Open access peer-reviewed chapter

Assessment of Immune Reconstitution Following Hematopoietic Stem Cell Transplantation

Written By

Meenakshi Singh, Selma Z. D’Silva and Abhishweta Saxena

Submitted: 16 May 2019 Reviewed: 16 August 2019 Published: 03 December 2019

DOI: 10.5772/intechopen.89198

From the Edited Volume

Cells of the Immune System

Edited by Ota Fuchs and Seyyed Shamsadin Athari

Chapter metrics overview

1,254 Chapter Downloads

View Full Metrics

Abstract

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potential curative treatment for both congenital and hematological malignancies. Immune reconstitution after allogeneic hematopoietic stem cell transplantation is implicated in successful transplant outcomes such as overall survival and relapse-free survival. The reconstitution of immune cell subsets after HSCT occurs in different phases at different time points encompassing pre-engraftment, engraftment, and post-engraftment. The recovery of innate cellular immunity with the appearance of monocytes, dendritic cells, and natural killer cells in peripheral blood correlates with initiation of cellular engraftment. The cellular adaptive immunity is characterized by both thymic-independent expansion of T cells infused with graft and thymus-dependent expansion of naïve T cells derived from donor stem cells. The humoral immunity consists of B-cell reconstitution, which consists primarily of transitional and naïve subsets with the recovery of memory B cells that occur much later. In this review, we highlight the factors affecting immune reconstitution, the reconstitution of innate and adaptive immunity, techniques to assess immune reconstitution, and ways to enhance it.

Keywords

  • immune reconstitution
  • hematopoietic stem cell transplantation
  • innate immunity
  • adaptive immunity

1. Introduction

Hematopoietic stem cell transplantation (HSCT) is a choice of treatment for thousands of leukemic patients. The main outcome expected from HSCT is the lifetime engraftment of the donor graft. The preferred donor is a HLA matched-related donor; however, this is available in about 25% of the patients. Other options such as matched unrelated, matched cord blood units, and haploidentical-related donor also do exist. The success of HSCT is marred by conditions such as graft-versus-host disease (GvHD), relapse, treatment-related toxicity, and infection, which lead to higher morbidity and mortality [1]. The effectiveness of HSCT is dependent on the immune reconstitution in the host, which is linked to the number of active T and NK cells present in the graft. Delayed immune reconstitution results in unfavorable transplant outcomes; hence, faster immune reconstitution of donor origin is required for long-term survival of patients.

Soon after HSCT using myeloablative conditioning, the patient experiences a period of pancytopenia. It takes several months or years for immune reconstitution and for patients to regain immunocompetence after transplant. The immune cells start re-appearing in the following order: neutrophils (0.5 months), monocytes (1 month), NK cells (1 month), T cells (2 months), and B cells (3 months); however, the normal levels are reached much later (Figure 1) [2].

Figure 1.

The time taken for different immune subsets to (A) reappear in circulation and (B) reach normal levels after hematopoietic stem cell transplantation.

There are various factors affecting immune reconstitution after transplant such as

  1. thymic damage (age-related or pre-transplant conditioning regimens)

  2. source of stem cells

  3. HLA disparity between donor and host

  4. post-transplant immune suppressant

  5. occurrence of graft-versus-host disease.

Age or pre transplant chemotherapy or radiation leads to thymic damage. The severity of the damage caused to the thymus depends on the dose of the drugs used and also on the age of the patients, which in turn affect the immune recovery. In younger patients (<18 years), there is faster thymic regeneration after chemotherapy than older patients [3]. The age of the donor also affects the engraftment and reconstitution potential of hematopoietic stem cells as shown in mouse models [4]. Moreover, the thymic recovery is faster and is associated with faster T-cell reconstitution and recovery of normal T-cell repertoire in autologous (9 months) than allo-HSCT (12 months) [5]. This delayed thymic-dependent immune reconstitution is further reduced by the occurrence of aGvHD after allogeneic HSCT [6, 7].

The source of stem cells used as graft could be either bone marrow, peripheral blood, or cord blood. Source of stem cells used predicts the rate of immune reconstitution. It has been observed that platelet (20 × 109/L) reconstitution is faster in peripheral blood (11–18 days) than bone marrow (17–25 days) HSCT. Similarly, neutrophil (>0.5 × 109/L) reconstitution is also faster in peripheral blood (12–19 days) than bone marrow (15–23 days) HSCT. This is because of the presence of long-term HSCs and more committed multipotent progenitors in the peripheral blood than bone marrow [8]. Further as compared to transplantation using in vivo or ex vivo T-cell depleted graft, faster immune reconstitution is seen in unmodified graft transplantation [9]. Using peripheral blood graft, faster reconstitution of CMV-specific cytotoxic T cells and CD4+ T cells is observed than stem cells from bone marrow source [10, 11]. The advantages of using umbilical cord blood units are its ready availability and its ability to cross the HLA barrier. The rates of engraftment and post-transplant outcomes are dependent on the number of total nucleated cells (TNCs) and CD34+ cell dose present in the graft source. Martin et al. [12] previously reported high TNC dose in association with positive transplant outcomes such as improved overall survival (OS), lower relapse rate (RR), and increased risk of chronic GvHD. Since there is a higher number of TNCs in the bone marrow and peripheral blood, there is faster engraftment (~14–21 days) after HSCT using this source of graft than umbilical cord blood source (~30 days) [13, 14]. Remberger et al. [15] reported faster engraftment but poor survival and higher relapse after HSCT using high CD34+ cell dose peripheral blood as graft source. Various researchers have reported immune cell reconstitution using different cell sources (Table 1).

Cells/L type of transplant NK cells 1 month CD4+ T cells 90 days CD8+ T cells 90 days B cells 90 days Reference
Matched sibling donor 220 645 33 [16]
Matched unrelated donor 253 198 447 43 [17]
Haploidentical donor 152 672 23 [16]
T-cell depleted 357 7 7 55 [18]
T-cell replete 183 127 181 64 [18]

Table 1.

Reconstitution of various immune subsets in different types of HSCT.

Graft manipulations such as T-cell depletion (TCD) have resulted in lower chances of GvHD and graft rejection in unrelated and HLA mismatched transplants. However, T-cell depletion results in delayed immune reconstitution and increased morbidity and mortality due to infection [19, 20, 21]. An advantage of using T-cell depletion is that in case of malignancies it also leads to better GVL effect depending on the malignant disease being treated. For example, in CML, TCD is related to increased relapse rate [22], whereas in AML and AML cohorts, lower rate of relapse has been observed in TCD transplantation [23, 24, 25].

The degree of HLA mismatch is an important factor in immune reconstitution. It has been observed that the outcomes from matched unrelated transplantation are at par with that of matched related transplantation [1]. Chang et al. reported similar reconstitution of T-cell subsets, except for CD4+ cells and CD4+ naïve T cells, in haploidentical and HLA-matched transplantation [16]. Various researchers have reported reconstitution of immune cells following different transplant strategies. It has been observed that the immune reconstitution is best in matched sibling related followed by matched unrelated donor, haploidentical donor, T-cell replete, and T-cell depleted transplants.

Conditioning regimens deplete host immune system, eliminate the leukemic cells, and create space for engraftment of the donor cells. Although this eliminates the patient’s leukemic cells, it also reduces the alloreactivity between host and donor cells after HSCT and further results in severe depletion of all immune cells. The use of drugs such as ATG or alemtuzumab depletes the host T cells further and results in a delayed recovery of donor-derived T cells. Increase in the severity of the conditioning regimen results in prolonged immune deficiency after transplant [26].

Both thymus-dependent and thymus-independent T-cell reconstitutions are affected by the increase in HLA mismatch between the patient and the donor, probably because of higher risk of GvHD [27]. Clave et al. [28] reported higher reconstitution of both CD4+ and CD8+ T cells in transplants involving unrelated cord blood grafts (190 cells × 103/μL for CD4+ and 280 cells × 103/μL for CD8+) than CD34 selected peripheral blood haploidentical donor grafts (68 cells × 103/μL for CD4+ and 80 cells × 103/μL for CD8+). Mehta et al. [29] showed lower reconstitution of absolute CD4+ and CD8+ T cells at 3 months and higher B-cell counts (6 months) after unrelated cord blood HSCT than HLA matched HSCT (121.53 vs. 261.18 for CD4+, 36.03 vs. 190.56 for CD8+, and 210 vs. 31.2 for B cells). There was similar reconstitution of B cells but lower CD4+ and CD8+ T-cell reconstitution in single unit umbilical cord blood transplantation than HLA mismatched donor HSCT (11 vs. 9 for B cells, 15 vs. 21 for CD4+ cells, and 14 vs. 21 for CD8+ cells) [30].

Acute graft-versus-host disease occurs when donor lymphocytes react against normal host tissue to cause serious complications after allogeneic HSCT. Although there is faster recovery of the innate immune system after allo-HSCT, lymphocyte recovery is delayed due to aGvHD [3, 31]. The recovery of T cells depends on the thymic efficiency as well as the peripheral niche, which provides resources for T-cell survival. As GvHD targets the bone marrow, in patients with graft-versus-host disease, the peripheral resources are reduced because of which there is increased immunosuppression leading to delayed T-cell reconstitution in allogeneic HSCT as compared to autologous HSCT. The options to increase the efficiency of T-cell reconstitution must be selected in a manner so as to not aggravate the already present GvHD [32, 33]. Similarly, the drugs used to treat GvHD can also result in delayed immune reconstitution. Drugs such as cyclosporine A and methotrexate interfere with the T-cell receptor signaling and hence result in alteration of peripheral T-cell survival and B-cell differentiation [34, 35]. Tyrosine kinase inhibitors like imatinib mesylate used for controlling refractory cGvHD also lower T-cell survival by interfering with T-cell receptor (TCR) or IL7 signaling [36, 37]. Reconstitution of dendritic cells is decreased in GvHD [38]. Conversely, it has been suggested that depletion or inactivation of the host dendritic cells before allogeneic HSCT reduces the occurrence of GvHD [39, 40, 41].

Advertisement

2. Reconstitution of innate immunity

After HSCT, the first cells to engraft are the monocytes, followed by granulocytes, platelets, and NK cells [42]. Monocytes are primarily involved in phagocytosis and release of cytokines. They are classified into classical (CD14++CD16-), intermediate (CD14++CD16+), and nonclassical (CD14+CD16++) based on the expression of CD14 and CD16 [43, 44]. Monocytes remain below the normal levels for up to a year [45, 46].

The conditioning regimen used prior transplant results in a neutropenic phase till the neutrophils reconstitute, which takes approximately 11–12 days in T-cell depleted haploidentical HSCT [47, 48]. Although neutrophil counts rise to normal numbers within 2 weeks after transplant [49], they become functionally competent only after 2 months [50, 51]. The type of graft affects the reconstitution of neutrophils: 2 weeks in case of GCSF mobilized grafts, 3 weeks in case of bone marrow, and around 4 weeks in umbilical cord blood [1]. Use of peripheral blood has decreased the neutrophil recovery time from an average of 16 to 12 days [52].

NK cells recover in both number and function within the first few weeks after transplant [53], and functional reconstitution of NK cells is reached within 2 months [1]. The time taken for NK-cell reconstitution is dependent on the occurrence of GvHD [47, 54] and does not differ if the source of stem cells is peripheral blood or bone marrow [55]. However, the number of functional NK cells is higher when the transplant involves T-cell replete grafts than T-cell depleted grafts [56]. The most prominent functional NK cells after transplant are CD56brightCD16dim [57, 58]. Also, higher overall survival is seen in patients with high CD56bright NK cells at day 14 after unmanipulated haploidentical HSCT. The cytolytic function of NK cells is regulated by the interaction of inhibitory/activating killer immunoglobulin like receptors (KIRs) present on their surface and their specific HLA class I ligands. The reconstitution of the inhibitory and activating KIRs is dependent on factors such as conditioning regimen, T-cell deplete/replete graft, and immunosuppression used after transplant.

In a study evaluating NK-cell reconstitution after matched related/unrelated donor HSCT, it has been reported that the NK-cell counts are lower for longer period (2-3 months) after MUD (156/μL) than MRD (265/μL). The most frequent immature NK cells were CD56bright and NKG2A+CD57-CD56dim NK cells [59]. Russo et al. [60] reported that in haploidentical HSCT using after transplant cyclophosphamide, the immature NK cell starts appearing at 2 weeks; however, the mature NK cells expressing CD16 and CD56 and NKG2A appear at about a year.

Host dendritic cells that escape chemotherapy/radiation activate alloantigenic T cells in the donor and hence play an important role in GvHD. Since host dendritic cells present MHC antigens to donor CD8+ T cells after transplant, depleting these cells could result in lower risk of GvHD [61, 62]. Lower reconstitution of lymphoid dendritic cells has been associated with inferior overall survival [63].

Gamma delta T cells make up ~5% of the T-cell population, and their receptors are composed to gamma and delta chains. These T cells have been reported to enhance engraftment and graft-versus-leukemia effect without an increase in GvHD [64]. Gamma delta T cells reconstitute faster in patients in whom bone marrow (60 days) is used as the graft source than peripheral blood (200 days) [65].

Advertisement

3. Reconstitution of adaptive immunity

T-cell reconstitution is faster in transplantation with peripheral blood as graft source than bone marrow due to higher number of T cells present in the graft [55]. Ciurea et al. [18] reported better T-cell reconstitution in recipients of T-cell replete haploidentical HSCT than recipients of T-cell depleted haploidentical HSCT at 6 months after transplant. Use of ATG for T-cell depletion also affects the rate of immune reconstitution. This effect is more prominent in umbilical cord blood transplantation than bone marrow transplantation. T-cell reconstitution in allo-HSCT without the use of ATG is seen in about 7–12 months when using bone marrow and umbilical cord as stem cell source as compared to 6–24 months when using peripheral blood as stem cell source [66]. T cells recover primarily via peripheral expansion of memory T cells or endogenous T-cell development. Hence, functional thymus is required for effective reconstitution of T cells [67]. This is an issue in aging patients where there is thymus atrophy [68]. Due to this, although full immune recovery is possible in middle-aged patients, it is not possible in older patients and is a cause of morbidity and mortality [69]. Reconstitution of T cells is slow probably due to the prolonged depletion and reduced function of naïve T cells [70]. T cells that reconstitute are primarily from the donor origin in case of T-cell replete transplant or host T cells that have escaped the conditioning regimen in case of T-cell depleted transplant. Naïve T cells/T-cell receptor excision circles (TRECs) are lower for approximately 10–30 years after transplant [71, 72]. Reconstitution of functional T cells as observed by their ability to secrete interferon gamma and interleukin-4 to normal levels returns in 30 days after haploidentical HSCT for patients in whom acute GvHD is not observed [73]. Recipients of T-cell depleted haploidentical HSCT show higher CD31+ naïve CD4+ T cells than their donors at approximately 4–6 years [74]. Homeostatic peripheral expansion is induced by various homeostatic cytokines such as IL7 and IL15, inflammatory cytokines, and viral exposure. Peripheral homeostatic expansion leads to an inverse CD4/CD8 ratio in patients for several months after transplant. CD4 counts are considered as the best predictive marker for the recovery of immune competence after HSCT, and its recovery has also been associated with lower risk of infections and improved transplant outcomes [1]. CD4+ T-cell counts are as low as <200 cells/μL in the first 3 months and reach levels of 450 cells/μL at about 5 years after transplant [55, 75]. CD8+ T-cell counts increase rapidly during the first 3 months after transplant possibly due to the expansion of herpesvirus-specific CD8 T cells [55, 76]. GvHD reduces the number of CD4+ T cells by inhibiting the thymic output, whereas CD8+ cells increase in number during GvHD or CMV reactivation [77, 78]. The reconstituting CD4+ T cells have a higher expression of CD11a, CD29, CD45RO, and HLA-DR and a lower expression of CD28, CD45RA, and CD62L than normal individuals [79, 80]. The early reconstituting CD8+ T cells are mostly memory or effector cells. Naïve or TREC+CD8+ T cells recover at a slower rate [77, 81]. The number of regulatory T cells (Tregs) is much higher after transplant than normal individuals and may contribute to remission [82, 83]. A Treg:CD4+ T cell ratio of less than 9% has been associated with higher risk of aGvHD [84]. Chang et al. [16] reported lower CD4+ T cells, dendritic cells, and higher CD28 expression on CD4+ and CD8+ T cells in patients receiving haploidentical HSCT than patients receiving HLA matched HSCT.

B-cell reconstitution is also delayed after HSCT: ~6 months for autologous and ~9 months after allogeneic transplantation and is mainly due to GvHD or its treatment. In the first 2 months after transplant, B-cell counts are low but rise higher than the normal levels in approximately 1–2 years [55, 85]. Since restoration of full humoral immune functioning requires both naïve and memory B cells, all patients who have undergone HSCT remain susceptible to infections for at least a year after transplant [1]. The reconstituted B cells express higher levels of CD1c, CD38, CD5, membrane IgM, and membrane IgD and lower levels of CD25 and CD26L than normal individuals [86].

A number of studies have reported comparisons between reconstitution of different immune cells depending on the graft source. Faster reconstitution of different immune cells was observed when bone marrow was used as graft source as compared to peripheral blood or cord blood (Table 2).

Cell type and numbers Bone marrow Peripheral blood Unrelated cord blood Reference
Neutrophils (>0.5 × 109/L) 16 days 15 days 19 days [87]
Natural killer cells (>0.1 × 109/L) 1.5 months 4 months 4 months [16, 87]
T cells (>0.5 × 109/L) CD4 2–3 months 6 months 3 months [28, 88]
Naïve T cells (>0.5 × 109/L) 9 months 24 months 12 months [87, 89]
Cytotoxic T cells (>0.25 × 109/L 3 months 9 months 8 months [65, 90]
T helper cells (>0.2 × 109/L) 4 months 10 months 1 months [65, 90]

Table 2.

Reconstitution of different immune cells depending on the graft source.

Advertisement

4. Assessment of post-transplant immune recovery

There are different methods to assess the immune recovery after transplant, such as estimation of absolute lymphocyte count (ALC), levels of immune cell subsets (NK cells, B cells, and T cells), and antibody titers to assays for T- and B-cell repertoires [91].

ALC levels have been reported in association with overall survival and rate of relapse. ALC >500 cells/μL on day 15 is linked with better OS and lower relapse after autologous as well as allogeneic transplantation [92, 93]. An increase in the levels of CD16+ monocytes has been associated with aGvhD [94].

Early recovery of CD4+ T cells is associated with overall survival, nonrelapse mortality, and risk of infections [95, 96]. Admiral et al. [97] reported the time taken by circulating CD4+ T cells to reach 0.5× 109/L as a strong marker for probability of relapse. In myeloablative allogeneic HSCT, higher levels of CD3+, CD8+ T cells, regulatory T cells, and myeloid dendritic cells are correlated with relapse-free survival [98].

Recently, flow cytometric analysis has been used to differentiate between the T, B, and NK-cell subpopulations. Low levels of NK cells within the first few weeks after transplant have been associated with poor transplant outcomes like lower overall survival and higher risk of infection [99, 100]. Surface markers such as CD45RA, CD28, CD27, CD62L, and CCR7 can be used to differentiate naïve, effector, effector memory, and central memory CD4+ and CD8+ subsets [101, 102]. The surface markers expressed by naïve T cells are CD45RA+CCR7+; central memory T cells are CD45RA-CCR7+; effector memory T cells are CD45RA-CCR7–; and effector T cells are CD45RA+CCR7– [91]. CD4+ T cells also include regulatory T cells (CD25+FoxP3+) and Th17 cells [103, 104]. The expression of CD27, IgM, and IgD helps in distinguishing between naïve B cells (CD27-IgD+), memory B cells (CD27+IgD+), and isotype switched memory B cells (CD27+IgD-) [105]. Myeloid and plasmacytoid dendritic cells can be distinguished based on the expression of CD123 and CD11c: CD123low CD11c+(myeloid) and CD123bright CD11c- (plasmacytoid) [106].

TRECs have been suggested as a marker for reconstitution of naïve T cells (CD4+CD45RA+) derived from the thymus. TRECs, however, remain low up to 6 months after HSCT [107]. Due to thymic atrophy with age, older patients have T cells with low TCR repertoire, which leads to higher risk of infections leading to lower transplant outcomes [108, 109]. Thymopoiesis can also be evaluated by measuring the number of TRECs by real-time quantitative in purified CD4+ and CD8+ T cells [110]. Lewin et al. [111] reported faster recovery of TRECs in younger patients and patients who received conventional grafts as compared to T-cell depleted grafts. Lower levels of TRECs are associated with GvHD and opportunistic infections [77, 112].

Certain cytokines can also be used as predictive markers for transplant outcomes. One such marker is IL7, which can be used to evaluate successful T-cell recovery. Increased IL7 is associated with delayed reconstitution and increased mortality and aGvHD [113]. High levels of IL6, GCSF, and IL2α have also been indicated in association with risk of aGvHD [96, 114]. For assessing chronic GvHD, high levels of IL8 and low levels of IL17A have been suggested [103, 115]. Min et al. [104] have also correlated high levels of IL6 and IL10 with poor transplant-related outcomes.

Further, T- and B-cell receptor repertoire gene arrangements can be evaluated by molecular techniques such as next generation sequencing [116, 117]. Michalek et al. [118] have demonstrated β chain sequencing of the T-cell receptor in order to identify the T-cell clones that could mediate either graft-versus-host disease or graft-versus-leukemia effect. Brink et al. [9] reported higher diversity in CD4+ T cells than CD8+ T cells following allogeneic HSCT. Greater diversity was observed in cord blood grafts, followed by unmanipulated grafts and T-cell depleted grafts.

Advertisement

5. Strategies to improve immune reconstitution

Many strategies, such as administration of recombinant cytokines, adoptive cell therapy, and hormone-based therapies, have recently been used to improve immune reconstitution after transplantation.

IL7 cytokine has been shown to effectively enhance reconstitution of T and B lymphoid cells by enabling thymopoiesis [105, 119]. It has been demonstrated that IL7 increased the CD3+, CD4+, and CD8+ T-cell levels to more than four folds and also leads to increase in functional and diverse T cells [120]. Administering IL-7 predominantly increases the naïve CD8+ T cells. The timing of administering is, however, important, as administering early after transplant aggravates GvHD [116, 121], whereas administering it at a later stage after HSCT results in lower risk of GvHD. This is contributed by the activation of alloreactive T cells that express lower IL-7Rα levels [32, 38]. Other cytokines that enable immune reconstitution are insulin-like growth factor 1(IGF-1), IL22, IL15, and IL12 [122, 123, 124]. IL15 has been shown to significantly increase the reconstitution of CD8+ T cells and NK cells and improve the GvL effect in haploidentical murine models [125]. Sauter et al. [126] reported better lymphocyte reconstitution after IL-15 administration in T-cell depleted allogeneic HSCT; however, it has been shown to worsen GvHD.

Recently, it has been suggested that modulating the function of dendritic cells could reduce GvHD while maximizing GvL [127]. Studies on reconstitution of dendritic cells after HSCT have been contradictory. Maraskovsky et al. [128] have shown that treatment with Flt3-L can expand DC subsets; however, when administered after HSCT, it can worsen GvHD [38]. Gauthier et al. [38] have demonstrated that SDF-1α therapy can expand the DC1 subsets and lower the severity of GvHD. Because of their immunosuppressive properties, mesenchymal stem cells have recently been used for suppressing GvHD [129, 130, 131]. Mesenchymal stem cells release cytokines such as IL-7, which improve T-cell survival and promote reconstitution of dendritic cells by secreting SDF-1α [132].

NK-cell immunotherapy is one of the novel strategies underway to reduce GvHD and enhance graft-versus-leukemia effect in a KIR-HLA mismatched haploidentical HSCT [133, 134, 135].

Advertisement

6. Future directions

Recently, few studies have identified the association of reconstitution of certain immune subsets with predicting post-HSCT outcomes. However, these studies are often limited by small sample size, lack of detailed immune reconstitution, and secretome profile, which could be used as biomarkers to predict immune reconstitution. Prospective studies involving a large number of patients should be conducted to determine which immune factors and tests to detect the same could have prognostic value and understand the impact of such predictive risk factors on transplant outcomes. This is most beneficial, especially for recipients of haploidentical HSCT, in which a routine strategy could be adopted to result in faster immune reconstitution and hence lower probability of poor transplant outcomes, such as TRM, relapse, and GvHD.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Mackall C, Fry T, Gress R, Peggs K, Storek J, Toubert A. Background to hematopoietic cell transplantation, including post transplant immune recovery. Bone Marrow Transplantation. 2009;44:457-462. DOI: 10.1038/bmt.2009.255
  2. 2. Storek J. Immunological reconstitution after hematopoietic cell transplantation – Its relation to the contents of the graft. Expert Opinion on Biological Therapy. 2008;8(5):583-597. DOI: 10.1517/14712598.8.5.583
  3. 3. Mackall CL, Fleisher TA, Brown MR, Andrich MP, Chen CC, Feuerstein IM, et al. Age, thymopoiesis, and CD4+ T-lymphocyte regeneration after intensive chemotherapy. The New England Journal of Medicine. 1995;332:143-149. DOI: 10.1056/NEJM199501193320303
  4. 4. Liang Y, Van Zant G, Szilvassy SJ. Effects of aging on the homing and engraftment of murine hematopoietic stem and progenitor cells. Blood 2005; 106: 1479-1487. DOI: 10.1182/blood-2004-11-4282
  5. 5. Olkinuora H, Talvensaari K, Kaartinen T, Siitonen S, Saarinen-Pihkala U, Partanen J, et al. T cell regeneration in pediatric allogeneic stem cell transplantation. Bone Marrow Transplantation. 2007;39(3):149-156. DOI: 10.1038/sj. bmt.1705557
  6. 6. Muller-Hermelink HK, Sale GE, Borisch B, Storb R. Pathology of the thymus after allogeneic bone marrow transplantation in man. A histologic immu-nohistochemical study of 36 patients. The American Journal of Pathology. 1987;129(2):242-256
  7. 7. Lum LG. The kinetics of immune reconstitution after human marrow transplantation. Blood. 1987;69(2):369-380
  8. 8. Korbling M, Anderlini P. Peripheral blood stem cell versus bone marrow allotransplantation: Does the source of hematopoietic stem cells matter? Blood. 2001;98:2900-2908. DOI: 10.1182/blood.v98.10.2900
  9. 9. van den Brink MRM, Velardi E, Perales MA. Immune reconstitution following stem cell transplantation. Hematology. 2015;2015(1):215-219. doi: 10.1182/asheducation-2015.1.215
  10. 10. Hakki M, Riddell SR, Storek J, Carter RA, Stevens-Ayers T, Sudour P, et al. Immune reconstitution to cytomegalovirus after allogeneic hematopoietic stem cell transplantation: Impact of host factors, drug therapy, and sub-clinical reactivation. Blood. 2003;102(8):3060-3067. DOI: 10.1182/blood-2002- 11-3472
  11. 11. Cwynarski K, Ainsworth J, Cobbold M, Wagner S, Mahendra P, Apperley J, et al. Direct visualization of cytomegalovirus-specific T-cell reconstitution after allogeneic stem cell transplantation. Blood. 2001;97(5):1232-1240. DOI: 10.1182/blood.V97.5.1232
  12. 12. Martin PS, Li S, Nikiforow S, Alyea EP, Antin JH, Armand P, et al. Infused total nucleated cell dose is a better predictor of transplant outcomes than CD34(+) cell number in reduced-intensity mobilized peripheral blood allogeneic hematopoietic cell transplantation. Haematologica. 2016;101(4):499-505. DOI: 10.3324/haematol.2015.134841
  13. 13. Seggewiss R, Einsele H. Immune reconstitution after allogeneic transplanta-tion and expanding options for immunomodulation: An update. Blood. 2010;115(19):3861-3868. DOI: 10.1182/blood-2009-12-234096
  14. 14. Danby R, Rocha V. Improving engraftment and immune reconstitution in umbilical cord blood transplantation. Frontiers in Immunology. 2014;5:68. DOI: 10.3389/fimmu.2014.00068
  15. 15. Remberger M, Törlén J, Ringdén O, Engström M, Watz E, Uhlin M, et al. Effect of total nucleated and CD34+ cell dose on outcome after allogeneic hematopoietic stem cell transplantation. Biology of Blood and Marrow Transplantation. 2015;21(5):889-893. DOI: 10.1016/j.bbmt.2015.01.025
  16. 16. Chang YJ, Zhao XY, Huo MR, Xu LP, Liu DH, Liu KY, et al. Immune reconstitution following Unmanipulated HLAMismatched/Haploidentical transplantation compared with HLA-identical sibling transplantation. Journal of Clinical Immunology. 2012;32:268-280. DOI: 10.1007/s10875-011-9630-7
  17. 17. Pérez-Martínez A, González-Vicent M, Valentín J, Aleo E, Lassaletta A, Sevilla J, et al. Early evaluation of immune reconstitution following allogeneic CD3/CD19-depleted grafts from alternative donors in childhood acute leukemia. Bone Marrow Transplantation. 2012;47:1419-1427. DOI: 10.1038/bmt.2012.43
  18. 18. Ciurea SO, Mulanovich V, Saliba RM, Bayraktar UD, Jiang Y, Bassett R, et al. Improved early outcomes using a T cell replete graft compared with T cell depleted Haploidentical hematopoietic stem cell transplantation. Biology of Blood and Marrow Transplantation. 2012;18(12):1835-1844. DOI: 10.1016/j.bbmt.2012.07.003
  19. 19. Cavazzana-Calvo M, Carlier F, Le Deist F, Morillon E, Taupin P, Gautier D, et al. Long-term T-cell reconstitution after hematopoietic stem-cell transplantation in primary T-cell-immunodeficient patients is associated with myeloid chimerism and possibly the primary disease phenotype. Blood. 2007;109:4575-4581. DOI: 10.1182/blood-2006-07-029090
  20. 20. Müller SM, Kohn T, Schulz AS, Debatin KM, Friedrich W. Similar pattern of thymic-dependent T-cell reconstitution in infants with severe combined immunodeficiency after human leukocyte antigen (HLA)-identical and HLA-nonidentical stem cell transplantation. Blood. 2000;96:4344-4349
  21. 21. Neven B, Leroy S, Decaluwe H, Le Deist F, Picard C, Moshous D, et al. Long-term outcome after hematopoietic stem cell transplantation of a single-center cohort of 90 patients with severe combined immunodeficiency. Blood. 2009;113:4114-4124. DOI: 10.1182/blood-2008-09-177923
  22. 22. Sehn LH, Alyea EP, Weller E, Canning C, Lee S, Ritz J, et al. Comparative outcomes of T-cell-depleted and non-T-cell-depleted allogeneic bone marrow transplantation for chronic myelogenous leukemia: Impact of donor lymphocyte infusion. Journal of Clinical Oncology. 1999;17:561-568
  23. 23. Papadopoulos EB, Carabasi MH, Castro-Malaspina H, Childs BH, Mackinnon S, Boulad F, et al. T-cell-depleted allogeneic bone marrow transplantation as postremission therapy for acute myelogenous leukemia: Freedom from relapse in the absence of graft-versus-host disease. Blood. 1998;91:1083-1090
  24. 24. Jakubowski AA, Small TN, Kernan NA, Castro-Malaspina H, Collins N, Koehne G, et al. T cell-depleted unrelated donor stem cell transplantation provides favorable disease-free survival for adults with hematologic malignancies. Biology of Blood and Marrow Transplantation. 2011;17:1335-1342. DOI: 10.1016/j.bbmt.2011.01.005
  25. 25. 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;17:1343-1351. DOI: 10.1016/j.bbmt.2011.02.002
  26. 26. Soderling CC, Song CW, Blazar BR, Vallera DA. A correlation between conditioning and engraftment in recipients of MHC-mismatched T cell-depleted murine bone marrow transplants. Journal of Immunology. 1985;135:941-946
  27. 27. Politikos I, Boussiotis VA. The role of the thymus in T-cell immune reconstitution after umbilical cord blood transplantation. Blood. 2014;124:3201-3211. DOI: 10.1182/blood-2014-07-589176
  28. 28. Clave E, Lisini D, Douay C, Giorgiani G, Busson M, Zecca M, et al. Thymic function recovery after unrelated donor cord blood or T-cell depleted HLA-haploidentical stem cell transplantation correlates with leukemia relapse. Frontiers in Immunology. 2013;4:54. DOI: 10.3389/fimmu.2013.00054
  29. 29. Mehta RS, Bejanyan N, Cao Q , Luo X, Brunstein C, Cooley S, et al. Immune reconstitution after umbilical cord blood versus peripheral blood progenitor cell transplantation in adults following myeloablative conditioning. Blood. 2016;22:2246
  30. 30. Servais S, Lengline E, Porcher R, Carmagnat M, Peffault de Latour R, Robin M, et al. Long-term immune reconstitution and infection burden after mismatched hematopoietic stem cell transplantation. Biology of Blood and Marrow Transplantation. 2014;20:507-517. DOI: 10.1016/j.bbmt.2014.01.001
  31. 31. Fry TJ, Mackall CL. Immune reconstitution following hematopoietic pro-genitor cell transplantation: Challenges for the future. Bone Marrow Transplantation. 2005;35:S53-S57. DOI: 10.1038/sj.bmt.1704848
  32. 32. Sinha ML, Fry TJ, Fowler DH, Miller G, Mackall CL. Interleukin 7 worsens graft-versus-host disease. Blood. 2002;100(7):2642-2649. DOI: 10.1182/blood- 2002-04-1082
  33. 33. Blaser BW, Roychowdhury S, Kim DJ, Schwind NR, Bhatt D, Yuan W, et al. Donor-derived IL-15 is critical for acute allogeneic graft-versus-host disease. Blood. 2005;105(2):894-901. DOI: 10.1182/blood-2004-05-1687
  34. 34. Hannam-Harris AC, Taylor DS, Nowell PC. Cyclosporin a directly inhibits human B-cell proliferation by more than a single mechanism. Journal of Leukocyte Biology. 1985;38(2):231-239. DOI: 10.1002/jlb.38.2.231
  35. 35. Gratama JW, Würsch AM, Nissen C, Gratwohl A, D'Amaro J, de Gast GC, et al. Influence of graft-versus-host disease prophylaxis on early T-lymphocyte regeneration following allogeneic bone marrow transplantation. British Journal of Haematology. 1986;62(2):355-365. DOI: 10.1111/j.1365-2141.1986.tb02939.x
  36. 36. Legros L, Ebran N, Stebe E, Rousselot P, Rea D, Cassuto JP, et al. Imatinib sensitizes T-cell lymphocytes from chronic myeloid leukemia patients to FasL-induced cell death: A brief communication. Journal of Immunotherapy. 2012;35(2):154-158. DOI: 10.1097/CJI.0b013e318243f238
  37. 37. Thiant S, Moutuou MM, Laflamme P, Sidi Boumedine R, Leboeuf DM, Busque L, et al. Imatinib mesylate inhibits STAT5 phosphorylation in response to IL-7 and promotes T cell lymphopenia in chronic myelogenous leukemia patients. Blood Cancer Journal. 2017;7(4):e551. DOI: 10.1038/bcj. 2017.29
  38. 38. Gauthier SD, Leboeuf D, Manuguerra-Gagne R, Gaboury L, Guimond M. Stromal-derived factor-1alpha and interleukin-7 treatment improves homeostatic proliferation of naive CD4(+) T cells after allogeneic stem cell transplantation. Biology of Blood and Marrow Transplantation. 2015;21(10):1721-1731. DOI: 10.1016/j. bbmt.2015.06.019
  39. 39. Chan GW, Gorgun G, Miller KB, Foss FM. Persistence of host dendritic cells after transplantation is associated with graft-versus-host disease. Biology of Blood and Marrow Transplantation. 2003;9(3):170-176. DOI: 10.1016/S1083-8791(03)70006-8
  40. 40. Arpinati M, Chirumbolo G, Urbini B, Bonifazi F, Bandini G, Saunthararajah Y, et al. Acute graft-versus-host disease and steroid treatment impair CD11c+ and CD123+ dendritic cell reconstitution after allogeneic peripheral blood stem cell transplantation. Biology of Blood and Marrow Transplantation. 2004;10(2):106-115. DOI: 10.1016/j.bbmt.2003.09.005
  41. 41. Vakkila J, Thomson AW, Hovi L, Vettenranta K, Saarinen-Pihkala UM. Circulating dendritic cell subset levels after allogeneic stem cell transplantation in children correlate with time post transplant and severity of acute graft-versus-host disease. Bone Marrow Transplantation. 2005;35(5):501-507. DOI: 10.1038/sj.bmt.1704827
  42. 42. Storek J, Geddes M, Khan F, Huard B, Helg C, Chalandon Y, et al. Reconstitution of the immune system after hematopoietic stem cell transplantation in humans. Seminars in Immunopathology. 2008;30:425-437. DOI: 10.1007/s00281-008-0132-5
  43. 43. Ziegler-Heitbrock L, Ancuta P, Crowe S, Dalod M, Grau V, Hart DN, et al. Nomenclature of monocytes and dendritic cells in blood. Blood. 2010;116:e74-e80. DOI: 10.1182/blood-2010-02-258558
  44. 44. Passlick B, Flieger D, Ziegler-Heitbrock HW. Identification and characterization of a novel monocyte subpopulation in human peripheral blood. Blood. 1989;74:2527-2534
  45. 45. Cayeux S, Meuer S, Pezzutto A, Körbling M, Haas R, Schulz R, et al. Allogeneic mixed lymphocyte reactions during a second round of ontogeny: Normal accessory cells did not restore defective interleukin-2 (IL-2) synthesis in T cells but induced responsiveness to exogeneous IL-2. Blood. 1989;74:2278-2284
  46. 46. Sahdev I, O’Reilly R, Black P, Heller G, Hoffmann M. Interleukin-1 production following T-cell-depleted and unmodified marrow grafts. Pediatric Hematology and Oncology. 1996;13:55-67
  47. 47. van Rood JJ, Loberiza Jr FR, Zhang MJ, Oudshoorn M, Claas F, Cairo MS, Champlin RE, Gale RP, Ringdén O, Hows JM, Horowitz MH. Effect of tolerance to noninherited maternal antigens on the occurrence of graft versus- host disease after bone marrow transplantation from a parent or an HLA-haploidentical sibling. Blood 2002;99:1572-1577. DOI: 10.1182/blood.v99.5.1572
  48. 48. Passweg JR, Tichelli A, Meyer-Monard S, Heim D, Stern M, Kühne T, et al. Purified donor NK-lymphocyte infusion to consolidate engraftment after haploidentical stem cell transplantation. Leukemia. 2004;18:1835-1838. DOI: 10.1038/sj.leu.2403524
  49. 49. Zimmerli W, Zarth A, Gratwohl A, Speck B. Neutrophil function and pyogenic infections in bone marrow transplant recipients. Blood. 1991;77:393-399
  50. 50. Atkinson K, Biggs JC, Downs K, Juttner C, Bradstock K, Lowenthal RM, et al. GM-CSF after allogeneic BMT: Accelerated recovery of neutrophils, monocytes and lymphocytes. Australian and New Zealand Journal of Medicine. 1991;21:686-692
  51. 51. Bensinger WI, Clift R, Martin P, Appelbaum FR, Demirer T, Gooley T, et al. Allogeneic peripheral blood stem cell transplantation in patients with advanced hematologic malignancies: A retrospective comparison with marrow transplantation. Blood. 1996;88:2794-2800
  52. 52. Przepiorka D, Smith TL, Folloder J, Anderlini P, Chan KW, Körbling M, et al. Controlled trial of filgrastim for acceleration of neutrophil recovery after allogeneic blood stem cell transplantation from human leukocyte antigen-matched related donors. Blood. 2001;97(11):3405-3410. DOI: 10.1182/blood.v97.11.3405
  53. 53. Jacobs R, Stoll M, Stratmann G, Leo R, Link H, Schmidt RE, et al. Natural killer cells after bone marrow transplantation. Blood. 1992;79:3239-3244
  54. 54. Chen H, Liu KY, Xu LP, Liu DH, Chen YH, Zhao XS, et al. Application of real time polymerase chain reaction to the diagnosis and treatment of cytomegalovirus infection after allogeneic hematopoietic stem cell transplantation. Zhonghua Xue Ye Xue Za Zhi. 2009;30:77-81
  55. 55. Storek J, Dawson MA, Storer B, Stevens-Ayers T, Maloney DG, Marr KA, et al. Immune reconstitution after allogeneic marrow transplantation compared with blood stem cell transplantation. Blood. 2001;97(11):3380-3389. DOI: 10.1182/blood.v97.11.3380
  56. 56. Gallez-Hawkins GM, Franck AE, Li X, Thao L, Oki A, Gendzekhadze K, et al. Expression of activating KIR2DS2 and KIR2DS4 genes after hematopoietic cell transplantation: Relevance to cytomegalovirus infection. Biology of Blood and Marrow Transplantation. 2011;17:1662-1672. DOI: 10.1016/j.bbmt.2011.04.008
  57. 57. De Angelis C, Mancusi A, Ruggeri L, Capanni M, Urbani E, Velardi A, et al. Expansion of CD56-negative, CD16-positive, KIR-expressing natural killer cells after T cell-depleted haploidentical hematopoietic stem cell transplantation. Acta Haematologica. 2011;126:13-20. DOI: 10.1159/000323661
  58. 58. Hokland M, Jacobsen N, Ellegaard J, Hokland P. Natural killer function following allogeneic bone marow transplantation. Transplantation. 1988;45:1080-1084. DOI: 10.1097/00007890-198806000-00016
  59. 59. Pical-Izard C, Crocchiolo R, Granjeaud S, Kochbati E, Just-Landi S, Chabannon C, et al. Reconstitution of natural killer cells in HLA-matched HSCT after reduced-intensity conditioning: Impact on clinical outcome. Biology of Blood and Marrow Transplantation. 2015;21:429-439. DOI: 10.1016/j.bbmt.2014.11.681
  60. 60. Russo A, Oliveira G, Berglund S, Greco R, Gambacorta V, Cieri N, et al. NK cell recovery after Haploidentical HSCT with post-TransplantCyclophosphamide: Dynamics and clinical implications. Blood. 2018;131:247-262. DOI: 10.1182/blood-2017-05-780668
  61. 61. Shlomchik WD. Antigen presentation in graft-vs-host disease. Experimental Hematology. 2003;31:1187-1197
  62. 62. Hashimoto D, Merad M. Harnessing dendritic cells to improve allogeneic hematopoietic cell transplantation outcome. Seminars in Immunology. 2011;23:50-57. DOI: 10.1016/j.smim.2011.01.005
  63. 63. Koehl U, Bochennek K, Zimmermann SY, Lehrnbecher T, Sörensen J, Esser R, et al. Immune recovery in children undergoing allogeneic stem cell transplantation: Absolute CD8+CD3+ count reconstitution is associated with survival. Bone Marrow Transplantation. 2007;39(5):269-278. DOI: 10.1038/sj.bmt.1705584
  64. 64. Booth C, Lawson S, Veys P. The current role of T cell depletion in paediatric stem cell transplantation. British Journal of Haematology. 2013;162:177-190. DOI: 10.1111/bjh.12400
  65. 65. Eyrich M, Leiler C, Lang P, Schilbach K, Schumm M, Bader P, et al. A prospective comparison of immune reconstitution in pediatric recipients of positively selected CD34þ peripheral blood stem cells from unrelated donors vs recipients of unmanipulated bone marrow from related donors. Bone Marrow Transplantation. 2003;32:379-390. DOI: 10.1038/sj.bmt.1704158
  66. 66. de Koning C, Plantinga M, Besseling P, Boelens JJ, Nierkens S. Immune reconstitution after allogeneic hematopoietic cell transplantation in children. Biology of Blood and Marrow Transplantation. 2016;22:195-206. DOI: 10.1016/j.bbmt.2015.08.028
  67. 67. Hakim FT, Memon SA, Cepeda R, Jones EC, Chow CK, Kasten-Sportes C, et al. Age-dependent incidence, time course, and consequences of thymic renewal in adults. The Journal of Clinical Investigation. 2005;115:930-939. DOI: 10.1172/JCI22492
  68. 68. Rodewald HR. The thymus in the age of retirement. Nature. 1998;396:630-631. DOI: 10.1038/25251
  69. 69. Storek J, Gooley T, Witherspoon RP, Sullivan KM, Storb R. Infectious morbidity in long-term survivors of allogeneic marrow transplantation is associated with low CD4 T cell counts. American Journal of Hematology. 1997;54:131-138
  70. 70. Roux E, Dumont-Girard F, Starobinski M, Siegrist CA, Helg C, Chapuis B, et al. Recovery of immune reactivity after T-cell-depleted bone marrow transplantation depends on thymic activity. Blood. 2000;96:2299-2303
  71. 71. Storek J, Joseph A, Espino G, Dawson MA, Douek DC, Sullivan KM, et al. Immunity of patients surviving 20 to 30 years after allogeneic or syngeneic bone marrow transplantation. Blood. 2001;98:3505-3512. DOI: 10.1182/blood.v98.13.3505
  72. 72. Le RQ , Melenhorst JJ, Battiwalla M, Hill B, Memon S, Savani BN, et al. Evolution of the donor T-cell repertoire in recipients in the second decade after allogeneic stem cell transplantation. Blood. 2011;117:5250-5256. DOI: 10.1182/blood-2011-01-329706
  73. 73. Fu YW, Wu DP, Cen JN, Feng YF, Chang WR, Zhu ZL, et al. Patterns of T-cell reconstitution by assessment of T-cell receptor excision circle and T-cell receptor clonal repertoire after allogeneic hematopoietic stem cell transplantation in leukemia patients – A study in Chinese patients. European Journal of Haematology. 2007;79(2):138-145. DOI: 10.1111/j.1600-0609.2007.00885.x
  74. 74. Azevedo RI, Soares MV, Albuquerque AS, Tendeiro R, Soares RS, Martins M, et al. Long-term immune reconstitution of naive and memory T cell pools after haploidentical hematopoietic stem cell transplantation. Biology of Blood and Marrow Transplantation. 2013;19(5):703-712. DOI: 10.1016/j.bbmt.2013.01.017
  75. 75. Atkinson K, Hansen JA, Storb R, Goehle S, Goldstein G, Thomas ED. T-cell subpopulations identified by monoclonal antibodies after human marrow transplantation. I. Helper-inducer and cytotoxic-suppressor subsets. Blood. 1982;59(6):1292-1298
  76. 76. Marshall NA, Howe JG, Formica R, Krause D, Wagner JE, Berliner N, et al. Rapid reconstitution of Epstein-Barr virus-specific T lymphocytes following allogeneic stem cell transplantation. Blood. 2000;96(8):2814-2821
  77. 77. Weinberg K, Blazar BR, Wagner JE, Agura E, Hill BJ, Smogorzewska M, et al. Factors affecting thymic function after allogeneic hematopoietic stem cell transplantation. Blood. 2001;97(5):1458e1466. DOI: 10.1182/blood.v97.5.1458
  78. 78. Storek J, Zhao Z, Lin E, Berger T, McSweeney PA, Nash RA, et al. Recovery from and consequences of severe iatrogenic lymphopenia (induced to treat autoimmune diseases). Clinical Immunology. 2004;113(3):285-298. DOI: 10.1016/j.clim.2004.07.006
  79. 79. Storek J, Witherspoon RP, Storb R. T cell reconstitution after bone marrow transplantation into adult patients does not resemble T cell development in early life. Bone Marrow Transplantation. 1995;16(3):413-425
  80. 80. Weinberg K, Annett G, Kashyap A, Lenarsky C, Forman SJ, Parkman R. The effect of thymic function on immunocompetence following bone marrow transplantation. Biology of Blood and Marrow Transplantation. 1995;1(1):18-23
  81. 81. Storek J, Joseph A, Dawson MA, Douek DC, Storer B, Maloney DG. Factors influencing T-lymphopoiesis after allogeneic hematopoietic cell transplantation. Transplantation. 2002;73(7):1154-1158. DOI: 10.1097/00007890-200204150-00026
  82. 82. Roord ST, de Jager W, Boon L, Wulffraat N, Martens A, Prakken B, et al. Autologous bone marrow transplantation in autoimmune arthritis restores immune homeostasis through CD4þCD25þFoxp3þ regulatory T cells. Blood. 2008;111:5233-5241. DOI: 10.1182/blood-2007-12-128488
  83. 83. Zhang L, Bertucci AM, Ramsey-Goldman R, Burt RK, Datta SK. Regulatory T cell (Treg) subsets return in patients with refractory lupus following stem cell transplantation, and TGF-beta-producing CD8þ Treg cells are associated with immunological remission of lupus. Journal of Immunology. 2009;183:6346-6358. DOI: 10.4049/jimmunol.0901773
  84. 84. Federmann B, Bornhauser M, Meisner C, Kordelas L, Beelen DW, Stuhler G, et al. Haploidentical allogeneic hematopoietic cell transplantation In adults using CD3/CD19 depletion and reduced intensity conditioning: A phase II study. Haematologica. 2012;97(10):1523-1531. DOI: 10.3324/haematol.2011.059378
  85. 85. Kook H, Goldman F, Padley D, Giller R, Rumelhart S, Holida M, et al. Reconstruction of the immune system after unrelated or partially matched T-cell-depleted bone marrow transplantation in children: Immunophenotypic analysis and factors affecting the speed of recovery. Blood. 1996;88(3):1089-1097
  86. 86. Storek J, Ferrara S, Ku N, Giorgi JV, Champlin RE, Saxon A. B cell reconstitution after human bone marrow transplantation: Recapitulation of ontogeny? Bone Marrow Transplantation. 1993;12(4):387e398
  87. 87. Oshrine BR, Li Y, Teachey DT, Heimall J, Barrett DM, Bunin N. Immunologic recovery in children after alternative donor allogeneic transplantation for hematologic malignancies: Comparison of recipients of partially T cell-depleted peripheral blood stem cells and umbilical cord blood. Biology of Blood and Marrow Transplantation. 2013;19:1581-1589. DOI: 10.1016/j.bbmt.2013.08.003
  88. 88. Moretta A, Maccario R, Fagioli F, Giraldi E, Busca A, Montagna D, et al. Analysis of immune reconstitution in children undergoing cord blood transplantation. Experimental Hematology. 2001;29:371-379
  89. 89. Olkinuora H, von Willebrand E, Kantele JM, Vainio O, Talvensaari K, Saarinen-Pihkala U, et al. The impact of early viral infections and graft-versus-host disease on immune reconstitution following paediatric stem cell transplantation. Scandinavian Journal of Immunology. 2011;73:586-593. DOI: 10.1111/j.1365-3083.2011.02530.x
  90. 90. Chiesa R, Gilmour K, Qasim W, Adams S, Worth AJ, Zhan H, et al. Omission of in vivo T cell depletion promotes rapid expansion of naïve CD4þ cord blood lymphocytes and restores adaptive immunity within 2 months after unrelated cord blood transplant. British Journal of Haematology. 2012;156:656-666. DOI: 10.1111/j.1365-2141.2011.08994.x
  91. 91. Dudakov JA, Perales MA, van den Brink MRM. Immune reconstitution following hematopoietic cell transplantation. In: Thomas’ Hematopoietic Cell Transplantation. 5th ed. Wiley & Sons Publishers; 2015. pp. 160-169. DOI: 10.1002/9781118416426.ch15
  92. 92. Porrata LF, Markovic SN. Timely reconstitution of immune competence affects clinical outcome following autologous stem cell transplantation. Clinical and Experimental Medicine. 2004;4:78-85
  93. 93. Kim DH, Kim JG, Sohn SK, Sung WJ, Suh JS, Lee KS, et al. Clinical impact of early absolute lymphocyte count after allogeneic stem cell transplantation. British Journal of Haematology. 2004;125:217-224. DOI: 10.1111/j.1365-2141.2004.04891.x
  94. 94. Döring M, Cabanillas Stanchi KM, Haufe S, Erbacher A, Bader P, Handgretinger R, et al. Patterns of monocyte subpopulations and their surface expression of HLA-DR during adverse events after hematopoietic stem cell transplantation. Annals of Hematology. 2014;94:825-836. DOI: 10.1007/s00277-014-2287-6
  95. 95. Kim DH, Sohn SK, Won DI, Lee NY, Suh JS, Lee KB. Rapid helper T-cell recovery above 200 × 106/l at 3 months correlates to successful transplant outcomes after allogeneic stem cell transplantation. Bone Marrow Transplantation. 2006;37:1119-1128. DOI: 10.1038/sj.bmt.1705381
  96. 96. Berger M, Figari O, Bruno B, Raiola A, Dominietto A, Fiorone M, et al. Lymphocyte subsets recovery following allogeneic bone marrow transplantation (BMT): CD4+ cell count and transplant-related mortality. Bone Marrow Transplantation. 2008;41:55-62. DOI: 10.1038/sj.bmt.1705870
  97. 97. Admiraal R, van Kesteren C, Jol-van der Zijde CM, Lankester AC, Bierings MB, Egberts TC, van Tol MJ, Knibbe CA, Bredius RG, Boelens JJ. Association between anti-thymocyte globulin (ATG) exposure and CD4þ immune reconstitution predicting overall survival in paediatric haematopoietic cell transplantation: A multicentre retrospective pharmacodynamic cohort analysis. The Lancet Haematology 2015;2: e194-e203. DOI: 10.1016/S2352-3026(15)00045-9
  98. 98. Kanda J, Chiou LW, Szabolcs P, Sempowski GD, Rizzieri DA, Long GD, et al. Immune recovery in adult patients after myeloablative dual umbilical cord blood, matched sibling, and matched unrelated donor hematopoietic cell transplantation. Biology of Blood and Marrow Transplantation. 2012;18:1664-1676.e1. DOI: 10.1016/j.bbmt.2012.06.005
  99. 99. Bartelink IH, Belitser SV, Knibbe CA, Danhof M, de Pagter AJ, Egberts TC, et al. Immune reconstitution kinetics as an early predictor for mortality using various hematopoietic stem cell sources in children. Biology of Blood and Marrow Transplantation. 2013;19:305-313. DOI: 10.1016/j.bbmt.2012.10.010
  100. 100. Thomson BG, Roberston KA, Gowan D, Heilman D, Broxmeyer HE, Emanuel D, et al. Analysis of engraftment, graft-versus-host disease, and immune recovery following unrelated donor cord blood transplantation. Blood. 2000;96:2703-2711
  101. 101. Campbell JJ, Murphy KE, Kunkel EJ, Brightling CE, Soler D, Shen Z, et al. CCR7 expression and memory T cell diversity in humans. Journal of Immunology. 2001;166:877-884. DOI: 10.4049/jimmunol.166.2.877
  102. 102. Hamann D, Baars PA, Rep MH, Hooibrink B, Kerkhof-Garde SR, Klein MR, et al. Phenotypic and functional separation of memory and effector human CD8+ T cells. The Journal of Experimental Medicine. 1997;186:1407-1418. DOI: 10.1084/jem.186.9.1407
  103. 103. Resende RG, de Correia-Silva J, Silva TA, Salomão UE, Marques-Silva L, Vieira ÉL, et al. IL-17 genetic and immunophenotypic evaluation in chronic graft-versus-host disease. Mediators of Inflammation. 2014;2014:571231. DOI: 10.1155/2014/571231
  104. 104. Min CK, Lee WY, Min DJ, Lee DG, Kim YJ, Park YH, et al. The kinetics of circulating cytokines including IL-6, TNF-, IL-8 and IL-10 following allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplantation. 2001;28:935-940. DOI: 10.1038/sj.bmt.1703258
  105. 105. Kang J, Der SD. Cytokine functions in the formative stages of a lymphocyte’s life. Current Opinion in Immunology. 2004;16:180-190. DOI: 10.1016/j.coi.2004.02.002
  106. 106. Kim JM, Rudensky A. The role of the transcription factor Foxp3 in the development of regulatory T cells. Immunological Reviews. 2006;212:86-98. DOI: 10.1111/j.0105-2896.2006.00426.x
  107. 107. Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM, et al. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nature Immunology. 2005;6:1123-1132. DOI: 10.1038/ni1254
  108. 108. Small TN, Robinson WH, Miklos DB. B cells and transplantation: An educational resource. Biology of Blood and Marrow Transplantation. 2009;15(1 Suppl):104-113. DOI: 10.1016/j.bbmt.2008.10.016
  109. 109. Rossi M, Young JW. Human dendritic cells: Potent antigen-presenting cells at the crossroads of innate and adaptive immunity. Journal of Immunology. 2005;175:1373-1381. DOI: 10.4049/jimmunol.175.3.1373
  110. 110. Douek DC, Vescio RA, Betts MR, Brenchley JM, Hill BJ, Zhang L, et al. Assessment of thymic output in adults after haematopoietic stem-cell transplantation and prediction of T-cell reconstitution. Lancet. 2000;355:1875-1881. DOI: 10.1016/S0140-6736(00)02293-5
  111. 111. Lewin SR, Heller G, Zhang L, Rodrigues E, Skulsky E, van den Brink MR, Small TN, Kernan NA, O'Reilly RJ, Ho DD, Young JW. Direct evidence for new T-cell generation by patients after either T-cell-depleted or unmodified allogeneic hematopoietic stem cell transplantations. Blood 2002; 100: 2235-2242
  112. 112. Wils EJ, van der Holt B, Broers AE, Posthumus-van Sluijs SJ, Gratama JW, Braakman E, et al. Insufficient recovery of thymopoiesis predicts for opportunistic infections in allogeneic hematopoietic stem cell transplant recipients. Haematologica. 2011;96:1846-1854. DOI: 10.3324/haematol.2011.047696
  113. 113. Kielsen K, Jordan KK, Uhlving HH, Pontoppidan PL, Shamim Z, Ifversen M, et al. T cell reconstitution in allogeneic haematopoietic stem cell transplantation: Prognostic significance of plasma interleukin-7. Scandinavian Journal of Immunology. 2015;81:72-80. DOI: 10.1111/sji.12244
  114. 114. Paczesny S, Krijanovski OI, Braun TM, Choi SW, Clouthier SG, Kuick R, et al. A biomarker panel for acute graft-versus-host disease. Blood. 2009;113:273-278. DOI: 10.1182/blood-2008-07-167098
  115. 115. Berger M, Signorino E, Muraro M, Quarello P, Biasin E, Nesi F, et al. Monitoring of TNFR1, IL-2Ra, HGF, CCL8, IL-8 and IL-12p70 following HSCT and their role as GVHD biomarkers in paediatric patients. Bone Marrow Transplantation. 2013;48:1230-1236. DOI: 10.1038/bmt.2013.41
  116. 116. Perales MA, Goldberg JD, Yuan J, Koehne G, Lechner L, Papadopoulos EB, et al. Recombinant human interleukin- 7 (CYT107) promotes T-cell recovery after allogeneic stem cell transplantation. Blood. 2012;120:4882-4891. DOI: 10.1182/blood-2012-06-437236
  117. 117. Keller T, Weber S, Gombert M, Schuster FR, Asang C, Stepensky P, et al. Next-generation-sequencing spectratyping reveals public T-cell receptor repertoires in pediatric very severe aplastic anemia and identifies a beta chain CDR3 sequence associated with hepatitis-induced pathogenesis. Haematologica. 2013;98:1388-1396. DOI: 10.3324/haematol.2012.069708
  118. 118. Michalek J, Collins RH, Hill BJ, Brenchley JM, Douek DC. Identification and monitoring of graft-versus-host specific T-cell clone in stem cell transplantation. The Lancet. 2003;361:1183-1185. DOI: 10.1016/S0140-6736(03)12917-0
  119. 119. van Heijst JW, Ceberio I, Lipuma LB, Samilo DW, Wasilewski GD, Gonzales AM, et al. Quantitative assessment of T cell repertoire recovery after hematopoietic stem cell transplantation. Nature Medicine. 2013;19:372-377. DOI: 10.1038/nm.3100
  120. 120. Sudo T, Nishikawa S, Ohno N, Akiyama N, Tamakoshi M, Yoshida H, et al. Expression and function of the interleukin 7 receptor in murine lymphocytes. Proceedings of the National Academy of Sciences of the United States of America. 1993;90:9125-9129. DOI: 10.1073/pnas.90.19.9125
  121. 121. Hennion-Tscheltzoff O, Leboeuf D, Gauthier SD, Dupuis M, Assouline B, Gregoire A, et al. TCR triggering modulates the responsiveness and homeostatic proliferation of CD4+ thymic emigrants to IL-7 therapy. Blood. 2013;121(23):4684-4693. DOI: 10.1182/blood-2012-09-458174
  122. 122. Alpdogan O, Muriglan SJ, Eng JM, Willis LM, Greenberg AS, Kappel BJ, et al. IL-7 enhances peripheral T cell reconstitution after allogeneic hematopoietic stem cell transplantation. The Journal of Clinical Investigation. 2003;112(7):1095-1107. DOI: 10.1172/JCI200317865
  123. 123. Dudakov JA, Hanash AM, Jenq RR, Young LF, Ghosh A, Singer NV, et al. Interleukin-22 drives endogenous thymic regeneration in mice. Science. 2012;336:91-95. DOI: 10.1126/science.1218004
  124. 124. Eisenring M, vom Berg J, Kristiansen G, Saller E, Becher B. IL-12 initiates tumor rejection via lymphoid tissue-inducer cells bearing the natural cytotoxicity receptor NKp46. Nature Immunology. 2010;11:1030-1038. DOI: 10.1038/ni.1947
  125. 125. Satoh-Takayama N, Lesjean-Pottier S, Vieira P, Sawa S, Eberl G, Vosshenrich CA, et al. IL-7 and IL-15 independently program the differentiation of intestinal CD3–NKp46+ cell subsets from Id2-dependent precursors. The Journal of Experimental Medicine. 2010;207:273-280. DOI: 10.1084/jem.20092029
  126. 126. Sauter CT, Bailey CP, Panis MM, Biswas CS, Budak-Alpdogan T, Durham A, et al. Interleukin-15 administration increases graft-versus-tumor activity in recipients of haploidentical hematopoietic SCT. Bone Marrow Transplantation. 2013;48(9):1237-1242. DOI: 10.1038/bmt.2013.47
  127. 127. Thiant S, Moutuou MM, Leboeuf D, Guimond M. Homeostatic cytokines in immune reconstitution and graft-versus-host disease. Cytokine. 2016;82:24-32. DOI: 10.1016/j.cyto.2016.01.003
  128. 128. Maraskovsky E, Brasel K, Teepe M, Roux ER, Lyman SD, Shortman K, et al. Dramatic increase in the numbers of functionally mature dendritic cells in Flt3 ligand-treated mice: Multiple dendritic cell subpopulations identified. The Journal of Experimental Medicine. 1996;184(5):1953-1962. DOI: 10.1084/jem.184.5.1953
  129. 129. Spaggiari GM, Capobianco A, Becchetti S, Mingari MC, Moretta L. Mesen-chymal stem cell-natural killer cell interactions: Evidence that activated NK cells are capable of killing MSCs, whereas MSCs can inhibit IL-2- induced NK-cell proliferation. Blood. 2006;107(4):1484-1490. DOI: 10.1182/ blood-2005-07-2775
  130. 130. Nemeth K, Leelahavanichkul A, Yuen PS, Mayer B, Parmelee A, Doi K, et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)- dependent reprogramming of host macrophages to increase their inter-leukin-10 production. Nature Medicine. 2009;15(1):42-49. DOI: 10.1038/nm.1905
  131. 131. Bouchlaka MN, Moffitt AB, Kim J, Kink JA, Bloom DD, Love C, et al. Human mesenchymal stem cell-educated macrophages are a distinct high IL-6- producing subset that confer protection in graft-versus-host-disease and radiation injury models. Biology of Blood and Marrow Transplantation. 2017;23(6):897-905. DOI: 10.1016/j.bbmt.2017.02.018
  132. 132. Fujii S, Miura Y, Fujishiro A, Shindo T, Shimazu Y, Hirai H, et al. Graft- versus-host disease amelioration by human bone marrow mesenchymal stromal/stem cell-derived extracellular vesicles is associated with peripheral preservation of naive T cell populations. Stem Cells. 2018;36(3):434-445. DOI: 10.1002/stem.2759
  133. 133. Laport GG, Sheehan K, Baker J, Armstrong R, Wong RM, Lowsky R, et al. Adoptive immunotherapy with cytokineinduced killer cells for patients with relapsed hematologic malignancies after allogeneic hematopoietic cell transplantation. Biology of Blood and Marrow Transplantation. 2011;17:1679-1687. DOI: 10.1016/j.bbmt.2011.05.012
  134. 134. Miller JS, Soignier Y, Panoskaltsis-Mortari A, McNearney SA, Yun GH, Fautsch SK, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood. 2005;105:3051-3057. DOI: 10.1182/blood-2004-07-2974
  135. 135. Rubnitz JE, Inaba H, Ribeiro RC, Pounds S, Rooney B, Bell T, et al. NKAML: A pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia. Journal of Clinical Oncology. 2010;28:955-959. DOI: 10.1200/JCO.2009.24.4590

Written By

Meenakshi Singh, Selma Z. D’Silva and Abhishweta Saxena

Submitted: 16 May 2019 Reviewed: 16 August 2019 Published: 03 December 2019