar]
- 58.Korngold R, Sprent J. T cell subsets and graft-versus-host disease. Transplantation. 1987;44(3):335–9. [DOI] [PubMed] [Google Scholar]
- 59.Impola U, Larjo A, Salmenniemi U, Putkonen M, Itälä-Remes M, Partanen J. Graft Immune Cell Composition Associates with Clinical Outcome of Allogeneic Hematopoietic Stem Cell Transplantation in Patients with AML. Frontiers in Immunology. 2016;7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Dutt S, Tseng D, Ermann J, George TI, Liu YP, Davis CR, et al. Naive and memory T cells induce different types of graft-versus-host disease. J Immunol. 2007;179(10):6547–54. [DOI] [PubMed] [Google Scholar]
- 61.Bleakley M, Otterud BE, Richardt JL, Mollerup AD, Hudecek M, Nishida T, et al. Leukemia-associated minor histocompatibility antigen discovery using T-cell clones isolated by in vitro stimulation of naive CD8+ T cells. Blood. 2010;115(23):4923–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Chen BJ, Deoliveira D, Cui X, Le NT, Son J, Whitesides JF, et al. Inability of memory T cells to induce graft-versus-host disease is a result of an abortive alloresponse. Blood. 2006;109(7):3115–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Hill GR, Crawford JM, Cooke KR, Brinson YS, Pan L, Ferrara JL. Total body irradiation and acute graft-versus-host disease: the role of gastrointestinal damage and inflammatory cytokines. Blood, The Journal of the American Society of Hematology. 1997;90(8):3204–13. [PubMed] [Google Scholar]
- 64.Hill GR, Betts BC, Tkachev V, Kean LS, Blazar BR. Current Concepts and Advances in Graft-Versus-Host Disease Immunology. Annual Review of Immunology. 2021;39(1):19–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Burman AC, Banovic T, Kuns RD, Clouston AD, Stanley AC, Morris ES, et al. IFNγ differentially controls the development of idiopathic pneumonia syndrome and GVHD of the gastrointestinal tract. Blood. 2007;110(3):1064–72. [DOI] [PubMed] [Google Scholar]
- 66.Yi T, Chen Y, Wang L, Du G, Huang D, Zhao D, et al. Reciprocal differentiation and tissue-specific pathogenesis of Th1, Th2, and Th17 cells in graft-versus-host disease. Blood. 2009;114(14):3101–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Yu Y, Wang D, Liu C, Kaosaard K, Semple K, Anasetti C, et al. Prevention of GVHD while sparing GVL effect by targeting Th1 and Th17 transcription factor T-bet and RORγt in mice. Blood. 2011;118(18):5011–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Jiang Q, Yang G, Xiao F, Xie J, Wang S, Lu L, et al. Role of Th22 Cells in the Pathogenesis of Autoimmune Diseases. Frontiers in Immunology. 2021;12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Gartlan KH, Bommiasamy H, Paz K, Wilkinson AN, Owen M, Reichenbach DK, et al. A critical role for donor-derived IL-22 in cutaneous chronic GVHD. American Journal of Transplantation. 2018;18(4):810–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Couturier M, Lamarthee B, Arbez J, Renauld J-C, Bossard C, Malard F, et al. IL-22 deficiency in donor T cells attenuates murine acute graft-versus-host disease mortality while sparing the graft-versus-leukemia effect. Leukemia. 2013;27(7):1527–37. [DOI] [PubMed] [Google Scholar]
- 71.Ni M, Wang L, Ding Y, Gong W, Wang S, Neuber B, et al. Response to extracorporeal photopheresis therapy of patients with steroid-refractory/-resistant GvHD is associated with up-regulation of Th22 cells and Tfh cells. Cytotherapy. 2022;24(3):311–9. [DOI] [PubMed] [Google Scholar]
- 72.Hanash AM, Dudakov JA, Hua G, O’Connor MH, Young LF, Singer NV, et al. Interleukin-22 protects intestinal stem cells from immune-mediated tissue damage and regulates sensitivity to graft versus host disease. Immunity. 2012;37(2):339–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ponce DM, Alousi AM, Nakamura R, Slingerland J, Calafiore M, Sandhu KS, et al. A phase 2 study of interleukin-22 and systemic corticosteroids as initial treatment for acute GVHD of the lower GI tract. Blood. 2023;141(12):1389–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Allogeneic Peripheral Blood Stem-Cell Compared With Bone Marrow Transplantation in the Management of Hematologic Malignancies: An Individual Patient Data Meta-Analysis of Nine Randomized Trials. Journal of Clinical Oncology. 2005;23(22):5074–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.McCurdy SR, Kanakry CG, Tsai H-L, Gojo I, Smith BD, Gladstone DE, et al. Development of Grade II Acute Graft-versus-Host Disease Is Associated with Improved Survival after Myeloablative HLA-Matched Bone Marrow Transplantation using Single-Agent Post-Transplant Cyclophosphamide. Biology of Blood and Marrow Transplantation. 2019;25(6):1128–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Nagler A, Labopin M, Shimoni A, Niederwieser D, Mufti GJ, Zander AR, et al. Mobilized Peripheral Blood Stem Cells Compared with Bone Marrow as the Stem Cell Source for Unrelated Donor Allogeneic Transplantation with Reduced-Intensity Conditioning in Patients with Acute Myeloid Leukemia in Complete Remission: An Analysis from the. Biology of Blood and Marrow Transplantation. 2012;18(9):1422–9. [DOI] [PubMed] [Google Scholar]
- 77.Jo T, Arai Y, Kondo T, Mizuno S, Hirabayashi S, Inamoto Y, et al. Advantages of peripheral blood stem cells from unrelated donors versus bone marrow transplants in outcomes of adult acute myeloid leukemia patients. Cytotherapy. 2022;24(10):1013–25. [DOI] [PubMed] [Google Scholar]
- 78.Marmont A, Horowitz M, Gale R, Sobocinski K, Ash R, van Bekkum D, et al. T-cell depletion of HLA-identical transplants in leukemia. Blood. 1991;78(8):2120–30. [PubMed] [Google Scholar]
- 79.Champlin RE, Passweg JR, Zhang M-J, Rowlings PA, Pelz CJ, Atkinson KA, et al. T-cell depletion of bone marrow transplants for leukemia from donors other than HLA-identical siblings: advantage of T-cell antibodies with narrow specificities. Blood. 2000;95(12):3996–4003. [PubMed] [Google Scholar]
- 80.Ho VT, Soiffer RJ. The history and future of T-cell depletion as graft-versus-host disease prophylaxis for allogeneic hematopoietic stem cell transplantation. Blood. 2001;98(12):3192–204. [DOI] [PubMed] [Google Scholar]
- 81.Keever C, Small T, Flomenberg N, Heller G, Pekle K, Black P, et al. Immune reconstitution following bone marrow transplantation: comparison of recipients of T-cell depleted marrow with recipients of conventional marrow grafts. Blood. 1989;73(5):1340–50. [PubMed] [Google Scholar]
- 82.Wu CJ, Chillemi A, Alyea EP, Orsini E, Neuberg D, Soiffer RJ, et al. Reconstitution of T-cell receptor repertoire diversity following T-cell depleted allogeneic bone marrow transplantation is related to hematopoietic chimerism. Blood. 2000;95(1):352–9. [PubMed] [Google Scholar]
- 83.Devine SM, Carter S, Soiffer RJ, Pasquini MC, Hari PN, Stein A, et al. Low Risk of Chronic Graft-versus-Host Disease and Relapse Associated with T Cell–Depleted Peripheral Blood Stem Cell Transplantation for Acute Myelogenous Leukemia in First Remission: Results of the Blood and Marrow Transplant Clinical Trials Network Protocol 0303. Biology of Blood and Marrow Transplantation. 2011;17(9):1343–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Abdelhakim H, Abdel-Azim H, Saad A. Role of αβ T Cell Depletion in Prevention of Graft versus Host Disease. Biomedicines. 2017;5(4):35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Bleakley M, Sehgal A, Seropian S, Biernacki MA, Krakow EF, Dahlberg A, et al. Naive T-Cell Depletion to Prevent Chronic Graft-Versus-Host Disease. Journal of Clinical Oncology. 2022;40(11):1174–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Aschan J, Ringden O, Sundberg B, Gahrton G, Ljungman P, Winiarski J. Methotrexate combined with cyclosporin A decreases graft-versus-host disease, but increases leukemic relapse compared to monotherapy. Bone marrow transplantation. 1991;7(2):113–9. [PubMed] [Google Scholar]
- 87.Aschan J, Ringden O, Sundberg B, Klaesson S, Ljungman P, Lönnqvist B. Increased risk of relapse in patients with chronic myelogenous leukemia given T-cell depleted marrow compared to methotrexate combined with cyclosporin or monotherapy for the prevention of graft-versus-host disease. European journal of haematology. 1993;50(5):269–74. [DOI] [PubMed] [Google Scholar]
- 88.Baron F, Zachée P, Maertens J, Kerre T, Ory A, Seidel L, et al. Non-myeloablative allogeneic hematopoietic cell transplantation following fludarabine plus 2 Gy TBI or ATG plus 8 Gy TLI: a phase II randomized study from the Belgian Hematological Society. Journal of Hematology & Oncology. 2015;8(1):4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Storek J, Mohty M, Boelens JJ. Rabbit Anti–T Cell Globulin in Allogeneic Hematopoietic Cell Transplantation. Biology of Blood and Marrow Transplantation. 2015;21(6):959–70. [DOI] [PubMed] [Google Scholar]
- 90.Naeije L, Kariminia A, Abdossamadi S, Azadpour S, Subrt P, Kuzeljevic B, et al. Anti-Thymocyte Globulin Prophylaxis Induces a Decrease in Naive Th Cells to Inhibit the Onset of Chronic Graft-versus-Host Disease: Results from the Canadian Bone Marrow Transplant Group (CBMTG) 0801 Study. Biology of Blood and Marrow Transplantation. 2020;26(3):438–44. [DOI] [PubMed] [Google Scholar]
- 91.Dalle J-H, Rk Dardari, Menezes J, Cordeiro P, Champagne MA, Duval M. Binding of Thymoglobulin to Natural Killer Cells Leads to Cell Activation and Interferon-γ Production. Transplantation. 2009;87(4). [DOI] [PubMed] [Google Scholar]
- 92.Penack O, Fischer L, Stroux A, Gentilini C, Nogai A, Muessig A, et al. Serotherapy with thymoglobulin and alemtuzumab differentially influences frequency and function of natural killer cells after allogeneic stem cell transplantation. Bone Marrow Transplant. 2008;41(4):377–83. [DOI] [PubMed] [Google Scholar]
- 93.Mohty M Mechanisms of action of antithymocyte globulin: T-cell depletion and beyond. Leukemia. 2007;21(7):1387–94. [DOI] [PubMed] [Google Scholar]
- 94.Kekre N, Antin JH. ATG in allogeneic stem cell transplantation: standard of care in 2017? Counterpoint. Blood Advances. 2017;1(9):573–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Vo PT, Pantin J, Ramos C, Cook L, Cho E, Kurlander R, et al. Conditioning with rabbit versus horse ATG dramatically alters clinical outcomes in identical twins with severe aplastic anemia transplanted with the same allogeneic donor. J Hematol Oncol. 2015;8:78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Kröger N, Solano C, Wolschke C, Bandini G, Patriarca F, Pini M, et al. Antilymphocyte Globulin for Prevention of Chronic Graft-versus-Host Disease. New England Journal of Medicine. 2016;374(1):43–53. [DOI] [PubMed] [Google Scholar]
- 97.Bonifazi F, Solano C, Wolschke C, Sessa M, Patriarca F, Zallio F, et al. GVHD prophylaxis plus ATLG after myeloablative allogeneic haemopoietic peripheral blood stem-cell transplantation from HLA-identical siblings in patients with acute leukaemia in remission: final results of quality of life and long-term outcome analysis of a phase 3 randomised study. The Lancet Haematology. 2019;6(2):e89–e99. [DOI] [PubMed] [Google Scholar]
- 98.Finke J, Bethge WA, Schmoor C, Ottinger HD, Stelljes M, Zander AR, et al. Standard graft-versus-host disease prophylaxis with or without anti-T-cell globulin in haematopoietic cell transplantation from matched unrelated donors: a randomised, open-label, multicentre phase 3 trial. The Lancet Oncology. 2009;10(9):855–64. [DOI] [PubMed] [Google Scholar]
- 99.Kumar A, Reljic T, Hamadani M, Mohty M, Kharfan-Dabaja MA. Antithymocyte globulin for graft-versus-host disease prophylaxis: an updated systematic review and meta-analysis. Bone Marrow Transplantation. 2019;54(7):1094–106. [DOI] [PubMed] [Google Scholar]
- 100.Huang X-J, Liu D-H, Liu K-Y, Xu L-P, Chen H, Han W, et al. Treatment of Acute Leukemia with Unmanipulated HLA-Mismatched/Haploidentical Blood and Bone Marrow Transplantation. Biology of Blood and Marrow Transplantation. 2009;15(2):257–65. [DOI] [PubMed] [Google Scholar]
- 101.Soiffer RJ, Kim HT, McGuirk J, Horwitz ME, Johnston L, Patnaik MM, et al. Prospective, Randomized, Double-Blind, Phase III Clinical Trial of Anti-T-Lymphocyte Globulin to Assess Impact on Chronic Graft-Versus-Host Disease-Free Survival in Patients Undergoing HLA-Matched Unrelated Myeloablative Hematopoietic Cell Transplantation. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2017;35(36):4003–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Lehnert S, Rybka WB. Amplification of the graft-versus-host reaction by cyclophosphamide: dependence on timing of drug administration. Bone Marrow Transplant. 1994;13(4):473–7. [PubMed] [Google Scholar]
- 103.Prigozhina TB, Elkin G, Khitrin S, Slavin S. Prevention of acute graft-vs-host disease by a single low-dose cyclophosphamide injection following allogeneic bone marrow transplantation. Exp Hematol. 2008;36(12):1750–9. [DOI] [PubMed] [Google Scholar]
- 104.Luznik L, O’Donnell PV, Symons HJ, Chen AR, Leffell MS, Zahurak M, et al. HLA-Haploidentical Bone Marrow Transplantation for Hematologic Malignancies Using Nonmyeloablative Conditioning and High-Dose, Posttransplantation Cyclophosphamide. Biology of Blood and Marrow Transplantation. 2008;14(6):641–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Nunes NS, Kanakry CG. Mechanisms of Graft-versus-Host Disease Prevention by Post-transplantation Cyclophosphamide: An Evolving Understanding. Front Immunol. 2019;10:2668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Hadjis AD, Nunes NS, Khan SM, Fletcher RE, Pohl AP, Venzon DJ, et al. Post-Transplantation Cyclophosphamide Uniquely Restrains Alloreactive CD4(+) T-Cell Proliferation and Differentiation After Murine MHC-Haploidentical Hematopoietic Cell Transplantation. Front Immunol. 2022;13:796349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Iwamoto M, Ikegawa S, Kondo T, Meguri Y, Nakamura M, Sando Y, et al. Post-transplantation cyclophosphamide restores early B-cell lymphogenesis that suppresses subsequent chronic graft-versus-host disease. Bone Marrow Transplantation. 2021;56(4):956–9. [DOI] [PubMed] [Google Scholar]
- 108.Ghosh N, Karmali R, Rocha V, Ahn KW, DiGilio A, Hari PN, et al. Reduced-intensity transplantation for lymphomas using haploidentical related donors versus HLA-matched sibling donors: a Center for International Blood and Marrow Transplant Research analysis. Journal of Clinical Oncology. 2016;34(26):3141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Hoover A, O’Leary D, Cao Q, Gupta AO, Ebens CL, Maakaron JE, et al. Phase II Study of Myeloablative 8/8- or 7/8-Matched Allotransplantation with Post-Transplant Cyclophosphamide, Tacrolimus, and Mycophenolate Mofetil: Marked Reduction in Gvhd Risk without Increased Relapse Risk Compared to Historical Cyclosporine/Methotrexate. Blood. 2022;140(Supplement 1):282–3. [Google Scholar]
- 110.Jurdi NE, Hoover A, O’Leary D, Cao Q, Gupta A, Ebens C, et al. Phase II Study of Myeloablative 8/8- or 7/8-Matched Allotransplantation with Post-Transplant Cyclophosphamide, Tacrolimus, and Mycophenolate Mofetil: Marked Reduction in GVHD Risk Without Increased Relapse Risk Compared to Historical Cyclosporine/Methotrexate. medRxiv. 2023. [Google Scholar]
- 111.Luznik L, Pasquini MC, Logan B, Soiffer RJ, Wu J, Devine SM, et al. Randomized Phase III BMT CTN Trial of Calcineurin Inhibitor–Free Chronic Graft-Versus-Host Disease Interventions in Myeloablative Hematopoietic Cell Transplantation for Hematologic Malignancies. Journal of Clinical Oncology. 2022;40(4):356–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Holtan SG, Hamadani M, WU J, AL Malki MM, Runaas L, Elmariah H, et al. Post-Transplant Cyclophosphamide, Tacrolimus, and Mycophenolate Mofetil As the New Standard for Graft-Versus-Host Disease (GVHD) Prophylaxis in Reduced Intensity Conditioning: Results from Phase III BMT CTN 1703. Blood. 2022;140(Supplement 2):LBA-4-LBA-. [Google Scholar]
- 113.Shimoni A, Labopin M, Angelucci E, Blaise D, Ciceri F, Koc Y, et al. The association of graft-versus-leukemia effect and graft-versus host disease in haploidentical transplantation with post-transplant cyclophosphamide for AML. Bone Marrow Transplant. 2022;57(3):384–90. [DOI] [PubMed] [Google Scholar]
- 114.Baron F, Labopin M, Tischer J, Raiola AM, Vydra J, Blaise D, et al. GVHD occurrence does not reduce AML relapse following PTCy-based haploidentical transplantation: a study from the ALWP of the EBMT. Journal of Hematology & Oncology. 2023;16(1):10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.McCurdy SR, Kasamon YL, Kanakry CG, Bolaños-Meade J, Tsai HL, Showel MM, et al. Comparable composite endpoints after HLA-matched and HLA-haploidentical transplantation with post-transplantation cyclophosphamide. Haematologica. 2017;102(2):391–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.McCurdy SR, Radojcic V, Tsai H-L, Vulic A, Thompson E, Ivcevic S, et al. Signatures of GVHD and relapse after posttransplant cyclophosphamide revealed by immune profiling and machine learning. Blood, The Journal of the American Society of Hematology. 2022;139(4):608–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Stokes J, Hoffman EA, Zeng Y, Larmonier N, Katsanis E. Post-transplant bendamustine reduces GvHD while preserving GvL in experimental haploidentical bone marrow transplantation. British Journal of Haematology. 2016;174(1):102–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Katsanis E, Maher K, Roe DJ, Simpson RJ. Progressive substitution of posttransplant cyclophosphamide with bendamustine: A phase I study in haploidentical bone marrow transplantation. EJHaem. 2020;1(1):286–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Moiseev I, Bondarenko S, Morozova E, Vlasova Y, Dotsenko A, Epifanovskaya O, et al. Graftversus-Host Disease Prophylaxis with Post-Transplantation Bendamustine in Patients with Refractory Acute Leukemia: A Dose-Ranging Study. Transplantation and Cellular Therapy. 2021;27(7):601.e1–e7. [DOI] [PubMed] [Google Scholar]
- 120.Guo W-w, Su X-h, Wang M-y, Han M-z, Feng X-m, Jiang E-l. Regulatory T Cells in GVHD Therapy. Frontiers in Immunology. 2021;12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Leveque-El mouttie L, Koyama M, Le Texier L, Markey KA, Cheong M, Kuns RD, et al. Corruption of dendritic cell antigen presentation during acute GVHD leads to regulatory T-cell failure and chronic GVHD. Blood. 2016;128(6):794–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Matsuoka K-i, Koreth J, Kim HT, Bascug G, McDonough S, Kawano Y, et al. Low-Dose Interleukin-2 Therapy Restores Regulatory T Cell Homeostasis in Patients with Chronic Graft-Versus-Host Disease. Science Translational Medicine. 2013;5(179):179ra43–ra43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Koreth J, Matsuoka K-I, Kim HT, McDonough SM, Bindra B, Alyea EP, et al. Interleukin-2 and Regulatory T Cells in Graft-versus-Host Disease. New England Journal of Medicine. 2011;365(22):2055–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Whangbo JS, Kim HT, Mirkovic N, Leonard L, Poryanda S, Silverstein S, et al. Dose-escalated interleukin-2 therapy for refractory chronic graft-versus-host disease in adults and children. Blood Adv. 2019;3(17):2550–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Kennedy-Nasser AA, Ku S, Castillo-Caro P, Hazrat Y, Wu M-F, Liu H, et al. Ultra Low-Dose IL-2 for GVHD Prophylaxis after Allogeneic Hematopoietic Stem Cell Transplantation Mediates Expansion of Regulatory T Cells without Diminishing Antiviral and Antileukemic Activity. Clinical Cancer Research. 2014;20(8):2215–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Zhao X-Y, Zhao X-S, Wang Y-T, Chen Y-H, Xu L-P, Zhang X-H, et al. Prophylactic use of low-dose interleukin-2 and the clinical outcomes of hematopoietic stem cell transplantation: A randomized study. OncoImmunology. 2016;5(12):e1250992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Brunstein CG, Miller JS, McKenna DH, Hippen KL, DeFor TE, Sumstad D, et al. Umbilical cord blood–derived T regulatory cells to prevent GVHD: kinetics, toxicity profile, and clinical effect. Blood. 2016;127(8):1044–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.MacMillan ML, Hippen KL, McKenna DH, Kadidlo D, Sumstad D, DeFor TE, et al. First-in-human phase 1 trial of induced regulatory T cells for graft-versus-host disease prophylaxis in HLA-matched siblings. Blood Advances. 2021;5(5):1425–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Peck A, Mellins ED. Plasticity of T-cell phenotype and function: the T helper type 17 example. Immunology. 2010;129(2):147–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Zanin-Zhorov A, Blazar BR. ROCK2, a critical regulator of immune modulation and fibrosis has emerged as a therapeutic target in chronic graft-versus-host disease. Clinical Immunology. 2021;230:108823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Shlomchik WD, Couzens MS, Tang CB, McNiff J, Robert ME, Liu J, et al. Prevention of graft versus host disease by inactivation of host antigen-presenting cells. Science. 1999;285(5426):412–5. [DOI] [PubMed] [Google Scholar]
- 132.Koyama M, Kuns RD, Olver SD, Raffelt NC, Wilson YA, Don ALJ, et al. Recipient nonhematopoietic antigen-presenting cells are sufficient to induce lethal acute graft-versus-host disease. Nature Medicine. 2012;18(1):135–42. [DOI] [PubMed] [Google Scholar]
- 133.Toubai T, Tawara I, Sun Y, Liu C, Nieves E, Evers R, et al. Induction of acute GVHD by sex-mismatched H-Y antigens in the absence of functional radiosensitive host hematopoietic–derived antigen-presenting cells. Blood. 2012;119(16):3844–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Markey KA, Schluter J, Gomes ALC, Littmann ER, Pickard AJ, Taylor BP, et al. The micro-bederived short-chain fatty acids butyrate and propionate are associated with protection from chronic GVHD. Blood. 2020;136(1):130–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Koyama M, Mukhopadhyay P, Schuster IS, Henden AS, Hülsdünker J, Varelias A, et al. MHC Class II Antigen Presentation by the Intestinal Epithelium Initiates Graft-versus-Host Disease and Is Influenced by the Microbiota. Immunity. 2019;51(5):885–98.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Zhang Y, Shlomchik WD, Joe G, Louboutin J-P, Zhu J, Rivera A, et al. APCs in the Liver and Spleen Recruit Activated Allogeneic CD8+ T Cells to Elicit Hepatic Graft-Versus-Host Disease1. The Journal of Immunology. 2002;169(12):7111–8. [DOI] [PubMed] [Google Scholar]
- 137.Koyama M, Cheong M, Markey KA, Gartlan KH, Kuns RD, Locke KR, et al. Donor colonic CD103+ dendritic cells determine the severity of acute graft-versus-host disease. Journal of Experimental Medicine. 2015;212(8):1303–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Koyama M, Hill GR. Alloantigen presentation and graft-versus-host disease: fuel for the fire. Blood. 2016;127(24):2963–70. [DOI] [PubMed] [Google Scholar]
- 139.Li H, Demetris AJ, McNiff J, Matte-Martone C, Tan HS, Rothstein DM, et al. Profound Depletion of Host Conventional Dendritic Cells, Plasmacytoid Dendritic Cells, and B Cells Does Not Prevent Graft-versus-Host Disease Induction. The Journal of Immunology. 2012;188(8):3804–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Matte CC, Liu J, Cormier J, Anderson BE, Athanasiadis I, Jain D, et al. Donor APCs are required for maximal GVHD but not for GVL. Nature Medicine. 2004;10(9):987–92. [DOI] [PubMed] [Google Scholar]
- 141.Li JM, Waller EK. Donor antigen-presenting cells regulate T-cell expansion and antitumor activity after allogeneic bone marrow transplantation. Biol Blood Marrow Transplant. 2004;10(8):540–51. [DOI] [PubMed] [Google Scholar]
- 142.Markey KA, Gartlan KH, Kuns RD, Lane SW, Hill GR. Conventional dendritic cells are required for the cross-presentation of leukemia-specific antigen in a model of AML relapse post-BMT. Bone Marrow Transplantation. 2018;53(6):800–3. [DOI] [PubMed] [Google Scholar]
- 143.Kuzmina Z, Eder S, Böhm A, Pernicka E, Vormittag L, Kalhs P, et al. Significantly worse survival of patients with NIH-defined chronic graft-versus-host disease and thrombocytopenia or progressive onset type: results of a prospective study. Leukemia. 2012;26(4):746–56. [DOI] [PubMed] [Google Scholar]
- 144.Khoder A, Alsuliman A, Basar R, Sobieski C, Kondo K, Alousi AM, et al. Evidence for B Cell Exhaustion in Chronic Graft-versus-Host Disease. Frontiers in Immunology. 2018;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Cooke KR, Luznik L, Sarantopoulos S, Hakim FT, Jagasia M, Fowler DH, et al. The Biology of Chronic Graft-versus-Host Disease: A Task Force Report from the National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease. Biology of Blood and Marrow Transplantation. 2017;23(2):211–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Sarantopoulos S, Stevenson KE, Kim HT, Bhuiya NS, Cutler CS, Soiffer RJ, et al. High Levels of B-Cell Activating Factor in Patients with Active Chronic Graft-Versus-Host Disease. Clinical Cancer Research. 2007;13(20):6107–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Sarantopoulos S, Stevenson KE, Kim HT, Cutler CS, Bhuiya NS, Schowalter M, et al. Altered B-cell homeostasis and excess BAFF in human chronic graft-versus-host disease. Blood. 2009;113(16):3865–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Kuzmina Z, Krenn K, Petkov V, Körmöczi U, Weigl R, Rottal A, et al. CD19+CD21low B cells and patients at risk for NIH-defined chronic graft-versus-host disease with bronchiolitis obliterans syndrome. Blood. 2013;121(10):1886–95. [DOI] [PubMed] [Google Scholar]
- 149.Zeiser R, Sarantopoulos S, Blazar BR. B-cell targeting in chronic graft-versus-host disease. Blood. 2018;131(13):1399–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Miklos D, Cutler CS, Arora M, Waller EK, Jagasia M, Pusic I, et al. Ibrutinib for chronic graft-versus-host disease after failure of prior therapy. Blood. 2017;130(21):2243–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Lin C, DiCioccio RA, Haykal T, McManigle WC, Li Z, Anand SM, et al. A Phase I Trial of SYK Inhibition with Fostamatinib in the Prevention and Treatment of Chronic Graft-Versus-Host Disease. Transplantation and Cellular Therapy. 2023;29(3):179.e1–e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Ritz J Role of B Cells in Graft-Versus-Leukemia and Graft-Versus-Host Disease. Blood. 2013;122(21):SCI-3-SCI-. [Google Scholar]
- 153.Simonetta F, Alvarez M, Negrin RS. Natural Killer Cells in Graft-versus-Host-Disease after Allogeneic Hematopoietic Cell Transplantation. Front Immunol. 2017;8:465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Palmer JM, Rajasekaran K, Thakar MS, Malarkannan S. Clinical Relevance of Natural Killer Cells Following Hematopoietic Stem Cell Transplantation. Journal of Cancer. 2013;4(1):25–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Ruggeri L, Capanni M, Urbani E, Perruccio K, Shlomchik WD, Tosti A, et al. Effectiveness of Donor Natural Killer Cell Alloreactivity in Mismatched Hematopoietic Transplants. Science. 2002;295(5562):2097–100. [DOI] [PubMed] [Google Scholar]
- 156.Bednarski JJ, Zimmerman C, Berrien-Elliott MM, Foltz JA, Becker-Hapak M, Neal CC, et al. Donor memory-like NK cells persist and induce remissions in pediatric patients with relapsed AML after transplant. Blood. 2022;139(11):1670–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Shapiro RM, Birch GC, Hu G, Vergara Cadavid J, Nikiforow S, Baginska J, et al. Expansion, persistence, and efficacy of donor memory-like NK cells infused for posttransplant relapse. Journal of Clinical Investigation. 2022;132(11). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Gao F, Ye Y, Gao Y, Huang H, Zhao Y. Influence of KIR and NK Cell Reconstitution in the Outcomes of Hematopoietic Stem Cell Transplantation. Frontiers in Immunology. 2020;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Olson JA, Leveson-Gower DB, Gill S, Baker J, Beilhack A, Negrin RS. NK cells mediate reduction of GVHD by inhibiting activated, alloreactive T cells while retaining GVT effects. Blood. 2010;115(21):4293–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Ullrich E, Salzmann-Manrique E, Bakhtiar S, Bremm M, Gerstner S, Herrmann E, et al. Relation between Acute GVHD and NK Cell Subset Reconstitution Following Allogeneic Stem Cell Transplantation. Frontiers in Immunology. 2016;7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Huenecke S, Cappel C, Esser R, Pfirrmann V, Salzmann-Manrique E, Betz S, et al. Development of Three Different NK Cell Subpopulations during Immune Reconstitution after Pediatric Allogeneic Hematopoietic Stem Cell Transplantation: Prognostic Markers in GvHD and Viral Infections. Front Immunol. 2017;8:109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Nalle SC, Aimee Kwak H, Edelblum KL, Joseph NE, Singh G, Khramtsova GF, et al. Recipient NK cell inactivation and intestinal barrier loss are required for MHC-matched graft-versus-host disease. Science Translational Medicine. 2014;6(243):243ra87–ra87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Gaballa A, Arruda LCM, Uhlin M. Gamma delta T-cell reconstitution after allogeneic HCT: A platform for cell therapy. Frontiers in Immunology. 2022;13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Handgretinger R, Schilbach K. The potential role of γδ T cells after allogeneic HCT for leukemia. Blood. 2018;131(10):1063–72. [DOI] [PubMed] [Google Scholar]
- 165.Merli P, Algeri M, Galaverna F, Milano GM, Bertaina V, Biagini S, et al. Immune Modulation Properties of Zoledronic Acid on TcRγδ T-Lymphocytes After TcRαβ/CD19-Depleted Haploidentical Stem Cell Transplantation: An analysis on 46 Pediatric Patients Affected by Acute Leukemia. Front Immunol. 2020;11:699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Cela ME, Holladay MS, Rooney CM, Richardson S, Alexander B, Krance RA, et al. Gamma delta T lymphocyte regeneration after T lymphocyte-depleted bone marrow transplantation from mismatched family members or matched unrelated donors. Bone marrow transplantation. 1996;17(2):243–7. [PubMed] [Google Scholar]
- 167.Drobyski WR, Majewski D, Hanson G. Graft-facilitating doses of ex vivo activated gammadelta T cells do not cause lethal murine graft-vs.-host disease. Biology of Blood and Marrow Transplantation. 1999;5(4):222–30. [DOI] [PubMed] [Google Scholar]
- 168.Lamb LS Jr, Gee AP, Hazlett LJ, Musk P, Parrish RS, O’Hanlon TP, et al. Influence of T Cell Depletion Method on Circulating γδ T Cell Reconstitution and Potential Role in the Graft-VersusLeukemia Effect. Cytotherapy. 1999;1(1):7–19. [DOI] [PubMed] [Google Scholar]
- 169.Godder KT, Henslee-Downey PJ, Mehta J, Park BS, Chiang KY, Abhyankar S, et al. Long term disease-free survival in acute leukemia patients recovering with increased γδ T cells after partially mismatched related donor bone marrow transplantation. Bone Marrow Transplantation. 2007;39(12):751–7. [DOI] [PubMed] [Google Scholar]
- 170.Klyuchnikov E, Badbaran A, Massoud R, Fritsche-Friedland U, Janson D, Ayuk F, et al. Enhanced Immune Reconstitution of γδ T Cells after Allogeneic Stem Cell Transplantation Overcomes the Negative Impact of Pretransplantation Minimal Residual Disease-Positive Status in Patients with Acute Myelogenous Leukemia. Transplantation and Cellular Therapy. 2021;27(10):841–50. [DOI] [PubMed] [Google Scholar]
- 171.Perko R, Kang G, Sunkara A, Leung W, Thomas PG, Dallas MH. Gamma Delta T Cell Reconstitution Is Associated with Fewer Infections and Improved Event-Free Survival after Hematopoietic Stem Cell Transplantation for Pediatric Leukemia. Biology of Blood and Marrow Transplantation. 2015;21(1):130–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Arruda LCM, Gaballa A, Uhlin M. Impact of γδ T cells on clinical outcome of hematopoietic stem cell transplantation: systematic review and meta-analysis. Blood Advances. 2019;3(21):3436–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Saura-Esteller J, de Jong M, King LA, Ensing E, Winograd B, de Gruijl TD, et al. Gamma Delta T-Cell Based Cancer Immunotherapy: Past-Present-Future. Frontiers in Immunology. 2022;13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Oran B, Champlin RE, Wang F, Tanaka T, Saliba RM, Al-Atrash G, et al. Donor clonal hematopoiesis increases risk of acute graft versus host disease after matched sibling transplantation. Leukemia. 2022;36(1):257–62. [DOI] [PubMed] [Google Scholar]
- 175.Frick M, Chan W, Arends CM, Hablesreiter R, Halik A, Heuser M, et al. Role of Donor Clonal Hematopoiesis in Allogeneic Hematopoietic Stem-Cell Transplantation. Journal of Clinical Oncology. 2019;37(5):375–85. [DOI] [PubMed] [Google Scholar]
- 176.Gibson CJ, Kim HT, Zhao L, Murdock HM, Hambley B, Ogata A, et al. Donor Clonal Hematopoiesis and Recipient Outcomes After Transplantation. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2022;40(2):189–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Newell LF, Williams T, Liu J, Yu Y, Chen Y, Booth GC, et al. Engrafted Donor-Derived Clonal Hematopoiesis after Allogenic Hematopoietic Cell Transplantation is Associated with Chronic Graft-versus-Host Disease Requiring Immunosuppressive Therapy, but no Adverse Impact on Overall Survival or Relapse. Transplant Cell Ther. 2021;27(8):662.e1–e9. [DOI] [PubMed] [Google Scholar]
- 178.Boettcher S, Wilk CM, Singer J, Beier F, Burcklen E, Beisel C, et al. Clonal hematopoiesis in donors and long-term survivors of related allogeneic hematopoietic stem cell transplantation. Blood. 2020;135(18):1548–59. [DOI] [PubMed] [Google Scholar]
- 179.Wang R, Yang X, Liu J, Zhong F, Zhang C, Chen Y, et al. Gut microbiota regulates acute myeloid leukaemia via alteration of intestinal barrier function mediated by butyrate. Nature Communications. 2022;13(1):2522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Beelen DW, Elmaagacli A, Müller KD, Hirche H, Schaefer UW. Influence of intestinal bacterial decontamination using metronidazole and ciprofloxacin or ciprofloxacin alone on the development of acute graft-versus-host disease after marrow transplantation in patients with hematologic malignancies: final results and long-term follow-up of an open-label prospective randomized trial. Blood. 1999;93(10):3267–75. [PubMed] [Google Scholar]
- 181.Severyn CJ, Siranosian BA, Kong ST, Moreno A, Li MM, Chen N, et al. Microbiota dynamics in a randomized trial of gut decontamination during allogeneic hematopoietic cell transplantation. JCI Insight. 2022;7(7). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Jenq RR, Taur Y, Devlin SM, Ponce DM, Goldberg JD, Ahr KF, et al. Intestinal Blautia Is Associated with Reduced Death from Graft-versus-Host Disease. Biol Blood Marrow Transplant. 2015;21(8):1373–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Simms-Waldrip TR, Sunkersett G, Coughlin LA, Savani MR, Arana C, Kim J, et al. Antibiotic-Induced Depletion of Anti-inflammatory Clostridia Is Associated with the Development of Graft-versus-Host Disease in Pediatric Stem Cell Transplantation Patients. Biology of Blood and Marrow Transplantation. 2017;23(5):820–9. [DOI] [PubMed] [Google Scholar]
- 184.Biernat MM, Urbaniak-Kujda D, Dybko J, Kapelko-Słowik K, Prajs I, Wróbel T. Fecal microbiota transplantation in the treatment of intestinal steroid-resistant graft-versus-host disease: two case reports and a review of the literature. J Int Med Res. 2020;48(6):300060520925693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Gu Y, Sun J, Li K, Wu X, Zhang J. Alteration in the Skin Microbiome in Cutaneous Graft Versus Host Disease. Acta Derm Venereol. 2021;101(1):adv00374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Fredricks DN. The gut microbiota and graft-versus-host disease. Journal of Clinical Investigation. 2019;129(5):1808–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.van der Velden WJ, Plantinga TS, Feuth T, Donnelly JP, Netea MG, Blijlevens NM. The incidence of acute graft-versus-host disease increases with Candida colonization depending the dectin-1 gene status. Clin Immunol. 2010;136(2):302–6. [DOI] [PubMed] [Google Scholar]
- 188.Bejanyan N, Weisdorf DJ, Logan BR, Wang H-L, Devine SM, De Lima M, et al. Survival of Patients with Acute Myeloid Leukemia Relapsing after Allogeneic Hematopoietic Cell Transplantation: A Center for International Blood and Marrow Transplant Research Study. Biology of Blood and Marrow Transplantation. 2015;21(3):454–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Kunadt D, Stölzel F. Effective Immunosurveillance After Allogeneic Hematopoietic Stem Cell Transplantation in Acute Myeloid Leukemia. Cancer Manag Res. 2021;13:7411–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Schmid C, Labopin M, Schaap N, Veelken H, Brecht A, Stadler M, et al. Long-term results and GvHD after prophylactic and preemptive donor lymphocyte infusion after allogeneic stem cell transplantation for acute leukemia. Bone Marrow Transplantation. 2022;57(2):215–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Pelot MR, Pearson DA, Swenson K, Zhao G, Sachs J, Yang Y-G, et al. Lymphohematopoietic graft-vs.-host reactions can be induced without graft-vs.-host disease in murine mixed chimeras established with a cyclophosphamide-based nonmyeloablative conditioning regimen. Biology of Blood and Marrow Transplantation. 1999;5(3):133–43. [DOI] [PubMed] [Google Scholar]
- 192.Mapara MY, Kim Y-M, Marx J, Sykes M. Donor lymphocyte infusion-mediated graft-versus-leukemia effects in mixed chimeras established with a nonmyeloablative conditioning regimen: extinction of graft-versus-leukemia effects after conversion to full donor chimerism1. Transplantation. 2003;76(2):297–305. [DOI] [PubMed] [Google Scholar]
- 193.Jedlickova Z, Schmid C, Koenecke C, Hertenstein B, Baurmann H, Schwerdtfeger R, et al. Long-term results of adjuvant donor lymphocyte transfusion in AML after allogeneic stem cell transplantation. Bone Marrow Transplantation. 2016;51(5):663–7. [DOI] [PubMed] [Google Scholar]
- 194.Wang Y, Liu D-H, Fan Z-P, Sun J, Wu X-J, Ma X, et al. Prevention of relapse using DLI can increase survival following HLA-identical transplantation in patients with advanced-stage acute leukemia: a multi-center study. Clinical Transplantation. 2012;26(4):635–43. [DOI] [PubMed] [Google Scholar]
- 195.Schmid C, Labopin M, Schaap N, Veelken H, Schleuning M, Stadler M, et al. Prophylactic donor lymphocyte infusion after allogeneic stem cell transplantation in acute leukaemia - a matched pair analysis by the Acute Leukaemia Working Party of EBMT. British Journal of Haematology. 2019;184(5):782–7. [DOI] [PubMed] [Google Scholar]
- 196.Goodyear OC, Dennis M, Jilani NY, Loke J, Siddique S, Ryan G, 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):3361–9. [DOI] [PubMed] [Google Scholar]
- 197.Schroeder T, Fröbel J, Cadeddu RP, Czibere A, Dienst A, Platzbecker U, et al. Salvage therapy with azacitidine increases regulatory T cells in peripheral blood of patients with AML or MDS and early relapse after allogeneic blood stem cell transplantation. Leukemia. 2013;27(9):1910–3. [DOI] [PubMed] [Google Scholar]
- 198.Santourlidis S, Trompeter HI, Weinhold S, Eisermann B, Meyer KL, Wernet P, et al. Crucial role of DNA methylation in determination of clonally distributed killer cell Ig-like receptor expression patterns in NK cells. J Immunol. 2002;169(8):4253–61. [DOI] [PubMed] [Google Scholar]
- 199.Sánchez-Abarca LI, Gutierrez-Cosio S, Santamaría C, Caballero-Velazquez T, Blanco B, Herrero-Sánchez C, et al. Immunomodulatory effect of 5-azacytidine (5-azaC): potential role in the transplantation setting. Blood. 2010;115(1):107–21. [DOI] [PubMed] [Google Scholar]
- 200.Pusic I, Choi J, Fiala MA, Gao F, Holt M, Cashen AF, et al. Maintenance Therapy with Decitabine after Allogeneic Stem Cell Transplantation for Acute Myelogenous Leukemia and Myelodysplastic Syndrome. Biol Blood Marrow Transplant. 2015;21(10):1761–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Craddock C, Jilani N, Siddique S, Yap C, Khan J, Nagra S, et al. Tolerability and Clinical Activity of Post-Transplantation Azacitidine in Patients Allografted for Acute Myeloid Leukemia Treated on the RICAZA Trial. Biol Blood Marrow Transplant. 2016;22(2):385–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Vij R, Le-Rademacher J, Laumann K, Hars V, Owzar K, Shore T, et al. A Phase II Multicenter Study of the Addition of Azacitidine to Reduced-Intensity Conditioning Allogeneic Transplant for High-Risk Myelodysplasia (and Older Patients with Acute Myeloid Leukemia): Results of CALGB 100801 (Alliance). Biol Blood Marrow Transplant. 2019;25(10):1984–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Keruakous AR, Holter-Chakrabarty J, Schmidt SA, Khawandanah MO, Selby G, Yuen C. Azacitidine maintenance therapy post-allogeneic stem cell transplantation in poor-risk acute myeloid leukemia. Hematol Oncol Stem Cell Ther. 2021. [DOI] [PubMed] [Google Scholar]
- 204.Oran B, de Lima M, Garcia-Manero G, Thall PF, Lin R, Popat U, et al. A phase 3 randomized study of 5-azacitidine maintenance vs observation after transplant in high-risk AML and MDS patients. Blood Advances. 2020;4(21):5580–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Gao L, Zhang Y, Wang S, Kong P, Su Y, Hu J, et al. Effect of rhG-CSF Combined With Decitabine Prophylaxis on Relapse of Patients With High-Risk MRD-Negative AML After HSCT: An Open-Label, Multicenter, Randomized Controlled Trial. Journal of Clinical Oncology. 2020;38(36):4249–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Kent A, Pollyea DA, Winters A, Jordan CT, Smith C, Gutman JA. Venetoclax Is Safe and Tolerable As Post-Transplant Maintenance Therapy for AML Patients at High Risk for Relapse. Blood. 2020;136:11–2. [DOI] [PubMed] [Google Scholar]
- 207.Wei Y, Xiong X, Li X, Lu W, He X, Jin X, et al. Low-dose decitabine plus venetoclax is safe and effective as post-transplant maintenance therapy for high-risk acute myeloid leukemia and myelodysplastic syndrome. Cancer Science. 2021;112(9):3636–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Craddock C, Platzbecker U, Heuser M, Pullarkat V, Chaudhury S, Wu D, et al. P561: VIALE-T: A RANDOMIZED, OPEN-LABEL, PHASE 3 STUDY OF VENETOCLAX IN COMBINATION WITH AZACITIDINE AFTER ALLOGENEIC STEM CELL TRANSPLANTATION IN PATIENTS WITH ACUTE MYELOID LEUKEMIA. HemaSphere. 2022;6. [Google Scholar]
- 209.Nayak RK, Chen Y-B. Maintenance therapy for AML after allogeneic HCT. Frontiers in Oncology. 2022;12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Xuan L, Wang Y, Huang F, Fan Z, Xu Y, Sun J, et al. Sorafenib maintenance in patients with FLT3-ITD acute myeloid leukaemia undergoing allogeneic haematopoietic stem-cell transplantation: an open-label, multicentre, randomised phase 3 trial. The Lancet Oncology. 2020;21(9):1201–12. [DOI] [PubMed] [Google Scholar]
- 211.Burchert A, Bug G, Fritz LV, Finke J, Stelljes M, Röllig C, et al. Sorafenib maintenance after allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia with FLT3–internal tandem duplication mutation (SORMAIN). Journal of Clinical Oncology. 2020;38(26):2993–3002. [DOI] [PubMed] [Google Scholar]
- 212.Maziarz RT, Levis M, Patnaik MM, Scott BL, Mohan SR, Deol A, et al. Midostaurin after allogeneic stem cell transplant in patients with FLT3-internal tandem duplication-positive acute myeloid leukemia. Bone Marrow Transplantation. 2021;56(5):1180–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Terao T, Matsuoka K-i, Ueda H, Matsumura A, Matsubara C, Kondo K, et al. Gilteritinib Maintenance Therapy Post-Allogenic Stem-Cell Transplantation Improves the Prognosis of Patients with FLT3-Mutated AML. Blood. 2022;140(Supplement 1):3290–1. [Google Scholar]
- 214.CTN APIB. Astellas and BMT CTN Announce Topline Results from Phase 3 MORPHO Trial of Gilteritinib 2023. [Available from: https://www.prnewswire.com/news-releases/astellas-and-bmt-ctn-announce-topline-results-from-phase-3-morpho-trial-of-gilteritinib-301767481.html.
- 215.Fathi AT, Kim HT, Soiffer RJ, Levis MJ, Li S, Kim AS, et al. Enasidenib as maintenance following allogeneic hematopoietic cell transplantation for IDH2-mutated myeloid malignancies. Blood Advances. 2022;6(22):5857–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Xu J, Niu T. Natural killer cell-based immunotherapy for acute myeloid leukemia. Journal of Hematology & Oncology. 2020;13(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Kulkarni UP, Arunachalam AK, Palani HK, Balasundaram N, Venkatraman A, Korula A, et al. Haploidentical Natural Killer Cell Therapy As an Adjunct to Stem Cell Transplantation for Refractory Acute Myeloid Leukemia. Blood. 2021;138(Supplement 1):3827–. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Lee DA, Denman CJ, Rondon G, Woodworth G, Chen J, Fisher T, et al. Haploidentical Natural Killer Cells Infused before Allogeneic Stem Cell Transplantation for Myeloid Malignancies: A Phase I Trial. Biology of Blood and Marrow Transplantation. 2016;22(7):1290–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Ciurea SO, Schafer JR, Bassett R, Denman CJ, Cao K, Willis D, et al. Phase 1 clinical trial using mbIL21 ex vivo–expanded donor-derived NK cells after haploidentical transplantation. Blood. 2017;130(16):1857–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Choi I, Yoon SR, Park S-Y, Kim H, Jung S-J, Jang YJ, et al. Donor-Derived Natural Killer Cells Infused after Human Leukocyte Antigen–Haploidentical Hematopoietic Cell Transplantation: A Dose-Escalation Study. Biology of Blood and Marrow Transplantation. 2014;20(5):696–704. [DOI] [PubMed] [Google Scholar]
- 221.Choi I, Yoon SR, Park S-Y, Kim H, Jung S-J, Kang Y-L, et al. Donor-Derived Natural Killer Cell Infusion after Human Leukocyte Antigen–Haploidentical Hematopoietic Cell Transplantation in Patients with Refractory Acute Leukemia. Biology of Blood and Marrow Transplantation. 2016;22(11):2065–76. [DOI] [PubMed] [Google Scholar]
- 222.Stern M, Passweg JR, Meyer-Monard S, Esser R, Tonn T, Soerensen J, et al. Pre-emptive immunotherapy with purified natural killer cells after haploidentical SCT: a prospective phase II study in two centers. Bone Marrow Transplantation. 2013;48(3):433–8. [DOI] [PubMed] [Google Scholar]
- 223.Motabi IH, Ghobadi A, Liu J, Schroeder M, Abboud CN, Cashen AF, et al. Chemotherapy versus Hypomethylating Agents for the Treatment of Relapsed Acute Myeloid Leukemia and Myelodysplastic Syndrome after Allogeneic Stem Cell Transplant. Biology of Blood and Marrow Transplantation. 2016;22(7):1324–9. [DOI] [PubMed] [Google Scholar]
- 224.Levine JE, Braun T, Penza SL, Beatty P, Cornetta K, Martino R, et al. Prospective trial of chemotherapy and donor leukocyte infusions for relapse of advanced myeloid malignancies after allogeneic stem-cell transplantation. Journal of Clinical Oncology. 2002;20(2):405–12. [DOI] [PubMed] [Google Scholar]
- 225.Devillier R, Crocchiolo R, Etienne A, Prebet T, Charbonnier A, Fürst S, et al. Outcome of relapse after allogeneic stem cell transplant in patients with acute myeloid leukemia. Leukemia & lymphoma. 2013;54(6):1228–34. [DOI] [PubMed] [Google Scholar]
- 226.Steinmann J, Bertz H, Wäsch R, Marks R, Zeiser R, Bogatyreva L, et al. 5-Azacytidine and DLI can induce long-term remissions in AML patients relapsed after allograft. Bone marrow transplantation. 2015;50(5):690–5. [DOI] [PubMed] [Google Scholar]
- 227.Ganguly S, Amin M, Divine C, Aljitawi OS, Abhyankar S, McGuirk JP. Decitabine in patients with relapsed acute myeloid leukemia (AML) after allogeneic stem cell transplantation (allo-SCT). Annals of hematology. 2013;92(4):549. [DOI] [PubMed] [Google Scholar]
- 228.Moukalled NM, El Darsa H, Haibe Y, Massoud R, Kanj SS, Mahfouz R, et al. Feasibility of Venetoclax-based combinations for adult patients with acute myeloid leukemia relapsing after allogenic stem cell transplantation. Bone Marrow Transplantation. 2019;54(4):620–4. [DOI] [PubMed] [Google Scholar]
- 229.Schuler E, Wagner-Drouet EM, Ajib S, Bug G, Crysandt M, Dressler S, et al. Treatment of myeloid malignancies relapsing after allogeneic hematopoietic stem cell transplantation with venetoclax and hypomethylating agents-a retrospective multicenter analysis on behalf of the German Cooperative Transplant Study Group. Ann Hematol. 2021;100(4):959–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Rettig AR, Ihorst G, Bertz H, Lübbert M, Marks R, Waterhouse M, et al. Donor lymphocyte infusions after first allogeneic hematopoietic stem-cell transplantation in adults with acute myeloid leukemia: a single-center landmark analysis. Ann Hematol. 2021;100(9):2339–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Schmid C, Labopin M, Nagler A, Bornhäuser M, Finke J, Fassas A, et al. 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. Journal of clinical oncology. 2007;25(31):4938–45. [DOI] [PubMed] [Google Scholar]
- 232.Mackinnon S, Papadopoulos EB, Carabasi MH, Reich L, Collins NH, Boulad F, et al. Adoptive immunotherapy evaluating escalating doses of donor leukocytes for relapse of chronic myeloid leukemia after bone marrow transplantation: separation of graft-versus-leukemia responses from graft-versus-host disease. Blood. 1995;86(4):1261–8. [PubMed] [Google Scholar]
- 233.Guglielmi C, Arcese W, Dazzi F, Brand R, Bunjes D, Verdonck LF, et al. Donor lymphocyte infusion for relapsed chronic myelogenous leukemia: prognostic relevance of the initial cell dose. Blood, The Journal of the American Society of Hematology. 2002;100(2):397–405. [DOI] [PubMed] [Google Scholar]
- 234.Orti G, Barba P, Fox L, Salamero O, Bosch F, Valcarcel D. Donor lymphocyte infusions in AML and MDS: Enhancing the graft-versus-leukemia effect. Experimental Hematology. 2017;48:1–11. [DOI] [PubMed] [Google Scholar]
- 235.Marks DI, Lush R, Cavenagh J, Milligan DW, Schey S, Parker A, et al. The toxicity and efficacy of donor lymphocyte infusions given after reduced-intensity conditioning allogeneic stem cell transplantation. Blood, The Journal of the American Society of Hematology. 2002;100(9):3108–14. [DOI] [PubMed] [Google Scholar]
- 236.Bar M, Sandmaier BM, Inamoto Y, Bruno B, Hari P, Chauncey T, et al. Donor Lymphocyte Infusion for Relapsed Hematological Malignancies after Allogeneic Hematopoietic Cell Transplantation: Prognostic Relevance of the Initial CD3+ T Cell Dose. Biology of Blood and Marrow Transplantation. 2013;19(6):949–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Miller JS, Weisdorf DJ, Burns LJ, Slungaard A, Wagner JE, Verneris MR, et al. Lymphodepletion followed by donor lymphocyte infusion (DLI) causes significantly more acute graft-versus-host disease than DLI alone. Blood, The Journal of the American Society of Hematology. 2007;110(7):2761–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Yang I, Weiss L, Abdul-Hai A, Kasir J, Reich S, Slavin S. Induction of early post-transplant graft-versus-leukemia effects using intentionally mismatched donor lymphocytes and elimination of alloantigen-primed donor lymphocytes for prevention of graft-versus-host disease. Cancer Res. 2005;65(21):9735–40. [DOI] [PubMed] [Google Scholar]
- 239.Shimoni A, Labopin M, Finke J, Ciceri F, Deconinck E, Kröger N, et al. Donor selection for a second allogeneic stem cell transplantation in AML patients relapsing after a first transplant: a study of the Acute Leukemia Working Party of EBMT. Blood Cancer Journal. 2019;9(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Toffalori C, Zito L, Gambacorta V, Riba M, Oliveira G, Bucci G, et al. Immune signature drives leukemia escape and relapse after hematopoietic cell transplantation. Nature Medicine. 2019;25(4):603–11. [DOI] [PubMed] [Google Scholar]
- 241.Yang X, Ma L, Zhang X, Huang L, Wei J. Targeting PD-1/PD-L1 pathway in myelodysplastic syndromes and acute myeloid leukemia. Experimental Hematology & Oncology. 2022;11(1):11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Oran B, Garcia-Manero G, Saliba RM, Alfayez M, Al-Atrash G, Ciurea SO, et al. Posttransplantation cyclophosphamide improves transplantation outcomes in patients with AML/MDS who are treated with checkpoint inhibitors. Cancer. 2020;126(10):2193–205. [DOI] [PubMed] [Google Scholar]
- 243.Nieto JC, Roldán E, Jiménez I, Fox L, Carabia J, Ortí G, et al. Posttransplant cyclophosphamide after allogeneic hematopoietic cell transplantation mitigates the immune activation induced by previous nivolumab therapy. Leukemia. 2020;34(12):3420–5. [DOI] [PubMed] [Google Scholar]
- 244.Ikegawa S, Meguri Y, Kondo T, Sugiura H, Sando Y, Nakamura M, et al. PTCy ameliorates GVHD by restoring regulatory and effector T-cell homeostasis in recipients with PD-1 blockade. Blood Advances. 2019;3(23):4081–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Daver NG, Vyas P, Kambhampati S, Malki MMA, Larson RA, Asch AS, et al. Tolerability and efficacy of the first-in-class anti-CD47 antibody magrolimab combined with azacitidine in frontline TP53m AML patients: Phase 1b results. Journal of Clinical Oncology. 2022;40(16_suppl):7020-. [Google Scholar]
- 246.Brunner AM, Esteve J, Porkka K, Knapper S, Traer E, Scholl S, et al. Efficacy and Safety of Sabatolimab (MBG453) in Combination with Hypomethylating Agents (HMAs) in Patients (Pts) with Very High/High-Risk Myelodysplastic Syndrome (vHR/HR-MDS) and Acute Myeloid Leukemia (AML): Final Analysis from a Phase Ib Study. Blood. 2021;138:244. [Google Scholar]
- 247.van Balen P, Jedema I, van Loenen MM, de Boer R, van Egmond HM, Hagedoorn RS, et al. HA-1H T-Cell Receptor Gene Transfer to Redirect Virus-Specific T Cells for Treatment of Hematological Malignancies After Allogeneic Stem Cell Transplantation: A Phase 1 Clinical Study. Frontiers in Immunology. 2020;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Huang R, Wen Q, Wang X, Yan H, Ma Y, Mai-Hong W, et al. Off-the-Shelf CD33 CAR-NK Cell Therapy for Relapse/Refractory AML: First-in-Human, Phase I Trial. Blood. 2022;140(Supplement 1):7450–1. [Google Scholar]