T-cell immune reconstitution (IR) after allogeneic hematopoietic cell transplantation (allo-HCT) is highly variable between patients and may take several months to even years. Patients with delayed or unbalanced T-cell IR have a higher probability of developing transplantation-related morbidity, mortality, and relapse of disease. Hence, there is a need for strategies to better predict and improve IR to reduce these limitations of allo-HCT. In this review, we provide an update of current and in-near-future clinically relevant strategies before, during, and after transplantation to achieve successful T-cell IR. Potent strategies are choosing the right HCT source (eg, donor-recipient matching, cell dose, graft manipulation), individualized conditioning and serotherapy (eg, antithymocyte globulin), nutritional status, exercise, home care, modulation of microbiota, enhancing homeostatic peripheral expansion, promoting thymopoiesis, and the use of adjuvant-targeted cellular immunotherapies. Strategies to prevent graft-versus-host disease are important as well because this complication and the subsequent need for immunosuppression affects T-cell IR and function. These options aim for personalized precision transplantation, where allo-HCT therapy is designed to boost a well-balanced T-cell IR and limit complications in individual patients, resulting in overall lower morbidity and higher survival chances.

Allogeneic hematopoietic cell transplantation (allo-HCT) nowadays is a widely accepted potentially curative treatment strategy for patients with refractory hematological malignancies and a variety of benign disorders. Immune reconstitution (IR) after allo-HCT, especially of the adaptive immune system, is highly variable between patients and can take several months to even years. A timely and balanced T-cell IR is especially important for sustained graft-versus-leukemia (GVL) effects, protection against opportunistic infections, and survival chances after allo-HCT.1-3 

Hence, there is a clear need to develop strategies to better predict and enhance T-cell recovery and function. The choice of the transplantation unit (eg, cell source, cell dose, donor-recipient matching, graft manipulation) and conditioning regimens are important variables pre-HCT that influence T-cell IR and subsequent complications (Figure 1). Also, peritransplantation parameters, such as the nutritional status of the patient and the composition of the gut microbiome, may influence outcome. Posttransplantation, T-cell reconstitution is dependent on 2 mechanisms: homeostatic peripheral expansion (HPE) and thymopoiesis. Therefore, T-cell IR would improve with strategies that stimulate HPE and thymic function. Graft-versus-host disease (GVHD) also is a major factor affecting T-cell IR,4  in part due to the immunosuppression needed to treat this complication after allo-HCT. As the thymus is especially sensitive to damage, such as during GVHD, even minor-grade GVHD might affect thymopoiesis.4-6  Therefore, strategies to prevent GVHD might improve T-cell IR as well.

Figure 1.

Pre-, peri-, and post-HCT factors affecting T-cell IR. Depicted in the bars are relative immune cell levels. Red, <reference values; risk for complications. Green, ≥reference values; adequate immunity.

Figure 1.

Pre-, peri-, and post-HCT factors affecting T-cell IR. Depicted in the bars are relative immune cell levels. Red, <reference values; risk for complications. Green, ≥reference values; adequate immunity.

Close modal

In this review, we comprehensively discuss current and in near-future clinically relevant strategies to better predict and improve T-cell IR and function after allo-HCT.

Choice of HCT source

Transplantation with bone marrow (BM) or peripheral blood (PB) (stem) cells from a matched family donor is considered the standard for allo-HCT. Alternative choices are BM/PB from a volunteer unrelated matched or mismatched donor, a haploidentical or mismatched family donor, and unrelated cord blood (CB). In addition, in an attempt to lower the risk of GVHD, the physician may choose for different strategies: CD34+ selection, ex vivo T-cell depletion (TCD) using negative selection with CD3+ or CD3αβ+ beads, in vivo TCD using serotherapy (T-cell–binding antibodies) such as antithymocyte globulin (ATG) or alemtuzumab, or for posttransplant cyclophosphamide (in haploidentical setting). The choice of the HCT source and its manipulation are important factors in the probability of T-cell IR, and therefore different HCT sources are associated with different probabilities for various outcomes; for example, probability on GVHD (acute and chronic), relapse,7  and immune recovery. Use of BM/PB grafts from younger donors is shown to improve survival chances,8,9  which may be due to enhanced immune-recovery probability. Overall, all HCT sources have advantages and disadvantages, and no clear superior cell source exists. The donor choice highly depends on institutional practices: some centers are more confident with CB, whereas others prefer BM/PB from matched unrelated donors, or haploidentical transplants.

HLA matching.

The availability of a donor is largely based on matching HLAs between patient and donor. For BM/PB, donor and recipient are typically matched on high resolution for HLA-A, HLA-B, HLA-C, HLA-DRB1, and some centers also match for HLA-DQB1 and HLA-DP. For CB transplantation (CBT), most centers use less stringent HLA-matching criteria: HLA-A, HLA-B (on low resolution), and HLA-DRB1 (on high resolution). This mismatching in CBT is permissive without increasing the probability on GVHD (in particular chronic).10  For BM/PB, mismatching at 1 or more HLA loci (on high resolution) increases the incidence of transplant-related mortality, resulting in lower survival chances. A mismatch of donor-specific antibodies may influence engraftment as well.11  Interestingly, in CBT, a certain degree of mismatching is suggested to be associated with increased GVL whereas GVHD probability is not increased.12,13  Importantly, to influence the probability of finding a (better matched) donor, the following are needed: recruitment of minority BM/PB donors, donor retention, improved efficiency of searches, and, for CB donor selection, a larger global inventory of diverse units in public banks.

Cell dose.

To permit any chance of T-cell recovery, myeloid engraftment needs to occur first. For this, the graft needs to contain a sufficient amount of (nucleated/CD34+) cells as a low cell dose hampers engraftment.14,15  CB units generally contain lower cell numbers than do BM/PB, which may be a problem for transplanting adults or larger children. Therefore, CB units are selected on cell dose, which is disease (malignant/nonmalignant) and HLA-matching dependent. Furthermore, recently, strategies to identify the better cryopreserved CB units have been developed as well, such as selection based on the total colony-forming unit16  or the amount of Aldehyde-dehydrogenase-Bright cells.17  These quality checks suggest better prediction of neutrophil engraftment, and may subsequently enhance the T-cell IR potential after CBT. For BM/PB, cell dose from HCT in adults is usually sufficient (although a minimum number is still required15 ) because an adequate amount of cells can readily be harvested from the donor (BM/apheresis).

For CB grafts that do not contain enough cells for transplantation, ex vivo expansion strategies are available: Delta1 Notch ligand,18  coculture with mesenchymal cells (eg, Mesoblast),19  aryl hydrocarbon receptor antagonist StemRegenin 1 (SR1; eg, HSC835),20  nicotinamide (eg, NiCord),21  and the copper chelator tetraethylenepentamine (TEPA; eg, StemEx).22  These expansion methods were successfully used in various clinical phase 1/2 studies and shown to be safe and effective. Overall, over 95% neutrophil engraftment was noted after a median time of 10 to 15 days. Another approach to overcome low cell dose is the transplantation of 2 CBT units, double CBT. In this, both CB units contribute to engraftment and reconstitution, but only 1 provides durable neutrophil engraftment.23,24  Neutrophil engraftment after double CBT (of usually inadequately dosed units for single-unit CBT) is successful and shows 85% to 100% engraftment at a median time of 24 days, which is comparable to single CBT with sufficient cell dose. Some reports suggested that double CBT is associated with lower relapse probability25  compared with single CBT. However, randomized controlled trials failed to prove this effect on relapse,26,27  and higher GVHD rates are shown after double CBT.28  Coinfusion with CD34+ cells from a haploidentical donor (haplo-cord) can also overcome the lower cell dose for CBT. Compared with double CBT, haplo-cord showed faster neutrophil recovery, lower risk of GVHD, but slower T-cell IR, and is suggested to be associated with lower relapse risk.29  Randomized controlled studies are, however, lacking.

Immune reconstitution.

Differences in IR rates between cell sources and manipulations might also influence the choice of HCT source. In adults, T-cell IR generally is considered most rapid after HCT with PB compared with BM, without ex vivo TCD.30,31  Not surprisingly, T-cell IR is more rapid after PB/BM from a HLA-matched donor compared with haploidentical BM/PB conditioned with ATG.32  Nevertheless, the effect of the nowadays-popular option of post-haplo cyclophosphamide on T-cell IR remains to be investigated. After ex vivo TCD with serotherapy, T-cell IR is similar in recipients of PB or BM.33  Obviously, T-cell reconstitution is severely hampered after HCT with CD34-selected PB/BM.34,35  T-cell IR is prolonged in adult recipients of ex vivo TCD-unrelated BM, compared with pediatric recipients of unrelated BM transplantation (BMT) and adults receiving related BMT.36  In children, data on PB are scarce as BM and CB are most frequently used as graft sources. Without serotherapy, overall T-cell IR is comparable after BMT or CBT in children, although CD8+ T-cell reconstitution is faster after BMT, whereas regulatory T cells (Tregs) and CD4+ T-cell recovery are faster after CBT.37  The use of ATG (aiming for in vivo T-cell depletion) appears to be critical and cell source dependent; T-cell IR after CBT with ATG especially is severely hampered compared with BMT.38 

Improving donor-recipient matching

PIRCHE.

Recently, an additional donor-recipient matching strategy was presented based on the probability of indirect HLA-molecule recognition represented by the number of predicted indirectly recognizable HLA epitopes (PIRCHEs).39  The number of PIRCHEs presented on HLA class I and II (PIRCHE-I and -II, respectively) is highly correlated with alloreactivity. For BM/PB donors with low PIRCHE-I and -II, the outcomes were similar to 10 of 10 HLA matching, whereas in CB a high PIRCHE-I is related to higher GVL.40 

HLA-KIR matching.

Natural killer (NK) cells are one of the first cells that reconstitute after allo-HCT, making them important for immunity when T-cell counts are still low. NK-cell function is regulated by the balance of activating and inhibitory signals via killer immunoglobulin-like receptors (KIRs), which interact with specific HLA class I ligands. This may have implications for GVL; when inhibitory KIRs are mismatched for HLA type (mainly HLA-C), donor NK cells recognize and kill recipient leukemia cells.41-43  However, KIR mismatching can also lead to GVHD-like syndromes.44  Future prospective trials should assess the effect of HLA-KIR matching on the balance between unwanted effects from mismatching (eg, graft failure and GVHD) and its wanted effects (eg, GVL).

Conditioning prior to HCT

Chemotherapeutics and total body irradiation.

Conditioning prior to HCT includes chemotherapy, such as cyclophosphamide, fludarabine, and busulfan, and/or total body irradiation. In addition, serotherapy, such as ATG or alemtuzumab, is added to reduce the risk of graft rejection and GVHD. Both chemotherapeutics and irradiation may severely affect T-cell IR after HCT,45  mainly due to thymic damage.46  Notably, T-cell IR is delayed with myeloablative conditioning compared with nonmyeloablative conditioning without serotherapy.47  Therefore, T-cell IR could be enhanced by optimizing the dosage of these chemotherapeutics via pharmacokinetic and dynamic modeling, and thymic shielding during irradiation, aiming for minimal thymic damage. Furthermore, some chemotherapy with longer half-life (eg, fludarabine) may still be present during the infusion of the cells and can subsequently deplete T cells in vivo. Also, sex steroid hormone ablation may be applied to prevent thymic damage by chemotherapy/irradiation (described in the paragraph on posttransplantation interventions).

Individualized serotherapy to improve T-cell IR.

ATG was introduced to the conditioning regimen for in vivo TCD to reduce GVHD risk which can reach a cumulative incidence of over 40% to 70% even with a well-matched unrelated donor. A major drawback of conditioning with serotherapy is hampered early T- and B-cell recovery.48,49  This effect is partly through direct binding and killing of T and B cells, and also through “off-target” cytotoxic effects on thymus cells, affecting thymopoiesis.50  However, abandoning ATG from the conditioning is associated with significantly higher incidences of graft rejection and GVHD.51 

Recently, high ATG exposure after allo-HCT was associated with detrimental effects on IR.38  The highly variable pharmacokinetics of ATG between patients causes underexposure or overexposure in a significant number of patients.52  CB cells were affected more by ATG compared with BM/PB; the tolerated post-HCT ATG exposure, which does not affect IR, is lower for CBT (<20 active units [AU] × day/mL) compared with BMT/peripheral blood cell transplantation (<50 AU × day/mL).38  CBT without ATG and CBT with very low ATG exposure were associated with excellent IR potential,49,51  stressing the importance for individualization. Interestingly, these studies also suggest that ATG exposure before, but not after, transplantation prevented GVHD (acute and chronic) not only in children, but also in adults (Rick Admiraal, Jurgen Kuball, and J.J.B., unpublished data). Sufficiently high exposure before CBT also prevented rejection. This indicates that there still is an important role for ATG, at least for HCT in nonmalignant diseases for graft rejection prevention, but earlier in the conditioning. In adult BM/PB recipients, this positive effect from conditioning with ATG on lower GVHD risk, but negative effects on T-cell IR, was also found.48,53  In HCT for hematopoietic malignancies, the decreased T-cell IR from posttransplantation ATG should especially be avoided, as this increases relapse risk and decreases survival.54,55  Taken together, these data suggest that personalized ATG dosing impacts T-cell IR potential after HCT, resulting in better survival chances.

Nutrition and microbiotics

Despite careful assessment and food supplementation, nutritional deficiency is common in allo-HCT patients. Nutritional requirements increase (up to 130%-150% of normal) due to catabolic stress, such as during IR and complications.56-58  Post-HCT calorie intake is directly related to time of neutrophil engraftment,56  and calorie requirements increase to ≥35 kcal/kg per day in addition to 1.5 to 2 g/kg per day of protein in the setting of GVHD.57 

To maintain nutritional status during and after allo-HCT treatment, patients often receive enteral nutrition or total parenteral nutrition (TPN).59  Recently, enteral nutrition in the form of an oral elemental diet was shown to be superior to TPN in decreasing the duration and severity of mucositis and duration of hospitalization.60  Even during intestinal GVHD, outcome (eg, diarrhea and time to complete dietary recovery) after enteral nutrition was comparable to TPN.61  TPN, compared with enteral nutrition, resulted in significantly higher morbidity and mortality.58  Therefore, recent studies suggest that enteral nutrition, seems to be the best approach to maintain nutritional status after allo-HCT, even in patients with mucositis or intestinal GVHD.

The composition of nutrients is important for IR after allo-HCT; a lack of zinc, selenium, iron, and antioxidant vitamins can lead to clinically significant immune deficiency and infections.62,63  On the other hand, immune-modifying nutrients like vitamin D could be used to dampen GVHD,64  as it increases Treg numbers.65  Notably, vitamin D supplementation is only beneficial in decreasing GVHD risk for patients with the low active AA or Aa vitamin D receptor (VDR) phenotype.66  Vitamin D increases the risk for GVHD in patients with the active aa VDR phenotype. This poses opportunities to improve allo-HCT outcome using vitamin D supplementation, at least in patients with low VDR activity, which is currently evaluated in prospective trials.

Recent studies investigating the relation between intestinal microbiotics and GVHD in the human HCT setting found that a lower diversity of microbiota relates to a higher risk of GVHD and mortality.67,68  Specifically, a shift toward higher presence of Enterococcus faecium and Enterococcus faecalis was found in GVHD patients,69  whereas genus Blautia bacteria was associated with a lower probability of GVHD.70  Therefore, choosing the right antibiotic regime may be of importance for its effects on microbiome diversity and GVHD probability.71  As prebiotics and probiotics maintain and restore microbiotic diversity, the use of probiotics to reduce GVHD risk is currently under investigation. The first results indicate that probiotic treatment prior to and after allo-HCT is safe,72  and might even reduce GVHD severity associated with better T-cell IR.67 

Exercise and home care

Daily exercise before and after HCT may fasten T-cell IR,73  although not all studies found a significant effect.74  Nevertheless, there is evidence indicating beneficial effects of physical exercise in the distribution and function of immune cells in healthy subjects.75  Prospective trials are warranted to prove this.

Some advantageous effects of home care in the neutropenic phase of the treatment are reported. Home care correlated with lower acute GVHD (aGVHD) incidence, and survival was at least as high as historic controls.76,77  Because home care would improve quality of life, reduce costs, and has been shown to be safe, further research on outcome and the effect on IR would be of interest.

For T-cell recovery, homeostatic peripheral expansion is the most important mechanism in absence of thymopoiesis, which takes at least 6 to 12 months. Improving HPE will, therefore, ultimately improve early T-cell IR. Some cytokine-based therapies for improving IR through HPE are currently under investigation (Figure 2). Nevertheless, early T-cell immunity through HPE is limited due to a limited T-cell receptor (TCR) diversity, as thymopoiesis is needed to provide new TCR clones in naive T cells. Treatment of patients before receiving allo-HCT (eg, conditioning, steroids, chemotherapy) causes damage to the thymus. GVHD after HCT can also significantly damage the thymus,4  whereas, on the other hand, immunosuppressive treatment of GVHD (eg, steroids, cyclosporine, tacrolimus) can hamper thymopoiesis as well. Therefore, thymic damage from GVHD needs to be balanced from that of immunosuppression. Therapeutic drug monitoring, such as for levels of tacrolimus,78  cyclosporine,79  and mycophenolate,80  can reduce the probability of thymic damage by GVHD, while limiting their effects on thymopoiesis by targeting to the appropriate exposure. Furthermore, of interest are several therapies linked to normal thymic function that are currently being investigated to directly promote thymopoiesis after allo-HCT (Figure 2).

Figure 2.

Overview on timing of noncellular posttransplantation interventions to improve T-cell IR via HPE and thymopoiesis.

Figure 2.

Overview on timing of noncellular posttransplantation interventions to improve T-cell IR via HPE and thymopoiesis.

Close modal

Improving T-cell IR through HPE

IL-2.

Interleukin-2 (IL-2) was introduced in the HCT setting for its role in the differentiation of T cells into effector cells and promotion of T-, B-, and NK-cell proliferation. Although IL-2 can have severe side effects at higher dosage (eg, capillary leak syndrome), low-dose IL-2 administration after HCT reduced relapse probability, probably due to an increased GVL response.81,82  It also restored homeostasis of CD4+ T cells, and increased Treg counts, associated with clinical improvement of chronic GVHD (cGVHD), whereas no adverse events were noted.83,84  Therefore, low-dose IL-2 therapy may improve T-cell IR, contributing to GVL, without increasing GVHD probability.

IL-15.

IL-15 is another interesting cytokine for improving HPE, as it is important for the expansion of B and T cells and for the survival of NK cells. In a murine HCT model, IL-15, during the first month after HCT, was associated with early T-cell and NK-cell IR.85  In humans, no systemic IL-15 therapy after allo-HCT has been evaluated so far. Only IL-15–stimulated CD3/CD19-depleted graft cells, from part of the HCT graft, or IL-15–activated cytokine-induced killer cells have been studied, albeit in few patients. Interestingly, this therapy increased NK, NKT, and T-cell numbers at 15 to 30 days after receipt, without exacerbating GVHD,86  and showed GVL potential when given at the initiation of relapse.87  IL-15 stimulation of stem cells would, therefore, be interesting to evaluate in future studies.

Improving T-cell IR through thymopoiesis

IL-7.

The administration of IL-7 was investigated in the HCT setting, as it naturally promotes the differentiation and proliferation of naive T cells. Treatment with recombinant IL-7 after HCT enhanced TCR diversity, induced a doubling of (mainly memory) CD4 and CD8 T cells, with no effects on Tregs, NK, or B cells.88  Interestingly, IL-7 also increased the amount of functional T cells, including virus-specific T cells, whereas no significant increase in GVHD or other serious toxicities were seen.88  These first results show promise for IL-7 in enhancing T-cell IR after HCT.

KGF.

Keratinocyte growth factor (KGF) promotes proliferation and maturation of immature T cells and plays a role in postnatal thymic regeneration. KGF, or palifermin, is used as standard-of-care peri-HCT therapy in some centers, and is US Food and Drug Administration (FDA) approved to prevent mucositis. Treatment of HCT patients with KGF, during 3 days before and 3 days after conditioning therapy, indeed not only decreased mucositis, but also prevented damage to the thymus.89,90  Although not all studies found any noteworthy impact of KGF on T-cell IR directly,89  this protection from thymic damage showed enhanced thymopoiesis, with early T-cell recovery probably caused by induction of IL-7, and lower GVHD incidence.90 

Sex steroid ablation.

Sex steroids are known to inhibit thymic function. Thus, sex steroid ablation may be applied to improve thymopoiesis. Administration of luteinizing hormone-releasing hormone (LHRH) agonists after HCT showed enhanced TCR excision circle production and T-cell repertoire regeneration, with enhanced total and naive CD4+ T-cell regeneration,91  indicating thymopoiesis. However, a major limitation of this approach is the surge in sex steroid after LHRH-agonist treatment, so a more rational approach could be to use LHRH antagonists.92  Another strategy for sex steroid ablation, leuprolide acetate (Lupron), in combination with KGF, showed increased reconstitution of naive CD4+ and CD8+ T cells, with a more diverse T-cell repertoire, in mice.93  Currently, phase 2 clinical studies are under way evaluating the effect of Lupron in the human allo-HCT setting (clinicaltrials.gov identifier: fNCT01338987).

Thymosin α 1.

Thymosin α 1 (Tα1) has recently been investigated in the HCT setting, as the thymus naturally promotes T-cell development by secreting this hormone. Tα1 (Thymalfasin/Zadaxin) is a FDA approved orphan drug for treatment of chronic hepatitis B, with no adverse effects found in recipients from 13 months to 99 years old.94  In the human HCT setting, Tα1 administration showed encouraging first results on T-cell IR. Subcutaneous administrations of Tα1, twice weekly for 4 weeks after allo-HCT, caused increased CD4+ T-cell recovery and pathogen-specific T cells to appear already at 1 month after HCT, which was earlier and in higher levels compared with controls.95  Additionally, Tα1 lowered the cumulative incidence of nonrelapse mortality (mainly infection-related) and increased event-free survival.94,95  Therefore, Tα1 therapy might be a promising novel therapy to enhance T-cell IR after allo-HCT.

Cellular immunotherapies to improve immunity

The lack of effector immune cells after allo-HCT may be compensated for with adjuvant cell-based immunotherapies. Due to high costs, regulatory burden, and complicated production, cellular immunotherapies are currently not widely being used in the HCT setting. They mainly have potency to be used to treat life-threatening complications such as relapse, viral reactivations, and GVHD, but could also be used prophylactically. Here, we discuss the most potent cellular immunotherapies currently used to improve immunity after allo-HCT (Figure 3).

Figure 3.

Overview on timing of cellular immunotherapies to improve immunity after allo-HCT. iNKT, invariant NKT; TRM, transplantation-related mortality.

Figure 3.

Overview on timing of cellular immunotherapies to improve immunity after allo-HCT. iNKT, invariant NKT; TRM, transplantation-related mortality.

Close modal

DC vaccination.

Dendritic cells (DCs) are highly specialized antigen-presenting cells that regulate the balance between action (relevant for GVL) and suppression of the immune system (relevant for GVHD).96  Therefore, DC vaccination is of major interest as a tool to modulate immune responses after allo-HCT. For BMT/peripheral blood cell transplantation, DCs can either be obtained by culturing monocytes into monocyte-derived DCs, or by directly isolating plasmacytoid DCs or conventional (myeloid) DCs from the peripheral blood of the donor. However, the low number of circulating DCs complicates their clinical application. In the case of CBT, CD34+ stem cells from part of the CB unit can easily be differentiated ex vivo into potent CB-DCs targeting viral and fungal infections after HCT, and even relapse.97,98  In addition, tolerogenic DCs might be a future therapy to suppress GVHD, as shown in a murine HCT model.99 

γδ T cells.

Increased numbers of donor γδ T cells early after allo-HCT correlate with better overall survival of leukemia patients, without increased risk of GVHD.100  Therefore, γδ T cells hold promise to improve outcome after allo-HCT. To date, trials using γδ T cells focus only on activated autologous Vγ9Vδ2 γδ T cells, combined with IL-2, or activated and amplified ex vivo.101  This stresses the need for future evaluation of the implementation of these potent cells in the allogeneic HCT setting, as is currently being investigated in a setting that combines γδ T cells and NK cells.102,103 

NK cells.

Adoptive NK-cell immunotherapy in the allo-HCT setting is under investigation as well. Results from one of the earliest studies suggest limited adverse events and no induction of GVHD.104  More recently, the combination of NK and γδ T cells was evaluated as a cell-based immunotherapy after allo-HCT. It is suggested that infusion of NK cells and γδ T lymphocytes around the time of HCT may protect the patient from early relapse, viruses (in particular, cytomegalovirus [CMV]), and graft rejection.105  Initial efforts are currently under way using TCR-αβ/CD19-depleted cell populations, which contain CD34+ progenitors, as well as mature NK cells and γδ T cells.102,103 

NKT cells.

For their immunomodulatory properties, immunotherapy using NKT cells could be useful in the allo-HCT setting as well. NKT cells play a role in both tumor surveillance and GVHD,106  with early reconstitution suggested to be predictive of lower relapse and GVHD risk and higher survival.107,108  Clinical trials implementing expanded NKT cells in the allo-HCT setting are currently under way.

Engineered T cells.

One method to improve T-cell recovery is donor lymphocyte infusion (DLI). This nonspecific T-cell therapy is, however, associated with increased risk on GVHD.109  Furthermore, because of the low frequency of T cells reactive against many common viruses within the circulation, higher DLI doses may be required to confer clinical benefit against viruses, consequently increasing the risk of GVHD.

A more targeted method to enhance T-cell immunity after allo-HCT is with adoptively transferred antigen-specific T cells. These T cells can be expanded ex vivo after stimulation with antigen-presenting cells presenting specific antigens, such as CMV, Epstein-Barr virus, and adenovirus,110  or against malignant cells to induce GVL.111  A drawback of this therapy is the long time needed for production of specific T cells. γ capture, based on interferon γ production, of functional antigen-specific T cells from a donor is a faster alternative. But for this method, the presence of specific T cells is essential and demands a seropositive donor. This is a challenge when the donor lacks viral immunity, or with CB which generally contains virus-naive immune cells. Nevertheless, virus-specific T cells can also be cultured from naive CB T cells.112 

Another way is T-cell gene therapy to produce chimeric antigen receptor (CAR) T cells or engineer T cells to express specific TCRs (TCR-T cells). Based on the chosen CAR or TCR binding domain, these modified T cells can be used to target viruses, and leukemic cells can be used to target relapse (eg, CART19, WT1-TCR-T cells).113-115  But they can potentially also be used as an alternative myeloablative therapy.116  Drawbacks of genetically modified T-cell therapy, however, are possible cytotoxic effects on normal cells that express target antigens.115  Nevertheless, the specificity and potency of this therapy is shown by the finding that donor-derived anti-CD19-CAR T cells induce complete remission in relapsed patients, without exacerbating GVHD.117  In addition, these cells have been shown to persist for over 3 years, providing long-term protection against their target.114 

Tregs and MSCs.

Tregs and mesenchymal stromal/stem cells (MSCs) can inhibit immune responses, and have been investigated as adjuvant cell therapies to treat or prevent GVHD. The nonspecific MSC therapy did not prevent GVHD when cotransplanted alongside the HCT graft,118  whereas even increased relapse rates and mortality were noted.119  However, reduced aGVHD risk was seen when given after HCT before GVHD onset.120  Coinfusion of Tregs alongside the HCT graft decreased the occurrence of aGVHD and cGVHD, while maintaining GVL.121,122  This also resulted in high frequencies of pathogen-specific CD4+ and CD8+ T cells already at 2 months after HCT and decreased incidence of CMV disease, showing strong antiviral protection after pandemic influenza vaccination.122  No negative effects due to the Treg infusion were reported. Furthermore, transplantation with CB-derived Tregs directly after CBT showed an increase in viral infections within 30 days of infusion, but lowered the occurrence of GVHD without increased risk of relapse.121,123  This may make Treg immunotherapy a promising strategy to decrease the risk for GVHD associated with better T-cell IR potential.

We have summarized a broad variety of novel strategies before, during, and after HCT to improve T-cell IR, aiming to decrease the risk of complications and increase survival chances. Selecting the appropriate HCT source and graft manipulations are the first critical factors affecting T-cell IR after transplantation. Individualizing the conditioning regimen is crucial as well, especially for compounds that dramatically influence T-cell IR, such as ATG. Furthermore, enhancing homeostatic peripheral expansion, preventing thymic damage, and promoting thymopoiesis are important in enhancing T-cell IR. Better disease control may be achieved with adjuvant (specific) cellular immunotherapies to prevent GVHD and promote immunity against pathogens and cancer cells. A combination of these pre-, peri-, and post-HCT strategies probably will most potently enable timely and balanced T-cell recovery.

Immunomonitoring will be of major importance to identify patients at risk, which would provide opportunities for early immune interventions. For instance, patients with low T-cell counts early after HCT, and/or low TCR diversity, are at risk for HCT-related complications and could benefit from adjuvant immunotherapies, such as low-dose IL-2, administration of Tregs, or leukemia/virus-specific DCs and/or specific T cells. New functional measures of immune recovery would, therefore, also guide clinicians in the assessment of IR beyond simple absolute lymphocyte counts. In addition, it is important to continue adequate supportive care with continuation of antimicrobial prophylaxis and polymerase chain reaction monitoring for viruses until adequate immune recovery occurs.

Standardization and harmonization of immunomonitoring would enable better comparison between multiple allo-HCT studies, in order to better understand the biology of IR, and find biomarkers.124  These biomarkers will help the physician guide the developing immune system, and anticipate complications in order to avert them. This will pave the path to precision transplantation in the future, in which allo-HCT therapy will be personalized, using a combination of strategies to boost T-cell IR, subsequently resulting in lower morbidity and higher survival after allo-HCT.

The authors thank Paul Besseling for his help with searching for recent, relevant literature on strategies to improve IR after HCT.

This work was supported by a Kinderen Kankervrij (KIKA) grant (project 142).

Contribution: C.d.K. wrote and edited the report; S.N. and J.J.B. reviewed the manuscript and provided critical comments; and all authors reviewed and approved the final report.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Jaap Jan Boelens, Blood and Marrow Transplantation Program and Laboratory of Translational Immunology, University Medical Center Utrecht, Lundlaan 6: KC 0.30.063.0, 3584 CX Utrecht, The Netherlands; e-mail: j.j.boelens@umcutrecht.nl.

1.
Kim
DH
,
Sohn
SK
,
Won
DI
,
Lee
NY
,
Suh
JS
,
Lee
KB
.
Rapid helper T-cell recovery above 200 x 10 6/l at 3 months correlates to successful transplant outcomes after allogeneic stem cell transplantation
.
Bone Marrow Transplant
.
2006
;
37
(
12
):
1119
-
1128
.
2.
Pourgheysari
B
,
Piper
KP
,
McLarnon
A
, et al
.
Early reconstitution of effector memory CD4+ CMV-specific T cells protects against CMV reactivation following allogeneic SCT
.
Bone Marrow Transplant
.
2009
;
43
(
11
):
853
-
861
.
3.
Edinger
M
,
Hoffmann
P
,
Ermann
J
, et al
.
CD4+CD25+ regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation
.
Nat Med
.
2003
;
9
(
9
):
1144
-
1150
.
4.
Clave
E
,
Busson
M
,
Douay
C
, et al
.
Acute graft-versus-host disease transiently impairs thymic output in young patients after allogeneic hematopoietic stem cell transplantation
.
Blood
.
2009
;
113
(
25
):
6477
-
6484
.
5.
Holländer
GA
,
Widmer
B
,
Burakoff
SJ
.
Loss of normal thymic repertoire selection and persistence of autoreactive T cells in graft vs host disease
.
J Immunol
.
1994
;
152
(
4
):
1609
-
1617
.
6.
Hauri-Hohl
MM
,
Keller
MP
,
Gill
J
, et al
.
Donor T-cell alloreactivity against host thymic epithelium limits T-cell development after bone marrow transplantation
.
Blood
.
2007
;
109
(
9
):
4080
-
4088
.
7.
Sirinoglu Demiriz
I
,
Tekgunduz
E
,
Altuntas
F
.
What is the most appropriate source for hematopoietic stem cell transplantation? Peripheral stem cell/bone marrow/cord blood
.
Bone Marrow Res
.
2012
;
2012
:
834040
.
8.
Kollman
C
,
Spellman
SR
,
Zhang
M-J
, et al
.
The effect of donor characteristics on survival after unrelated donor transplantation for hematologic malignancy
.
Blood
.
2016
;
127
(
2
):
260
-
267
.
9.
Kröger
N
,
Zabelina
T
,
de Wreede
L
, et al
;
MDS subcommittee of the Chronic Leukemia Working Party of the European Group for Blood and Marrow Transplantation (EBMT)
.
Allogeneic stem cell transplantation for older advanced MDS patients: improved survival with young unrelated donor in comparison with HLA-identical siblings
.
Leukemia
.
2013
;
27
(
3
):
604
-
609
.
10.
Horowitz
MM
.
High-resolution typing for unrelated donor transplantation: how far do we go?
Best Pract Res Clin Haematol
.
2009
;
22
(
4
):
537
-
541
.
11.
Takanashi
M
,
Atsuta
Y
,
Fujiwara
K
, et al
.
The impact of anti-HLA antibodies on unrelated cord blood transplantations
.
Blood
.
2010
;
116
(
15
):
2839
-
2846
.
12.
Brunstein
CG
,
Gutman
JA
,
Weisdorf
DJ
, et al
.
Allogeneic hematopoietic cell transplantation for hematologic malignancy: relative risks and benefits of double umbilical cord blood
.
Blood
.
2010
;
116
(
22
):
4693
-
4699
.
13.
Eapen
M
,
Rubinstein
P
,
Zhang
M-J
, et al
.
Outcomes of transplantation of unrelated donor umbilical cord blood and bone marrow in children with acute leukaemia: a comparison study
.
Lancet
.
2007
;
369
(
9577
):
1947
-
1954
.
14.
Wagner
JE
,
Barker
JN
,
DeFor
TE
, et al
.
Transplantation of unrelated donor umbilical cord blood in 102 patients with malignant and nonmalignant diseases: influence of CD34 cell dose and HLA disparity on treatment-related mortality and survival
.
Blood
.
2002
;
100
(
5
):
1611
-
1618
.
15.
Heimfeld
S
.
Bone marrow transplantation: how important is CD34 cell dose in HLA-identical stem cell transplantation?
Leukemia
.
2003
;
17
(
5
):
856
-
858
.
16.
Page
KM
,
Zhang
L
,
Mendizabal
A
, et al
.
Total colony-forming units are a strong, independent predictor of neutrophil and platelet engraftment after unrelated umbilical cord blood transplantation: a single-center analysis of 435 cord blood transplants
.
Biol Blood Marrow Transplant
.
2011
;
17
(
9
):
1362
-
1374
.
17.
Lee
HR
,
Shin
S
,
Yoon
JH
,
Roh
EY
,
Kim
BJ
,
Song
EY
.
Aldehyde dehydrogenase-bright cells correlated with the colony-forming unit-granulocyte-macrophage assay of thawed cord blood units
.
Transfusion
.
2014
;
54
(
7
):
1871
-
1875
.
18.
Delaney
C
,
Heimfeld
S
,
Brashem-Stein
C
,
Voorhies
H
,
Manger
RL
,
Bernstein
ID
.
Notch-mediated expansion of human cord blood progenitor cells capable of rapid myeloid reconstitution
.
Nat Med
.
2010
;
16
(
2
):
232
-
236
.
19.
de Lima
M
,
McNiece
I
,
Robinson
SN
, et al
.
Cord-blood engraftment with ex vivo mesenchymal-cell coculture
.
N Engl J Med
.
2012
;
367
(
24
):
2305
-
2315
.
20.
Boitano
AE
,
Wang
J
,
Romeo
R
, et al
.
Aryl hydrocarbon receptor antagonists promote the expansion of human hematopoietic stem cells
.
Science
.
2010
;
329
(
5997
):
1345
-
1348
.
21.
Horwitz
ME
,
Chao
NJ
,
Rizzieri
DA
, et al
.
Umbilical cord blood expansion with nicotinamide provides long-term multilineage engraftment
.
J Clin Invest
.
2014
;
124
(
7
):
3121
-
3128
.
22.
Stiff
PJ
,
Montesinos
P
,
Peled
T
, et al
.
StemEx (copper chelation based) ex vivo expanded umbilical cord blood stem cell transplantation (UCBT) accelerates engraftment and improves 100 day survival in myeloablated patients compared to a registry cohort undergoing double unit UCBT: results of a multicenter study of 101 patients with hematologic malignancies [abstract]
.
Blood
.
2013
;
122
(
21
).
Abstract 295
.
23.
Barker
JN
,
Weisdorf
DJ
,
DeFor
TE
, et al
.
Transplantation of 2 partially HLA-matched umbilical cord blood units to enhance engraftment in adults with hematologic malignancy
.
Blood
.
2005
;
105
(
3
):
1343
-
1347
.
24.
Somers
JA
,
Brand
A
,
van Hensbergen
Y
, et al
.
Double umbilical cord blood transplantation: a study of early engraftment kinetics in leukocyte subsets using HLA-specific monoclonal antibodies
.
Biol Blood Marrow Transplant
.
2013
;
19
(
2
):
266
-
273
.
25.
Verneris
MR
,
Brunstein
CG
,
Barker
J
, et al
.
Relapse risk after umbilical cord blood transplantation: enhanced graft-versus-leukemia effect in recipients of 2 units
.
Blood
.
2009
;
114
(
19
):
4293
-
4299
.
26.
Wagner
JE
,
Eapen
M
,
Carter
SL
, et al
.
No survival advantage after double umbilical cord blood (UCB) compared to single UCB transplant in children with hematological malignancy: results of the Blood and Marrow Transplant Clinical Trials Network (BMT CTN 0501) randomized trial [abstract]
.
Blood
.
2012
;
120
(
21
).
Abstract 359
.
27.
Ruggeri
A
,
Sanz
G
,
Bittencourt
H
, et al
;
Eurocord and Acute Leukemia Working Party of European Blood and Marrow Transplant Group
.
Comparison of outcomes after single or double cord blood transplantation in adults with acute leukemia using different types of myeloablative conditioning regimen, a retrospective study on behalf of Eurocord and the Acute Leukemia Working Party of EBMT
.
Leukemia
.
2014
;
28
(
4
):
779
-
786
.
28.
Ponce
DM
,
Gonzales
A
,
Lubin
M
, et al
.
Graft-versus-host disease after double-unit cord blood transplantation has unique features and an association with engrafting unit-to-recipient HLA match
.
Biol Blood Marrow Transplant
.
2013
;
19
(
6
):
904
-
911
.
29.
van Besien
K
,
Hari
P
,
Zhang
M-J
, et al
.
Reduced intensity haplo plus single cord transplant compared to double cord transplant: improved engraftment and graft-versus-host disease-free, relapse-free survival
.
Haematologica
.
2016
;
101
(
5
):
634
-
643
.
30.
Ottinger
HD
,
Beelen
DW
,
Scheulen
B
,
Schaefer
UW
,
Grosse-Wilde
H
.
Improved immune reconstitution after allotransplantation of peripheral blood stem cells instead of bone marrow
.
Blood
.
1996
;
88
(
7
):
2775
-
2779
.
31.
Storek
J
,
Dawson
MA
,
Storer
B
, et al
.
Immune reconstitution after allogeneic marrow transplantation compared with blood stem cell transplantation
.
Blood
.
2001
;
97
(
11
):
3380
-
3389
.
32.
Chang
Y-J
,
Zhao
X-Y
,
Huo
M-R
, et al
.
Immune reconstitution following unmanipulated HLA-mismatched/haploidentical transplantation compared with HLA-identical sibling transplantation
.
J Clin Immunol
.
2012
;
32
(
2
):
268
-
280
.
33.
Novitzky
N
,
Davison
GM
,
Hale
G
,
Waldmann
H
.
Immune reconstitution at 6 months following T-cell depleted hematopoietic stem cell transplantation is predictive for treatment outcome
.
Transplantation
.
2002
;
74
(
11
):
1551
-
1559
.
34.
Martínez
C
,
Urbano-Ispizua
A
,
Rozman
C
, et al
.
Immune reconstitution following allogeneic peripheral blood progenitor cell transplantation: comparison of recipients of positive CD34+ selected grafts with recipients of unmanipulated grafts
.
Exp Hematol
.
1999
;
27
(
3
):
561
-
568
.
35.
Damiani
D
,
Stocchi
R
,
Masolini
P
, et al
.
CD34+-selected versus unmanipulated autologous stem cell transplantation in multiple myeloma: impact on dendritic and immune recovery and on complications due to infection
.
Ann Oncol
.
2003
;
14
(
3
):
475
-
480
.
36.
Small
TN
,
Papadopoulos
EB
,
Boulad
F
, et al
.
Comparison of immune reconstitution after unrelated and related T-cell-depleted bone marrow transplantation: effect of patient age and donor leukocyte infusions
.
Blood
.
1999
;
93
(
2
):
467
-
480
.
37.
de Koning
C
,
Plantinga
M
,
Besseling
P
,
Boelens
JJ
,
Nierkens
S
.
Immune reconstitution after allogeneic hematopoietic cell transplantation in children
.
Biol Blood Marrow Transplant
.
2016
;
22
(
2
):
195
-
206
.
38.
Admiraal
R
,
van Kesteren
C
,
Jol-van der Zijde
CM
, et al
.
Association between anti-thymocyte globulin exposure and CD4+ immune reconstitution in paediatric haematopoietic cell transplantation: a multicentre retrospective pharmacodynamic cohort analysis
.
Lancet Haematol
.
2015
;
2
(
5
):
e194
-
e203
.
39.
Thus
KA
,
Ruizendaal
MT
,
de Hoop
TA
, et al
.
Refinement of the definition of permissible HLA-DPB1 mismatches with predicted indirectly recognizable HLA-DPB1 epitopes
.
Biol Blood Marrow Transplant
.
2014
;
20
(
11
):
1705
-
1710
.
40.
Thus
KA
,
de Hoop
TA
,
de Weger
RA
,
Bierings
MB
,
Boelens
JJ
,
Spierings
E
.
Predicted indirectly recognizable HLA epitopes class I promote antileukemia responses after cord blood transplantation: indications for a potential novel donor selection tool
.
Biol Blood Marrow Transplant
.
2016
;
22
(
1
):
170
-
173
.
41.
Venstrom
JM
,
Pittari
G
,
Gooley
TA
, et al
.
HLA-C-dependent prevention of leukemia relapse by donor activating KIR2DS1
.
N Engl J Med
.
2012
;
367
(
9
):
805
-
816
.
42.
Ruggeri
L
,
Capanni
M
,
Urbani
E
, et al
.
Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants
.
Science
.
2002
;
295
(
5562
):
2097
-
2100
.
43.
Rubnitz
JE
,
Inaba
H
,
Ribeiro
RC
, et al
.
NKAML: a pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia
.
J Clin Oncol
.
2010
;
28
(
6
):
955
-
959
.
44.
Miller
JS
,
Cooley
S
,
Parham
P
, et al
.
Missing KIR ligands are associated with less relapse and increased graft-versus-host disease (GVHD) following unrelated donor allogeneic HCT
.
Blood
.
2007
;
109
(
11
):
5058
-
5061
.
45.
Mackall
CL
,
Fleisher
TA
,
Brown
MR
, et al
.
Lymphocyte depletion during treatment with intensive chemotherapy for cancer
.
Blood
.
1994
;
84
(
7
):
2221
-
2228
.
46.
Mackall
CL
,
Fleisher
TA
,
Brown
MR
, et al
.
Age, thymopoiesis, and CD4+ T-lymphocyte regeneration after intensive chemotherapy
.
N Engl J Med
.
1995
;
332
(
3
):
143
-
149
.
47.
Petersen
SL
,
Ryder
LP
,
Björk
P
, et al
.
A comparison of T-, B- and NK-cell reconstitution following conventional or nonmyeloablative conditioning and transplantation with bone marrow or peripheral blood stem cells from human leucocyte antigen identical sibling donors
.
Bone Marrow Transplant
.
2003
;
32
(
1
):
65
-
72
.
48.
Bosch
M
,
Dhadda
M
,
Hoegh-Petersen
M
, et al
.
Immune reconstitution after anti-thymocyte globulin-conditioned hematopoietic cell transplantation
.
Cytotherapy
.
2012
;
14
(
10
):
1258
-
1275
.
49.
Lindemans
CA
,
Chiesa
R
,
Amrolia
PJ
, et al
.
Impact of thymoglobulin prior to pediatric unrelated umbilical cord blood transplantation on immune reconstitution and clinical outcome
.
Blood
.
2014
;
123
(
1
):
126
-
132
.
50.
Na
IK
,
Wittenbecher
F
,
Dziubianau
M
, et al
.
Rabbit antithymocyte globulin (thymoglobulin) impairs the thymic output of both conventional and regulatory CD4+ T cells after allogeneic hematopoietic stem cell transplantation in adult patients
.
Haematologica
.
2013
;
98
(
1
):
23
-
30
.
51.
Chiesa
R
,
Gilmour
K
,
Qasim
W
, et al
.
Omission of in vivo T-cell depletion promotes rapid expansion of naïve CD4+ cord blood lymphocytes and restores adaptive immunity within 2 months after unrelated cord blood transplant
.
Br J Haematol
.
2012
;
156
(
5
):
656
-
666
.
52.
Admiraal
R
,
van Kesteren
C
,
Jol-van der Zijde
CM
, et al
.
Population pharmacokinetic modeling of Thymoglobulin(®) in children receiving allogeneic-hematopoietic cell transplantation: towards improved survival through individualized dosing
.
Clin Pharmacokinet
.
2015
;
54
(
4
):
435
-
446
.
53.
Podgorny
PJ
,
Ugarte-Torres
A
,
Liu
Y
,
Williamson
TS
,
Russell
JA
,
Storek
J
.
High rabbit-antihuman thymocyte globulin levels are associated with low likelihood of graft-vs-host disease and high likelihood of posttransplant lymphoproliferative disorder
.
Biol Blood Marrow Transplant
.
2010
;
16
(
7
):
915
-
926
.
54.
Soiffer
RJ
,
LeRademacher
J
,
Ho
V
, et al
.
Impact of immune modulation with anti–T-cell antibodies on the outcome of reduced-intensity allogeneic hematopoietic stem cell transplantation for hematologic malignancies
.
Blood
.
2011
;
117
(
25
):
6963
-
6970
.
55.
Admiraal
R
,
Lindemans
CA
,
van Kesteren
C
,
Bierings
MB
,
Versluijs
B
,
Nierkens
SBJ
.
Excellent T-cell reconstitution and survival provided ATG exposure is low or absent after pediatric cord blood transplantation [published online ahead of print October 4, 2016]
.
Blood
.
56.
Forchielli
ML
,
Azzi
N
,
Cadranel
S
,
Paolucci
G
.
Total parenteral nutrition in bone marrow transplant: what is the appropriate energy level?
Oncology
.
2003
;
64
(
1
):
7
-
13
.
57.
Roberts
S
,
Thompson
J
.
Graft-vs-host disease: nutrition therapy in a challenging condition
.
Nutr Clin Pract
.
2005
;
20
(
4
):
440
-
450
.
58.
Sheean
PM
.
Nutrition support of blood or marrow transplant recipients: how much do we really know?
Pract Gastroenterol
.
2005
;
26
:
84
-
97
.
59.
Weisdorf
SA
,
Lysne
J
,
Wind
D
, et al
.
Positive effect of prophylactic total parenteral nutrition on long-term outcome of bone marrow transplantation
.
Transplantation
.
1987
;
43
(
6
):
833
-
838
.
60.
Morishita
T
,
Tsushita
N
,
Imai
K
, et al
.
The efficacy of oral elemental diet in patients with hematopoietic stem cell transplantation [abstract]
.
Blood
.
2014
;
124
(
21
).
Abstract 1136
.
61.
Imataki
O
,
Nakatani
S
,
Hasegawa
T
, et al
.
Nutritional support for patients suffering from intestinal graft-versus-host disease after allogeneic hematopoietic stem cell transplantation
.
Am J Hematol
.
2006
;
81
(
10
):
747
-
752
.
62.
Cunningham-Rundles
S
,
McNeeley
DF
,
Moon
A
.
Mechanisms of nutrient modulation of the immune response
.
J Allergy Clin Immunol
.
2005
;
115
(
6
):
1119-1128, quiz 1129
.
63.
Worthington
ML
,
Cresci
G
.
Immune-modulating formulas: who wins the meta-analysis race?
Nutr Clin Pract
.
2011
;
26
(
6
):
650
-
655
.
64.
Benrashid
M
,
Moyers
K
,
Mohty
M
,
Savani
BN
.
Vitamin D deficiency, autoimmunity, and graft-versus-host-disease risk: Implication for preventive therapy
.
Exp Hematol
.
2012
;
40
(
4
):
263
-
267
.
65.
Prietl
B
,
Pilz
S
,
Wolf
M
, et al
.
Vitamin D supplementation and regulatory T cells in apparently healthy subjects: vitamin D treatment for autoimmune diseases?
Isr Med Assoc J
.
2010
;
12
(
3
):
136
-
139
.
66.
Cho
HJ
,
Shin
DY
,
Kim
JH
, et al
.
Impact of vitamin D receptor gene polymorphisms on clinical outcomes of HLA-matched sibling hematopoietic stem cell transplantation
.
Clin Transplant
.
2012
;
26
(
3
):
476
-
483
.
67.
Jenq
RR
,
Ubeda
C
,
Taur
Y
, et al
.
Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation
.
J Exp Med
.
2012
;
209
(
5
):
903
-
911
.
68.
Taur
Y
,
Jenq
RR
,
Perales
MA
, et al
.
The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation
.
Blood
.
2014
;
124
(
7
):
1174
-
1182
.
69.
Holler
E
,
Butzhammer
P
,
Schmid
K
, et al
.
Metagenomic analysis of the stool microbiome in patients receiving allogeneic stem cell transplantation: loss of diversity is associated with use of systemic antibiotics and more pronounced in gastrointestinal graft-versus-host disease
.
Biol Blood Marrow Transplant
.
2014
;
20
(
5
):
640
-
645
.
70.
Jenq
RR
,
Taur
Y
,
Devlin
SM
, et al
.
Intestinal Blautia is associated with reduced death from graft-versus-host disease
.
Biol Blood Marrow Transplant
.
2015
;
21
(
8
):
1373
-
1383
.
71.
Shono
Y
,
Docampo
MD
,
Peled
JU
, et al
.
Increased GVHD-related mortality with broad-spectrum antibiotic use after allogeneic hematopoietic stem cell transplantation in human patients and mice
.
Sci Transl Med
.
2016
;
8
(
339
):
339ra71
.
72.
Ladas
EJ
,
Bhatia
M
,
Chen
L
, et al
.
The safety and feasibility of probiotics in children and adolescents undergoing hematopoietic cell transplantation
.
Bone Marrow Transplant
.
2016
;
51
(
2
):
262
-
266
.
73.
Kim
SD
,
Kim
HS
.
A series of bed exercises to improve lymphocyte count in allogeneic bone marrow transplantation patients
.
Eur J Cancer Care (Engl)
.
2006
;
15
(
5
):
453
-
457
.
74.
Hayes
SC
,
Rowbottom
D
,
Davies
PSW
,
Parker
TW
,
Bashford
J
.
Immunological changes after cancer treatment and participation in an exercise program
.
Med Sci Sports Exerc
.
2003
;
35
(
1
):
2
-
9
.
75.
Timmons
BW
.
Paediatric exercise immunology: health and clinical applications
.
Exerc Immunol Rev
.
2005
;
11
:
108
-
144
.
76.
Ringdén
O
,
Remberger
M
,
Törlén
J
, et al
.
Home care during neutropenia after allogeneic hematopoietic stem cell transplantation in children and adolescents is safe and may be more advantageous than isolation in hospital
.
Pediatr Transplant
.
2014
;
18
(
4
):
398
-
404
.
77.
Ringdén
O
,
Remberger
M
,
Holmberg
K
, et al
.
Many days at home during neutropenia after allogeneic hematopoietic stem cell transplantation correlates with low incidence of acute graft-versus-host disease
.
Biol Blood Marrow Transplant
.
2013
;
19
(
2
):
314
-
320
.
78.
Wallin
JE
,
Friberg
LE
,
Fasth
A
,
Staatz
CE
.
Population pharmacokinetics of tacrolimus in pediatric hematopoietic stem cell transplant recipients: new initial dosage suggestions and a model-based dosage adjustment tool
.
Ther Drug Monit
.
2009
;
31
(
4
):
457
-
466
.
79.
Dupuis
LL
,
Seto
W
,
Teuffel
O
, et al
.
Prediction of area under the cyclosporine concentration versus time curve in children undergoing hematopoietic stem cell transplantation
.
Biol Blood Marrow Transplant
.
2013
;
19
(
3
):
418
-
423
.
80.
Bhatia
M
,
Militano
O
,
Jin
Z
, et al
.
An age-dependent pharmacokinetic study of intravenous and oral mycophenolate mofetil in combination with tacrolimus for GVHD prophylaxis in pediatric allogeneic stem cell transplantation recipients
.
Biol Blood Marrow Transplant
.
2010
;
16
(
3
):
333
-
343
.
81.
Inamoto
Y
,
Fefer
A
,
Sandmaier
BM
, et al
.
A phase I/II study of chemotherapy followed by donor lymphocyte infusion plus interleukin-2 for relapsed acute leukemia after allogeneic hematopoietic cell transplantation
.
Biol Blood Marrow Transplant
.
2011
;
17
(
9
):
1308
-
1315
.
82.
Kennedy-Nasser
AA
,
Ku
S
,
Castillo-Caro
P
, 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
.
Clin Cancer Res
.
2014
;
20
(
8
):
2215
-
2225
.
83.
Matsuoka
K
,
Koreth
J
,
Kim
HT
, et al
.
Low-dose interleukin-2 therapy restores regulatory T cell homeostasis in patients with chronic graft-versus-host disease
.
Sci Transl Med
.
2013
;
5
(
179
):
179ra43
.
84.
Koreth
J
,
Matsuoka
K
,
Kim
HT
, et al
.
Interleukin-2 and regulatory T cells in graft-versus-host disease
.
N Engl J Med
.
2011
;
365
(
22
):
2055
-
2066
.
85.
Alpdogan
O
,
Eng
JM
,
Muriglan
SJ
, et al
.
Interleukin-15 enhances immune reconstitution after allogeneic bone marrow transplantation
.
Blood
.
2005
;
105
(
2
):
865
-
873
.
86.
Rettinger
E
,
Bonig
H
,
Wehner
S
, et al
.
Feasibility of IL-15-activated cytokine-induced killer cell infusions after haploidentical stem cell transplantation
.
Bone Marrow Transplant
.
2013
;
48
(
8
):
1141
-
1143
.
87.
Pfeiffer
MM
,
Schumm
M
,
Müller
I
,
Handgretinger
R
,
Lang
P
.
IL-15-stimulated CD3/CD19-depleted stem-cell boosts in relapsed pediatric patients after haploidentical SCT
.
Leukemia
.
2012
;
26
(
11
):
2435
-
2439
.
88.
Perales
M-A
,
Goldberg
JD
,
Yuan
J
, et al
.
Recombinant human interleukin-7 (CYT107) promotes T-cell recovery after allogeneic stem cell transplantation
.
Blood
.
2012
;
120
(
24
):
4882
-
4891
.
89.
Rizwan
R
,
Levine
JE
,
Defor
T
, et al
.
Peritransplant palifermin use and lymphocyte recovery after T-cell replete, matched related allogeneic hematopoietic cell transplantation
.
Am J Hematol
.
2011
;
86
(
10
):
879
-
882
.
90.
Min
D
,
Taylor
PA
,
Panoskaltsis-Mortari
A
, et al
.
Protection from thymic epithelial cell injury by keratinocyte growth factor: a new approach to improve thymic and peripheral T-cell reconstitution after bone marrow transplantation
.
Blood
.
2002
;
99
(
12
):
4592
-
4600
.
91.
Sutherland
JS
,
Spyroglou
L
,
Muirhead
JL
, et al
.
Enhanced immune system regeneration in humans following allogeneic or autologous hemopoietic stem cell transplantation by temporary sex steroid blockade
.
Clin Cancer Res
.
2008
;
14
(
4
):
1138
-
1149
.
92.
Velardi
E
,
Tsai
JJ
,
Holland
AM
, et al
.
Sex steroid blockade enhances thymopoiesis by modulating Notch signaling
.
J Exp Med
.
2014
;
211
(
12
):
2341
-
2349
.
93.
Kelly
RM
,
Highfill
SL
,
Panoskaltsis-Mortari
A
, et al
.
Keratinocyte growth factor and androgen blockade work in concert to protect against conditioning regimen-induced thymic epithelial damage and enhance T-cell reconstitution after murine bone marrow transplantation
.
Blood
.
2008
;
111
(
12
):
5734
-
5744
.
94.
Tuthill
C
,
King
RS
.
Thymosin apha 1-A peptide immune modulator with a broad range of clinical applications
.
Clin Exp Pharmacol
.
2013
;
3
(
4
):
133
.
95.
Perruccio
K
,
Bonifazi
P
,
Topini
F
, et al
.
Thymosin α1 to harness immunity to pathogens after haploidentical hematopoietic transplantation
.
Ann N Y Acad Sci
.
2010
;
1194
:
153
-
161
.
96.
Plantinga
M
,
de Haar
C
,
Nierkens
S
,
Boelens
JJ
.
Dendritic cell therapy in an allogeneic-hematopoietic cell transplantation setting: an effective strategy toward better disease control?
Front Immunol
.
2014
;
5
:
218
.
97.
de Haar
C
,
Plantinga
M
,
Blokland
NJ
, et al
.
Generation of a cord blood-derived Wilms tumor 1 dendritic cell vaccine for AML patients treated with allogeneic cord blood transplantation
.
OncoImmunology
.
2015
;
4
(
11
):
e1023973
.
98.
Chang
MC
,
Lee
CN
,
Chen
YL
, et al
.
Cord blood stem-cell-derived dendritic cells generate potent antigen-specific immune responses and anti-tumour effects
.
Clin Sci (Lond)
.
2012
;
123
(
6
):
347
-
360
.
99.
Hadeiba
H
,
Sato
T
,
Habtezion
A
,
Oderup
C
,
Pan
J
,
Butcher
EC
.
CCR9 expression defines tolerogenic plasmacytoid dendritic cells able to suppress acute graft-versus-host disease
.
Nat Immunol
.
2008
;
9
(
11
):
1253
-
1260
.
100.
Godder
KT
,
Henslee-Downey
PJ
,
Mehta
J
, et al
.
Long term disease-free survival in acute leukemia patients recovering with increased gammadelta T cells after partially mismatched related donor bone marrow transplantation
.
Bone Marrow Transplant
.
2007
;
39
(
12
):
751
-
757
.
101.
Scheper
W
,
Gründer
C
,
Straetemans
T
,
Sebestyen
Z
,
Kuball
J
.
Hunting for clinical translation with innate-like immune cells and their receptors
.
Leukemia
.
2014
;
28
(
6
):
1181
-
1190
.
102.
Schumm
M
,
Lang
P
,
Bethge
W
, et al
.
Depletion of T-cell receptor alpha/beta and CD19 positive cells from apheresis products with the CliniMACS device
.
Cytotherapy
.
2013
;
15
(
10
):
1253
-
1258
.
103.
Locatelli
F
,
Bauquet
A
,
Palumbo
G
,
Moretta
F
,
Bertaina
A
.
Negative depletion of α/β+ T cells and of CD19+ B lymphocytes: a novel frontier to optimize the effect of innate immunity in HLA-mismatched hematopoietic stem cell transplantation
.
Immunol Lett
.
2013
;
155
(
1-2
):
21
-
23
.
104.
Passweg
JR
,
Koehl
U
,
Uharek
L
,
Meyer-Monard
S
,
Tichelli
A
.
Natural-killer-cell-based treatment in haematopoietic stem-cell transplantation
.
Best Pract Res Clin Haematol
.
2006
;
19
(
4
):
811
-
824
.
105.
Norell
H
,
Moretta
A
,
Silva-Santos
B
,
Moretta
L
.
At the Bench: Preclinical rationale for exploiting NK cells and γδ T lymphocytes for the treatment of high-risk leukemias
.
J Leukoc Biol
.
2013
;
94
(
6
):
1123
-
1139
.
106.
Leveson-Gower
DB
,
Olson
JA
,
Sega
EI
, et al
.
Low doses of natural killer T cells provide protection from acute graft-versus-host disease via an IL-4-dependent mechanism
.
Blood
.
2011
;
117
(
11
):
3220
-
3229
.
107.
de Lalla
C
,
Rinaldi
A
,
Montagna
D
, et al
.
Invariant NKT cell reconstitution in pediatric leukemia patients given CD4- subset dynamics and correlates with transplantation defines distinct CD4 + and HLA-haploidentical stem cell remission state
.
J Immunol
.
2011
;
186
(
7
):
4490
-
4499
.
108.
Rubio
MT
,
Moreira-Teixeira
L
,
Bachy
E
, et al
.
Early posttransplantation donor-derived invariant natural killer T-cell recovery predicts the occurrence of acute graft-versus-host disease and overall survival
.
Blood
.
2012
;
120
(
10
):
2144
-
2154
.
109.
Or
R
,
Hadar
E
,
Bitan
M
, et al
.
Safety and efficacy of donor lymphocyte infusions following mismatched stem cell transplantation
.
Biol Blood Marrow Transplant
.
2006
;
12
(
12
):
1295
-
1301
.
110.
Hanley
PJ
,
Cruz
CR
,
Savoldo
B
, et al
.
Functionally active virus-specific T cells that target CMV, adenovirus, and EBV can be expanded from naive T-cell populations in cord blood and will target a range of viral epitopes
.
Blood
.
2009
;
114
(
9
):
1958
-
1967
.
111.
Linn
YC
,
Niam
M
,
Chu
S
, et al
.
The anti-tumour activity of allogeneic cytokine-induced killer cells in patients who relapse after allogeneic transplant for haematological malignancies
.
Bone Marrow Transplant
.
2012
;
47
(
7
):
957
-
966
.
112.
Leen
A
,
Gee
AP
,
Leung
K
, et al
.
Multi-virus-specific T-cell therapy for patients after hematopoietic stem cell and cord blood transplantation [abstract]
.
Blood
.
2013
;
122
(
21
).
Abstract 140
.
113.
Bar
M
.
Adoptive Immunotherapy for Acute Myeloid Leukemia: From Allogeneic Hematopoietic Cell Transplantation to CAR T Cells
.
J Leuk (Los Angel)
.
2014
;
2
:
134
.
114.
Kalos
M
,
Levine
BL
,
Porter
DL
, et al
.
T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia
.
Sci Transl Med
.
2011
;
3
(
95
):
95ra73
.
115.
Fujiwara
H
.
Adoptive immunotherapy for hematological malignancies using T cells gene-modified to express tumor antigen-specific receptors
.
Pharmaceuticals (Basel)
.
2014
;
7
(
12
):
1049
-
1068
.
116.
Gill
S
,
Tasian
SK
,
Ruella
M
, et al
.
Preclinical targeting of human acute myeloid leukemia and myeloablation using chimeric antigen receptor-modified T cells
.
Blood
.
2014
;
123
(
15
):
2343
-
2354
.
117.
Kochenderfer
JN
,
Dudley
ME
,
Carpenter
RO
, et al
.
Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation
.
Blood
.
2013
;
122
(
25
):
4129
-
4139
.
118.
Lazarus
HM
,
Koc
ON
,
Devine
SM
, et al
.
Cotransplantation of HLA-identical sibling culture-expanded mesenchymal stem cells and hematopoietic stem cells in hematologic malignancy patients
.
Biol Blood Marrow Transplant
.
2005
;
11
(
5
):
389
-
398
.
119.
Ning
H
,
Yang
F
,
Jiang
M
, et al
.
The correlation between cotransplantation of mesenchymal stem cells and higher recurrence rate in hematologic malignancy patients: outcome of a pilot clinical study
.
Leukemia
.
2008
;
22
(
3
):
593
-
599
.
120.
Kuzmina
LA
,
Petinati
NA
,
Parovichnikova
EN
, et al
.
Multipotent mesenchymal stromal cells for the prophylaxis of acute graft-versus-host disease-a phase II study
.
Stem Cells Int
.
2012
;
2012
:
968213
.
121.
Brunstein
CG
,
Miller
JS
,
Cao
Q
, et al
.
Infusion of ex vivo expanded T regulatory cells in adults transplanted with umbilical cord blood: safety profile and detection kinetics
.
Blood
.
2011
;
117
(
3
):
1061
-
1070
.
122.
Martelli
MF
,
Di Ianni
M
,
Ruggeri
L
, et al
.
HLA-haploidentical transplantation with regulatory and conventional T-cell adoptive immunotherapy prevents acute leukemia relapse
.
Blood
.
2014
;
124
(
4
):
638
-
644
.
123.
Brunstein
CG
,
Blazar
BR
,
Miller
JS
, et al
.
Adoptive transfer of umbilical cord blood-derived regulatory T cells and early viral reactivation
.
Biol Blood Marrow Transplant
.
2013
;
19
(
8
):
1271
-
1273
.
124.
Nierkens
S
,
Lankester
AC
,
Egeler
RM
, et al
;
Westhafen Intercontinental Group
.
Challenges in the harmonization of immune monitoring studies and trial design for cell-based therapies in the context of hematopoietic cell transplantation for pediatric cancer patients
.
Cytotherapy
.
2015
;
17
(
12
):
1667
-
1674
.
Sign in via your Institution