Umbilical cord blood (UCB) is an alternative source of hematopoietic stem cells for patients without HLA-matched adult donors. UCB contains a low number of nucleated cells and mostly naive T cells, resulting in prolonged time to engraftment and lack of transferred T-cell memory. Although the first phase of T-cell reconstitution after UCB transplantation (UCBT) depends on peripheral expansion of transferred T cells, permanent T-cell reconstitution is mediated via a central mechanism, which depends on de novo production of naive T lymphocytes by the recipient’s thymus from donor-derived lymphoid-myeloid progenitors (LMPs). Thymopoiesis can be assessed by quantification of recent thymic emigrants, T-cell receptor excision circle levels, and T-cell receptor repertoire diversity. These assays are valuable tools for monitoring posttransplantation thymic recovery, but more importantly they have shown the significant prognostic value of thymic reconstitution for clinical outcomes after UCBT, including opportunistic infections, disease relapse, and overall survival. Strategies to improve thymic entry and differentiation of LMPs and to accelerate recovery of the thymic stromal microenvironment may improve thymic lymphopoiesis. Here, we discuss the mechanisms and clinical implications of thymic recovery and new approaches to improve reconstitution of the T-cell repertoire after UCBT.

The first successful umbilical cord blood (UCB) transplantation (UCBT) was performed in 1988, using cord blood from an HLA-matched sibling in a patient with Fanconi anemia.1  Since then, multiple large-scale trials have proven the clinical utility of UCBT as an alternative source of hematopoietic stem cells (HSCs) for the treatment of children and adults who are in need for allogeneic HSC transplantation (HSCT).2-4  Compared with peripherally mobilized HSC or bone marrow (BM) HSC units from unrelated adult donors, UCB grafts have the advantage of immediate availability, absence of risk to the donors, and lower immunogenicity, which allows for a greater degree of HLA incompatibility.5  UCB contains mostly naive, antigen-inexperienced T lymphocytes which do not transfer protective T-cell memory function to the host. Additionally, UCB T cells display impaired capacity for effector cytokine production and reduced cytolytic activity.6,7  Furthermore, UCB contains high numbers of T regulatory cells (Tregs) with a more potent suppressor function compared with adult Tregs.8  Consequently, UCBT is associated with delayed and incomplete immune reconstitution due to the lack of transferred adoptive immunity and due to the delayed HSC engraftment and reconstitution of lymphopoiesis in the host thymus.9  As a result, infectious complications and viral reactivation remain the most important causes of peritransplant morbidity and mortality in UCBT recipients.

Allogeneic transplantation is followed by a period of profound lymphopenia and immunodeficiency, as a result of conditioning chemotherapy and use of immunosuppressive agents to prevent graft rejection or GVHD. The quantitative and qualitative reconstitution of the T-cell compartment is a slow process that can extend beyond the first year after HSCT and proceeds along 2 different pathways that act in parallel but follow distinct kinetics10  (Figure 1). In the early posttransplant period, the thymus-independent pathway predominates and is mediated by adoptively transferred donor T cells contained in the graft or recipient T cells that survive conditioning. These transferred T lymphocytes undergo homeostatic expansion in response to lymphopenia and high cytokine levels, which characterize the early posttransplant period, or oligoclonal proliferation upon interaction with cognate antigens. In contrast to peripherally mobilized HSC or BM grafts from adult donors, which are characterized by oligoclonal T-cell receptor (TCR) profiles and contain a considerable number of memory T cells, UCB grafts contain uniformly antigen-inexperienced T cells, which display a complete TCR repertoire at birth.11  The lymphopenia-driven homeostatic expansion of UCB T cells after transplantation leads to gradual loss of the naive phenotype and transition to an effector or memory-like phenotype.12,13  In addition, the antigen-driven peripheral T-cell expansion results in the contraction of T-cell repertoire diversity. These changes in the composition of the T-cell pool early after UCBT lead to an increasingly contracted and skewed T-cell repertoire that cannot sustain immune protection against a broad spectrum of antigens.

Figure 1

Reconstitution of the T-cell compartment after UCBT. Conditioning chemoradiation prior to UCBT results in profound lymphopenia and immunodeficiency of the host. T-cell reconstitution after UCBT is achieved by 2 independent mechanisms: the thymus-independent pathway of T-cell reconstitution predominates in the early posttransplant period and is mediated by adoptively transferred UCB T cells, which are uniformly naive and do not transfer protective immune memory, or recipient T cells that survive conditioning. These T-cell populations undergo peripheral expansion in response to lymphopenia and high cytokine levels (IL-7, IL-15, etc), or oligoclonal proliferation upon interaction with cognate antigen. Overtime, this early peripheral T-cell expansion results in late effector memory T-cell skewing and contraction of the T-cell repertoire diversity, and is associated with impaired immunologic responses to antigens. Reconstitution of a functionally competent T-cell compartment with broad antigenic specificity eventually requires the de novo production of naive T cells by the thymus of the UCBT recipient. This thymus-dependent pathway of T-cell reconstitution is a prolonged multistep process. LMPs contained in the UCB graft or arising from the engrafted donor-derived HSCs migrate via circulation and repopulate the thymus with thymocyte precursors that can reconstitute thymopoiesis. The thymus provides the essential microenvironment (stroma) that supports T-cell proliferation, selection, and differentiation into RTEs. Several factors can delay the recovery of thymopoiesis after UCBT, including low number of LMPs (as a result of low nucleated cell dose of UCB and delayed engraftment), advanced recipient age with resultant thymic involution and thymic damage from the conditioning chemoradiation or GVHD. Although slow, the thymus-dependent mechanism is imperative for the renewal of peripheral T-cell pool and constant export of new naive T cells with broad TCR repertoire diversity, capable of responding to a great spectrum of antigens.

Figure 1

Reconstitution of the T-cell compartment after UCBT. Conditioning chemoradiation prior to UCBT results in profound lymphopenia and immunodeficiency of the host. T-cell reconstitution after UCBT is achieved by 2 independent mechanisms: the thymus-independent pathway of T-cell reconstitution predominates in the early posttransplant period and is mediated by adoptively transferred UCB T cells, which are uniformly naive and do not transfer protective immune memory, or recipient T cells that survive conditioning. These T-cell populations undergo peripheral expansion in response to lymphopenia and high cytokine levels (IL-7, IL-15, etc), or oligoclonal proliferation upon interaction with cognate antigen. Overtime, this early peripheral T-cell expansion results in late effector memory T-cell skewing and contraction of the T-cell repertoire diversity, and is associated with impaired immunologic responses to antigens. Reconstitution of a functionally competent T-cell compartment with broad antigenic specificity eventually requires the de novo production of naive T cells by the thymus of the UCBT recipient. This thymus-dependent pathway of T-cell reconstitution is a prolonged multistep process. LMPs contained in the UCB graft or arising from the engrafted donor-derived HSCs migrate via circulation and repopulate the thymus with thymocyte precursors that can reconstitute thymopoiesis. The thymus provides the essential microenvironment (stroma) that supports T-cell proliferation, selection, and differentiation into RTEs. Several factors can delay the recovery of thymopoiesis after UCBT, including low number of LMPs (as a result of low nucleated cell dose of UCB and delayed engraftment), advanced recipient age with resultant thymic involution and thymic damage from the conditioning chemoradiation or GVHD. Although slow, the thymus-dependent mechanism is imperative for the renewal of peripheral T-cell pool and constant export of new naive T cells with broad TCR repertoire diversity, capable of responding to a great spectrum of antigens.

Close modal

Reconstitution of a functionally competent T-cell compartment with broad antigenic specificity depends on the de novo production of naive T cells by the thymus of the HSCT recipient.14  This thymus-dependent pathway is a prolonged process, which begins with the migration of early lymphoid-myeloid progenitors (LMPs), present in the graft or arising from donor-derived HSCs after engraftment, which circulate in the periphery and seed the recipient’s thymus. LMPs initially settle in the cortex where they undergo expansion and T-cell lineage commitment under the influence of Notch signaling.15  Furthermore, the thymus provides the essential microenvironment (stroma) that supports T-cell proliferation, differentiation, and selection via cytokines (interleukin-7 [IL-7], stem cell factor [SCF], keratinocyte growth factor [KGF], C-C motif chemokine ligand 25 [CCL25]) and surface proteins (chemokine receptors, peptide/major histocompatibility complex [MHC] complexes), which facilitate trafficking and cell-cell interactions.16  Thymocytes undergo sequential rearrangements of the loci encoding for the β- and α-chains of TCR, which result in the assembly of the αβ-TCR and are subject to positive selection. Positively selected thymocytes undergo differentiation into CD4 and CD8 single-positive (SP) thymocytes, according to the restriction of their TCR to recognize either MHC class II or I molecules, respectively. Finally, SP cells migrate to the medulla and undergo negative selection, a process that assures the elimination of self-reactive thymocytes. Only the small percentage of thymocytes that have successfully undergone positive and negative selection exit the thymus and are termed recent thymic emigrants (RTEs).

Regardless of the graft source, the efficiency of thymopoiesis after HSCT depends on several factors, including recipient age, intensity of the conditioning regimen, and graft-versus-host disease (GVHD). In UCBT, 2 additional factors are of particular importance. First, the low number of hematopoietic progenitor cells in the UCB graft results in delayed engraftment and emergence of LMPs that migrate to the thymus and reinitiate thymopoiesis. Second, the greater degree of MHC discordance between donor and host in UCBT compared with HSCT from adult donors, might adversely affect intrathymic T-cell selection, which occurs in an MHC-restricted manner. Specifically, during positive selection, cortical thymic epithelial cells (cTECs) expressing self-MHC mediate presentation of self-peptides and select TCRs with intermediate affinity for such antigens. Subsequently, T cells surviving positive selection are challenged during negative selection in the thymic medulla for reactivity with self-peptides presented by medullary epithelial cells (mTECs) or by BM-derived dendritic cells (DCs). Although after allogeneic HSCT, cTECs and mTECs remain of host origin, thymic DCs are of donor origin. Consequently, it has been proposed that the degree of MHC incompatibility between donor and host can adversely impact thymic-dependent T-cell reconstitution, resulting in altered peripheral T-cell repertoire due to perturbation of the physiologic mechanisms of the positive selection process in the thymus.17  This hypothesis has been supported by clinical findings showing that TCR repertoire diversity is only mildly skewed in recipients of BM from HLA-identical related donors, whereas recipients of BM from HLA-mismatched related donors or matched unrelated donors (MUDs) display a markedly skewed TCR repertoire during the first year after transplantation.14,18,19  In that regard, because the lower alloreactivity of UCB grafts allows for a greater degree of MHC discordance between donor and host, the markedly skewed TCR repertoire diversity in UCBT recipients during the first year after UCBT, might be a consequence of a perturbed thymic selection process due to the greater degree of MHC discordance between UCB and host.

Maintenance of a broad TCR repertoire depends on the constant export of naive T cells from the thymus, which persists throughout adult life.20  In the past, radiographic imaging had been used to monitor the size of the thymus as a surrogate marker of thymopoiesis. However, size alone does not reliably reflect the actual functional capacity of the organ. Consequently, volumetric studies have considerable limitations in the evaluation of thymic recovery. Identification of thymus-derived naive T cells by immunophenotyping provides a more accurate assessment of thymic activity and RTE export. The expression of CD45 isotopes has been most widely used for the enumeration of the naive (CD45RA+) T cells, although not always reliably.21  The use of additional surface markers, including CCR7, CD62L, CD31, CD27, and αEβ7 integrin (CD103), in conjunction with the CD45RA, has improved discrimination between naive and memory T cells.22,23  However, similarly to CD45RA, none of these surface markers are specific for RTEs. Therefore, although highly helpful for the assessment of T-cell reconstitution, immunophenotyping also has some limitations in the assessment of thymopoiesis.

A more accurate quantification of thymic RTE output is provided by the measurement of T-cell receptor excision circles (TRECs).24  These are circular molecules of extrachromosomal DNA and represent by-products of the TCR recombination events that take place during intrathymic T-cell development25  (Figure 2). Measurement of signal joint TRECs (sjTRECs) with quantitative competitive (QC)–polymerase chain reaction (PCR) has been extensively used for the assessment of thymopoiesis and RTE output.24,25  Although useful in monitoring thymopoietic recovery after HSCT, the sjTREC assay may be influenced by peripheral T-cell proliferation because TRECs do not replicate with mitosis and are progressively diluted out with each division cycle. Thus, the dependency of TREC levels on the proliferative history of the peripheral T-cell pool is a limiting factor for comparisons. Measuring TREC levels per volume of blood rather than per nanogram of DNA can attenuate this effect. Similarly, the determination of the sj/β TREC ratio (termed thymic factor [TF]) has been introduced as a more accurate method for the assessment of thymopoiesis.26,27  The sj/β TREC ratio reflects the intensity of the intrathymic proliferation between the double negative (DN) and double positive (DP) stages, which is the major determinant of thymic RTE output. Because both sj and β TREC levels are equally diluted with subsequent cell divisions, their ratio is independent of peripheral cell proliferation and it is considered as an “RTE signature” of the peripheral T-cell compartment.

Figure 2

Molecular generation of TRECs during thymic differentiation of T cells. DN thymocytes first undergo rearrangement of the TCRB locus (encoding segments of the TCR-β chain). This begins with the rearrangement of the TCRBD to TCRBJ, which gives rise to several DβJβTRECs, and is followed by the recombination of V to DJ segments, which generates a greater variety of VDβTRECs (A). The TCRA locus is rearranged next, which, similarly to the β-chain, is characterized by enormous diversity. However, a common requirement for productive TCRAVJ recombination is deletion of the TCRD locus that it encompasses. This 2-step process gives rise to a signal joint TREC and a coding joint TREC (B). Both DNA families of TRECs are stable and do not replicate during mitosis. These sequences are unique to naive αβ T cells. As a result, TRECs serve as a valuable marker of RTEs and their levels are indicative of thymic activity.

Figure 2

Molecular generation of TRECs during thymic differentiation of T cells. DN thymocytes first undergo rearrangement of the TCRB locus (encoding segments of the TCR-β chain). This begins with the rearrangement of the TCRBD to TCRBJ, which gives rise to several DβJβTRECs, and is followed by the recombination of V to DJ segments, which generates a greater variety of VDβTRECs (A). The TCRA locus is rearranged next, which, similarly to the β-chain, is characterized by enormous diversity. However, a common requirement for productive TCRAVJ recombination is deletion of the TCRD locus that it encompasses. This 2-step process gives rise to a signal joint TREC and a coding joint TREC (B). Both DNA families of TRECs are stable and do not replicate during mitosis. These sequences are unique to naive αβ T cells. As a result, TRECs serve as a valuable marker of RTEs and their levels are indicative of thymic activity.

Close modal

The diversity of T-cell repertoire, as assessed by CDR3 spectratyping or sequencing of the TCR β-chain, can also provide indirect information about thymic activity. T-cell diversity is almost exclusively accounted for by the naive population of lymphocytes. Consequently, TCR repertoire diversity after HSCT reflects the extent of naive T-cell production by the thymus and correlates with TREC levels.28 

Outcomes of single- and double-unit UCBT

The assessment of thymic function by measuring RTEs, TRECs, or TCR repertoire diversity has provided valuable insight into the kinetics and factors that affect thymic reconstitution after UCBT (Table 1). Studies of lymphocyte reconstitution in pediatric UCBT recipients have shown that despite a transient delay in the first few months, long-term lymphocyte recovery after single-unit UCBT (sUCBT) is comparable to matched sibling or unrelated HSCT recipients.29-31  These observations suggest a relative deficiency of the thymus-independent pathway of T-cell reconstitution after UCBT, which might be related to the low number and biologic properties of UCB T cells, but efficient support of T-cell recovery via the thymus-dependent pathway within the first year after UCBT. In such pediatric UCBT recipients, sjTREC and βTREC values reach a nadir at 3 months but recover to near pre-UCBT levels at 6 months after transplantation.32  By 1 year, recipients attain normal levels of sjTRECs,33  with a concomitant increase in naive CD4+ T-cell counts and TCR repertoire diversity.34,35  No significant differences in the timeline of thymic recovery have been observed compared with haploidentical or matched sibling HSCT recipients.32,35  In fact, at 2 years after transplantation, TREC values and TCR diversity were higher in UCBT recipients than in recipients of matched sibling donor grafts.35  These observations raise the intriguing hypothesis that LMPs of the UCB might have a superior potential to reconstitute thymopoiesis and TCR diversity compared with other HSC graft sources.

Table 1

Published studies reporting findings on thymic reconstitution after UCBT

ReferencePatients (Median age, y)Conditioning and no. of UCB unitsTRECs
32  UCBT: 24 pediatric patients (7.7) MAC, no ATG, 1 UCB unit No significant differences between the 2 groups. sjTRECs and βTRECs dropped at 3 mo and returned to ∼ pretransplant levels at 6 mo 
Haplo HSCT: 33 pediatric patients (4.7) 
33  10 adult patients (29.3) Variable, ATG, 1 UCB unit 7 of 10 adult patients had detectable sjTRECs at 18 mo after UCBT, with median 325 copies per μg DNA at a median of 36 mo. All pediatric patients had detectable sjTRECs at 12 mo (earliest time point tested), median 6741 copies per μg DNA at median of 30 mo 
8 pediatric patients (4.6) 
34  30 pediatric patients (1) No ATG, 1 UCB unit TRECs detectable at 3 mo and substantially increased at 6-12 mo 
35  29 patients: Variable ATG, 1 UCB unit Normal levels at 1 y; comparable between UCBT and BMT groups. At 2 y, UCBT recipients had statistically higher TRECs than BMT recipients and young adults 
UCBT: 10 patients (12.6) 
MSD BMT: 19 patients (15) 
13  32 patients (33.5) Variable, ATG, 1 UCB unit 6 of 21 patients had detectable sjTRECs at baseline; 24 of 26 had undetectable sjTRECs during first year after UCBT 
40  dUCBT: 29 patients (36) MAC without ATG, 2 UCB units TRECs lower in dUCBT recipients at 3 mo vs MRD/MUD group, but comparable in the 2 groups by 6 mo. Levels remained lower than normal in both groups at 12 mo 
MSD BMT: 33 patients (45) 
Matched unrelated: 33 patients (41) 
41  27 patients (48) RIC, ATG, 2 UCB units All evaluable patients had detectable sjTRECs at baseline; 3 of 10 at day 100; 10 of 15 at 6 mo (median 117 copies per μg DNA); 14 of 15 at 1 y (median 2136 copies per μg DNA) 
42  13 patients (42) MAC without ATG (9 patients) and RIC + ATG (4 patients), + post-UCBT PTH, 2 UCB units All patients had detectable sjTRECs at baseline; 3 of 7 at day 100; 4 of 6 at 6 mo (median 442 copies per μg DNA); all patients alive at 1 and 2 y had normal (>2000 copies per μg DNA) levels 
ReferencePatients (Median age, y)Conditioning and no. of UCB unitsTRECs
32  UCBT: 24 pediatric patients (7.7) MAC, no ATG, 1 UCB unit No significant differences between the 2 groups. sjTRECs and βTRECs dropped at 3 mo and returned to ∼ pretransplant levels at 6 mo 
Haplo HSCT: 33 pediatric patients (4.7) 
33  10 adult patients (29.3) Variable, ATG, 1 UCB unit 7 of 10 adult patients had detectable sjTRECs at 18 mo after UCBT, with median 325 copies per μg DNA at a median of 36 mo. All pediatric patients had detectable sjTRECs at 12 mo (earliest time point tested), median 6741 copies per μg DNA at median of 30 mo 
8 pediatric patients (4.6) 
34  30 pediatric patients (1) No ATG, 1 UCB unit TRECs detectable at 3 mo and substantially increased at 6-12 mo 
35  29 patients: Variable ATG, 1 UCB unit Normal levels at 1 y; comparable between UCBT and BMT groups. At 2 y, UCBT recipients had statistically higher TRECs than BMT recipients and young adults 
UCBT: 10 patients (12.6) 
MSD BMT: 19 patients (15) 
13  32 patients (33.5) Variable, ATG, 1 UCB unit 6 of 21 patients had detectable sjTRECs at baseline; 24 of 26 had undetectable sjTRECs during first year after UCBT 
40  dUCBT: 29 patients (36) MAC without ATG, 2 UCB units TRECs lower in dUCBT recipients at 3 mo vs MRD/MUD group, but comparable in the 2 groups by 6 mo. Levels remained lower than normal in both groups at 12 mo 
MSD BMT: 33 patients (45) 
Matched unrelated: 33 patients (41) 
41  27 patients (48) RIC, ATG, 2 UCB units All evaluable patients had detectable sjTRECs at baseline; 3 of 10 at day 100; 10 of 15 at 6 mo (median 117 copies per μg DNA); 14 of 15 at 1 y (median 2136 copies per μg DNA) 
42  13 patients (42) MAC without ATG (9 patients) and RIC + ATG (4 patients), + post-UCBT PTH, 2 UCB units All patients had detectable sjTRECs at baseline; 3 of 7 at day 100; 4 of 6 at 6 mo (median 442 copies per μg DNA); all patients alive at 1 and 2 y had normal (>2000 copies per μg DNA) levels 

ATG, anti-thymocyte globulin; BMT, bone marrow transplantation; dUCBT, double-unit UCBT; haplo, haploidentical; HSCT, hematopoietic stem cell transplantation; MAC, myeloablative conditioning; MRD, matched related donor; MSD, matched sibling donor; MUD, matched unrelated donor; PTH, parathyroid hormone; RIC, reduced-intensity conditioning; sjTREC, signal joint TREC; TREC, T-cell receptor excision circle; UCB, umbilical cord blood; UCBT, UCB transplantation.

In striking contrast to these findings in pediatric patients who undergo sUCBT, a marked delay in thymic recovery has been observed after sUCBT in adults. Komanduri et al studied immune reconstitution parameters in a cohort of 32 patients undergoing sUCBT.13  These patients were heavily pretreated, had impaired thymopoiesis at baseline, and received various conditioning regimens, with the inclusion of anti-thymocyte globulin (ATG). During the first year, patients displayed near complete absence of sjTRECs, paucity of naive T cells, and late memory T-cell skewing. Another study of sUCBT in adult recipients, reported TREC values below the age-adjusted normal controls, even at 36 months after transplantation.33  Whether this marked difference in the kinetics of thymic recovery between pediatric and adult sUCBT recipients is primarily related to age or to nucleated cell dose remains unclear.

Double-unit UCBT (dUCBT) has been used to circumvent the cell dose limitation in adult patients.36,37  Compared with sUCBT, dUCBT results in earlier thymic recovery. In a prospective analysis from Dana-Farber/Harvard Cancer Center,38  CD4 counts were significantly lower in the dUCBT recipients compared with MUD transplant recipients after reduced-intensity conditioning (RIC) for 6 months, but by 12 months the CD4 T-cell recovery was similar in the dUCBT and MUD cohort and reached normal levels by 24 months. Interestingly, the recovery of naive CD4 cells (CD4+CD45RO) in the dUCBT cohort was delayed in the first 6 months, but by 24 months it reached normal levels and significantly surpassed recovery in the MUD cohort.38  In a different report,39  paucity of naive (CD45RA+CCR7+) T cells in dUCBT recipients was observed during the first 6 months, followed by a gradual increase to levels comparable to those of healthy controls by 1 year. Thymopoiesis, as assessed by TREC levels, was also impaired until 9 months after transplantation, when a substantial increase of RTEs was observed. A more detailed comparison of immune reconstitution between adult dUCBT and matched sibling donor (MSD) or MUD transplant recipients after myeloablative conditioning (MAC) without ATG has been reported by Kanda et al.40  The RTE (CD4+CD45+CD62L+) count was significantly lower in dUCBT recipients during the first 6 months after transplantation but this difference disappeared at 1 year. TREC levels were also lower in dUCBT recipients at 3 months but this difference normalized between the 2 cohorts at 6 months and TCRβ repertoire was comparable by 12 months after transplantation.

In accord with those reports and in contrast to the protracted thymic deficiency observed after sUCBT, our group has shown early detection of TREC levels in adult dUCBT recipients both after RIC41  and MAC42  regimens. In a cohort of 27 adult patients who received identical RIC regimen with ATG, median sjTREC levels at the time of transplantation were slightly below normal range in this heavily pretreated population, fell dramatically after conditioning, and remained universally low through 100 days. However, at 6 months, 10 of 15 evaluable patients had detectable thymopoiesis and median TREC values reached normal limits by 1 year after dUCBT. In our second study,42  we analyzed 13 patients who enrolled in a phase 2 trial of dUCBT; 9 patients received MAC and 4 received RIC. Thymic reconstitution compared favorably to our previous study. TREC levels were detected as early as 100 days after dUCBT in a minority of patients and 67% of patients had detectable levels at 6 months. At 1 and 2 years, all surviving patients had normal levels of TRECs. The younger age of MAC recipients, omission of ATG in the MAC arm, and higher TREC levels at the time of transplantation in this cohort may have accounted for those differences.

Recently, Memorial Sloan Kettering Cancer Center (MSKCC) used a deep-sequencing analysis approach to measure TCR diversity with high resolution in 27 patients after conventional or T-cell depleted (TCD) peripheral blood stem cell (PBSC) transplantation or dUCBT.43  Interestingly, dUCBT recipients had the highest TCR diversity of all patients. The difference was more pronounced in the CD4+ T-cell diversity between dUCBT and TCD recipients after 6 months and was associated with a greater fraction of naive CD4+ T cells in dUCBT patients.

Taken together, these studies indicate that dUCBT supports improved quantitative and qualitative thymic recovery, compared with sUCBT. Although this might be related to a cell dose effect resulting in higher LMP numbers in the context of 2 UCB units, the majority of dUCBT recipients display hematopoietic reconstitution from 1 of the 2 UCB units well before the recovery of thymic function.37,44  This observation suggests that as-yet-unidentified mechanisms, other than cell dose-related HSC engraftment, might be involved in the improved thymic reconstitution after dUCBT.

Other factors affecting thymic reconstitution after UCBT

Several factors related to the characteristics of the host and the peritransplant clinical conditions have clinically important impact on the reconstitution of thymic function after allogeneic transplantation.26  Advanced recipient age has been associated with a delay in the recovery of TRECs and naive T cells, resulting in higher risk for opportunistic infections and inferior overall survival (OS).45,46  Similarly, in the setting of sUCBT, adult sUCBT recipients have markedly delayed thymic recovery compared with pediatric patients. Although one may consider that the lower cell dose per kilogram noted in adult sUCBT recipients may account for the inferior outcomes compared with pediatric populations, a multivariable analysis showed that recipient age and nucleated cell dose were independent prognostic factors in the setting of UCBT.47  The thymus is also a sensitive target of GVHD, which affects both its lymphoid and epithelial/stromal compartments. Features of “thymic GVHD” include thymocyte depletion, changes in the number and composition of thymic epithelial cells (TECs), disappearance of the corticomedullary demarcation, and absence of Hassall bodies. The distortion of normal architecture results in defective thymopoiesis.17  From a clinical standpoint, GVHD, both acute and chronic, has been identified as an independent determinant that impacts thymic recovery after HSCT or UCBT, and is associated with reduced naive T cells and TRECs and an oligoclonal T-cell repertoire.45,47,48  Importantly, immunosuppressive medications to prevent or treat GVHD can have detrimental effects on thymopoiesis.49,50  However, one study has suggested successful recovery of TRECs in a cohort of UCB recipients who received prophylactic immunosuppression in the absence of GVHD.48  An additional factor that can affect thymic reconstitution is related to the intensity of conditioning regimen. Cytotoxic chemotherapy and radiation have a negative impact on thymic function and RTE output.51,52  Theoretically, RIC regimens may cause less damage to the thymus resulting in faster regeneration of naive T cells. As an example, a multivariate analysis showed that the intensity of conditioning was the most important factor influencing TREC and CD4+ naive T-cell counts in the first 6 months after HSCT.53  In the same regard, Chao et al54  reported that patients undergoing sUCBT after RIC developed faster recovery of naive CD45RA+ T cells and TREC levels and more diverse T-cell repertoire compared with patients receiving sUCBT after MAC using identical ATG dose, GVHD prophylaxis, and supportive care.33  In another UCBT study, RIC was an independent factor for TREC recovery after UCBT in a multivariate model.47  Other factors, such as use of ATG,46  radiation, and pretransplant host TREC levels55  have also been implicated in the thymic recovery after transplantation. It is of note that due to the variable effect of the above factors, conclusions are not always consistent among different studies.

UCBT is associated with increased rates of opportunistic infections compared with other graft sources, especially from viral pathogens that require intact T-cell immunity such as herpesviruses, adenovirus, and BK virus.12,56-60  Cytomegalovirus (CMV) is the most frequent opportunistic pathogen thought to contribute significantly to HSCT morbidity and mortality.61  The CMV source after UCBT is almost exclusively of host origin because the incidence of congenital CMV infection is very low.62  The frequency of CMV reactivation after UCBT is variable between studies, depending on patient characteristics and pretransplant seropositivity status. In contrast, the risk of CMV infection in UCB recipients is not associated with donor serology, which reflects the maternal exposure history rather than active or latent infection. However, UCB lacks CMV-specific memory cells that would confer adoptive immunoprotection against CMV, and this might have significant implications in the clinical outcome of CMV reactivation in UCBT recipients. In the largest published series of 332 UCBT recipients from the University of Minnesota,59  CMV reactivation was observed in 51% of seropositive subjects, which is comparable to the rates observed in recipients of adult HSCT. However, a relatively high 27.1% of patients experiencing reactivation developed clinical CMV disease, resulting in higher TRM and reduced OS. In another report of 330 pediatric UCBT recipients from Duke University,12  CMV was the second most common cause of infectious-related deaths in the first 6 months after transplantation. Another study from Japan reported CMV antigenemia in 79% of adult UCBT recipients, who were also more likely to require repeated courses of preemptive gancyclovir therapy compared with recipients of HSCT from adult donors.63  These findings suggest a delayed recovery of CMV-specific immunity after UCBT.

To determine the immunological mechanisms that lead to the restoration of functional CMV-specific immune responses, our group has examined parameters of immune reconstitution in the context of CMV reactivation in a cohort of 27 adult dUCBT recipients. CMV-specific effectors were detected by interferon-γ (IFN-γ) enzyme-linked immunospot assay as early as 8 weeks posttransplant, before the recovery of thymopoiesis, as evidenced by undetectable TREC levels at this early time point.41  Similarly, McGoldrick et al were able to detect IFN-γ+ CMV-specific CD4+ and CD8+ T cells of UCB origin after in vitro stimulation in the majority of seropositive patients in the first 56 days after transplantation.64  These findings suggest that both CD8+ and CD4+ UCB-derived naive T cells are primed to CMV early after UCBT and can give rise to CMV effectors, independently of thymic recovery. However, the UCB-derived CD8+ CMV-specific T cells remain at low numbers and fail to control CMV reactivation in the early posttransplant period.64  The functional deficiency of CMV-specific CD8+ T cells may be explained by the profound paucity of CD4+ T helper cells after UCBT,41,64  which are imperative for the development of a functional CD8+ T-cell response.65  This conclusion is further supported by the fact that in our study, clearance of CMV viremia was increasingly observed after 6 months and significantly correlated with the recovery of naive CD4+CD45RA+ T cells.41  Furthermore, clearance of CMV viremia was associated with the reemergence of TRECs, and UCBT recipients that attained normal TREC levels were more likely to display absence of CMV viremia, suggesting a critical contribution of the recovering thymopoiesis in the clinical control of the virus in vivo.41 

The prognostic value of thymic recovery extends beyond its influence on reconstitution of pathogen-specific immunity. The COBLT study group has examined the clinical effect of the development of antigen-specific T-lymphocyte immunity against herpesviruses (herpes simplex virus [HSV], CMV, Epstein-Barr virus [EBV]) on outcomes in a cohort of 117 pediatric patients with acute myeloid leukemia (AML) or acute lymphocytic leukemia (ALL) who underwent UCBT.66  Recipients with a positive proliferative response against any herpesvirus in the first 3 years had lower infectious-related mortality67  but, more importantly, a markedly lower risk of leukemia relapse.66  Furthermore, recovery of thymic function has been correlated with decreased risk for leukemia relapse: in a combined analysis of 46 pediatric patients undergoing UCBT or haplo-HSCT, subjects who relapsed had significantly lower levels of sjTRECs or βTRECs before transplantation and during follow-up, at 3 and 6 months. In addition, lack of detectable sjTRECs and, more notably, βTREC levels, strongly correlated with increased incidence of relapse.32 

Findings in adult UCBT recipients are in accord with these observations in pediatric patients. Increased TREC levels displayed a strong correlation with attainment of CMV-specific immunity and absence of CMV viremia in adult dUCBT recipients.41  Because CMV immunity was used as a paradigm for successful immune reconstitution, this study also investigated whether reconstitution of CMV-specific immunity and parameters of cellular T-cell immunity may be directly linked to distinct outcomes of progression-free survival (PFS) and OS. Univariable and multivariable analysis demonstrated that improved OS and PFS were significantly associated with the ability of patients to develop CMV-specific responses. Given that reconstitution of functional CMV-specific immunity and absence of viremia were significantly associated with TREC recovery, it was hypothesized that the restoration of thymopoiesis might also imply a successful immune reconstitution and improved capacity for generation of immune responses against other pathogens or tumor antigens. Consistent with this hypothesis, the main causes of death in this patient cohort were relapse, posttransplant lymphoproliferative disorder (PTLD), and sepsis. Furthermore, assessment of OS showed that patients whose TREC levels were 2000 copies per μg DNA (the lowest limit of values range in healthy individuals) or more by 1 year after transplantation had significantly improved OS compared with patients whose TREC values remained <2000 copies per μg (Figure 3). Similarly to these findings, in a cohort of 50 patients who underwent UCBT at the Karolinska Institute, subjects with TREC levels above the median value at 6 months had a trend of increased OS.47  Although the relationship between TRECs and OS did not reach statistical significance, this observation is in accord with another report from the same institution showing a significant correlation between TREC levels and OS in adult patients who underwent HSCT from adult donors.46  Other studies have also reported the prognostic value of thymic recovery on clinical outcomes after adult HSCT.68,69  Taken together, these observations support the intriguing hypothesis that thymic differentiation of pathogen-specific and leukemia-specific T cells might occur in parallel in UCBT recipients and offer an opportunity to investigate further the kinetics and mechanisms of regeneration of such antigen-specific T-cell populations.

Figure 3

PFS and OS are higher in patients with regeneration of thymic function than in patients with impaired thymic regeneration after UCBT. A cohort of adult recipients of dUCBT treated with 1 protocol of pretransplant conditioning and posttransplantation immunosuppression41  was categorized into 2 groups: (1) patients with TREC values ≥2000 copies per μg DNA at 1 year after transplantation and (2) patients with TREC values <2000 copies per μg DNA at 1 year after transplantation. Kaplan-Meier estimates of PFS and OS were calculated as a function of the maximum TREC concentration attained at 1 year. Patients with TREC levels exceeding 2000 copies per μg DNA had a significantly improved PFS (P = .01) (A) and OS (P = .03) (B) compared with patients with TREC levels <2000 copies per μg DNA (n = 22).

Figure 3

PFS and OS are higher in patients with regeneration of thymic function than in patients with impaired thymic regeneration after UCBT. A cohort of adult recipients of dUCBT treated with 1 protocol of pretransplant conditioning and posttransplantation immunosuppression41  was categorized into 2 groups: (1) patients with TREC values ≥2000 copies per μg DNA at 1 year after transplantation and (2) patients with TREC values <2000 copies per μg DNA at 1 year after transplantation. Kaplan-Meier estimates of PFS and OS were calculated as a function of the maximum TREC concentration attained at 1 year. Patients with TREC levels exceeding 2000 copies per μg DNA had a significantly improved PFS (P = .01) (A) and OS (P = .03) (B) compared with patients with TREC levels <2000 copies per μg DNA (n = 22).

Close modal

It has been described that dUCBT is associated with lower rates of leukemia relapse compared with sUCBT70  or HSCT from adult related or unrelated donors.71,72  This compensates for the higher treatment related mortality (TRM) at earlier time points, resulting in comparable long-term survival between dUCBT and HSCT recipients from adult donors. Although the mechanisms for reduced leukemia relapse in dUCBT recipients have not been identified, intriguing studies from MSKCC showed that dUCBT recipients have superior TCR repertoire diversity at 6 and 12 months after transplantation compared with patients undergoing HSCT from adult donor grafts, with or without T-cell depletion.43  Considering the essential role of the thymus for the diversification of TCR repertoire and the prompt recovery of TRECs after dUCBT,41  these observations provide indirect evidence that enhanced thymopoiesis might lead to the development of T cells with a broad TCR repertoire, including clones specific for graft-versus-leukemia (GVL), which account for the decreased relapse risk in dUCBT recipients.

These extensive studies provide compelling evidence that recovery of thymopoiesis plays a critical role in the generation of competent immune responses against viral or tumor antigens both in pediatric and adult UCBT recipients. Therefore, assessment of thymic reconstitution might be a valuable predictive factor of clinical outcomes after UCBT.

The use of dUCBT is currently the standard of care for adult patients in most US centers and is associated with improved myeloid engraftment and reduced risk of graft failure.73  Moreover, studies suggest that dUCBT recipients may also display faster thymic reconstitution compared with sUCBT recipients.40-42  This outcome may be related to a cell dose effect, with higher numbers of LMPs infused in dUCBT recipients leading to more efficient thymic seeding. However, alternative yet unidentified mechanisms may also be involved, considering that the majority of patients demonstrate single-unit chimerism by 3 months,37,44  before the emergence of RTEs).

Cytokine-based approaches

Experimental approaches to improve thymopoietic recovery after HSCT are under investigation in preclinical models or human clinical trials, although they have not been tested specifically in the setting of UCBT. KGF is an important trophic factor for TECs.74  In several murine models of HSCT, exogenous KGF administration has been shown to protect TECs from chemotherapy, radiation, or GVHD-induced damage, leading to increased production of intrathymic IL-7 and enhanced thymopoiesis.75  However, in human trials, administration of KGF did not show a beneficial effect on immune recovery.76 

Administration of IL-7 in mice has shown not only to enhance thymic reconstitution, but also has a beneficial effect on the thymic-independent pathway of T-cell regeneration by mediating survival and proliferation of naive T cells.77,78  The immunorestorative effects of IL-7 have similarly been observed in primates and, more recently, in clinical trials in humans.79  However, studies have not provided definitive evidence for augmented thymopoiesis in humans. Patients with advanced malignancies80  or HIV81  treated with exogenous recombinant human IL-7 (rhIL-7) displayed a sustained expansion of T cells, preferentially of the naive subsets, higher TRECs, and a marked increase in the TCR repertoire diversity. On the other hand, no increase in thymic size was noted by positron emission tomography/computed tomography scanning, suggesting that IL-7 may primarily work at a postthymic level by enhancing proliferation of naive T cells including RTEs.80  In the same regard, it is important to note that IL-7 plays an important role in the trafficking of RTEs among peripheral blood and secondary lymphoid orgrans,82  possibly confounding the use of TREC assays as markers of thymic output. Recently, rhIL-7 was tested in a phase 1 trial of 12 patients undergoing T-cell-depleted HSCT.83  This resulted in significant expansion of effector memory T cells, which was associated with an increase in virus-specific T cells and enhanced TCR repertoire. However, no effect on TREC levels and RTE output was observed, at least at the tested IL-7 dose and follow-up period of the study.

In a murine model of HSCT, treatment with Fms-related tyrosine kinase 3 ligand (FLT3L) resulted in significantly increased numbers of thymic-dependent progeny and TREC+ T cells, suggesting increased thymopoiesis.84  However, although FLT3L increased thymic output in peripheral organs, the thymus itself was not evaluated in that study. Thus, it remains equivocal whether thymopoiesis was actually increased or this observation is due to pre- or postthymic mechanisms, such as thymic homing of LMPs or RTE trafficking, especially considering that FLT3L has potent effects on hematopoietic stem cells and also enhances the thymic-independent pathway of peripheral T-cell expansion.84,85  Sex steroid ablation with luteinizing hormone-releasing hormone analogue (LHRH-A) before HSCT in mice is associated with increased numbers of myeloid and lymphoid progenitors in the BM and improved T-cell recovery.86  In humans, LHRH-A administration prior to HSCT has been shown to lead to faster recovery of total and naive TREC+CD4+ T cells and diversification of T-cell repertoire, suggesting an effect of sex hormone blockade on thymic recovery.87  In a mouse model of radiation-induced thymic injury, it was determined that depletion of DP thymocytes triggered intrathymic production of IL-22,88  which appeared to have an active role in thymic recovery by promoting survival and proliferation of TECs. Importantly, systemic administration of IL-22 enhanced thymopoiesis following total body irradiation. Whether the above approaches are applicable to UCBT remains to be determined.

Cell-based approaches

To enhance HSC engraftment of the UCB, several groups have explored various strategies of ex vivo expansion of the UCB.89  Based on preclinical observations that the Notch signaling pathway plays a critical role in the regulation of self-renewal and repopulating ability of HSC progenitors of the UCB90,91  as well as fate decision of HSCs toward T-cell differentiation,92  Delaney et al have developed an ex vivo culture system for UCB progenitors based on immobilized Notch ligand Delta 1.93  However, in a phase 1 trial of dUCBT using 1 ex vivo–expanded and 1 unmanipulated unit, although a transient multilog increase in the ex vivo–expanded CD34+ and total nucleated cells (TNCs) was observed, long-term hematopoietic reconstitution was achieved by the nonexpanded unit. Similarly to the effects of ex vivo expansion via Notch-mediated signaling, ex vivo coculture of UCB mononuclear cells with mesenchymal cells resulted in a rapid increase of neutrophil numbers after dUCBT.94  However, this approach also induced only transient chimerism from the ex vivo–manipulated UCB, which eventually declined, resulting in long-term engraftment and hematopoiesis by the unmanipulated UCB in all patients. No effects on the reconstitution of adaptive immunity were noted by any of these cell-based approaches.

In an effort to selectively improve thymic dysfunction and T-cell reconstitution after HSCT, recent studies discovered that human progenitor T cells, developed in vitro from UCB stem cells with the use of OP9-DL1 culture system, could be a source of thymus-seeding progenitors when infused in NSG mice. Detailed cellular analysis revealed that between 2 distinct in vitro–derived progenitor human T-cell populations (proT1 and proT2), proT2 cells (CD34+CD45RA+CD7++) had superior engrafting capacity in the recipient thymus. More importantly, these in vitro–derived proT2 cells could also restore thymic architecture and when coinfused with HSCs, were able to promote HSC-derived T-cell lymphopoiesis.95  These findings suggest that Notch-based ex vivo culture of human progenitor T cells might represent a clinically applicable novel method to support T-cell reconstitution in UCBT recipients.

Based on previous observations that prostaglandin E2 (PGE2) is a critical regulator of HSC homeostasis,96  Dana-Farber Harvard Cancer Center conducted a phase 1 trial of dUCBT using 1 ex vivo PGE2-treated UCB unit and 1 unmanipulated unit.97  This study showed enhanced neutrophil recovery in comparison with historic controls and, more importantly, long-term dominance of the PGE2-treated unit. Besides the effect on myeloid engraftment, this study also determined that PGE2 promoted the survival of UCB T cells via the Wnt/β-catenin pathway.98  In a different experimental system it was previously determined that Wnt signals mediate proliferation and cell adhesion, but not differentiation of immature thymocytes.99  Therefore, modulation of the PGE2/Wnt axis might also have a beneficial effect on thymopoiesis. In support of this hypothesis, a preliminary immune reconstitution analysis showed less TCR clonality in patients with PGE2-treated UCB unit dominance compared with historic control dUCBT recipients and patients with dominance of the non-PGE2-treated unit.100  Although preliminary, these findings suggest that PGE2 treatment might alter the properties of UCB-derived thymocyte progenitors, including survival, thymus colonization, and expansion, leading to enhanced thymopoiesis.

UCB is a valuable HSC source for patients requiring allogeneic transplantation who lack suitable sibling or unrelated adult donors. The distinct biologic properties of UCB grafts allow for a greater degree of HLA disparity and lead to lower rates of GVHD in UCBT recipients, without compromising the GVL effect. However, UCBT is associated with delayed engraftment and immune reconstitution, resulting in higher rates of early posttransplant infectious complications. Recovery of thymopoiesis is an imperative component of immune reconstitution after UCBT and plays an essential role in the restoration of peripheral T-cell compartment by the de novo production of naive lymphocytes with diverse TCR repertoire. Furthermore, parameters of thymic function have significant prognostic value for clinical outcomes in UCBT recipients. The use of double UCBT has lead to faster thymic regeneration in adult patients. Further strategies to improve thymic seeding, intrathymic proliferation, and differentiation of LMPs, or to protect the thymic microenvironment from the detrimental effects of conditioning and GVHD may further improve the outcomes of UCB transplantation.

This work was supported by the National Institutes of Health grants R56A1098129 and RO1CA183605, and by the HHV-6 Foundation pilot grant.

Contribution: I.P. and V.A.B. wrote the paper.

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

Correspondence: Vassiliki A. Boussiotis, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Dana 513-518, Boston MA 02215; e-mail: vboussio@bidmc.harvard.edu.

1
Gluckman
 
E
Broxmeyer
 
HA
Auerbach
 
AD
, et al. 
Hematopoietic reconstitution in a patient with Fanconi’s anemia by means of umbilical-cord blood from an HLA-identical sibling.
N Engl J Med
1989
, vol. 
321
 
17
(pg. 
1174
-
1178
)
2
Gluckman
 
E
Rocha
 
V
Boyer-Chammard
 
A
, et al. 
Eurocord Transplant Group and the European Blood and Marrow Transplantation Group
Outcome of cord-blood transplantation from related and unrelated donors.
N Engl J Med
1997
, vol. 
337
 
6
(pg. 
373
-
381
)
3
Rubinstein
 
P
Carrier
 
C
Scaradavou
 
A
, et al. 
Outcomes among 562 recipients of placental-blood transplants from unrelated donors.
N Engl J Med
1998
, vol. 
339
 
22
(pg. 
1565
-
1577
)
4
Rocha
 
V
Labopin
 
M
Sanz
 
G
, et al. 
Acute Leukemia Working Party of European Blood and Marrow Transplant Group
Eurocord-Netcord Registry
Transplants of umbilical-cord blood or bone marrow from unrelated donors in adults with acute leukemia.
N Engl J Med
2004
, vol. 
351
 
22
(pg. 
2276
-
2285
)
5
Rocha
 
V
Wagner
 
JE
Sobocinski
 
KA
, et al. 
Eurocord and International Bone Marrow Transplant Registry Working Committee on Alternative Donor and Stem Cell Sources
Graft-versus-host disease in children who have received a cord-blood or bone marrow transplant from an HLA-identical sibling.
N Engl J Med
2000
, vol. 
342
 
25
(pg. 
1846
-
1854
)
6
Chalmers
 
IM
Janossy
 
G
Contreras
 
M
Navarrete
 
C
Intracellular cytokine profile of cord and adult blood lymphocytes.
Blood
1998
, vol. 
92
 
1
(pg. 
11
-
18
)
7
Risdon
 
G
Gaddy
 
J
Stehman
 
FB
Broxmeyer
 
HE
Proliferative and cytotoxic responses of human cord blood T lymphocytes following allogeneic stimulation.
Cell Immunol
1994
, vol. 
154
 
1
(pg. 
14
-
24
)
8
Godfrey
 
WR
Spoden
 
DJ
Ge
 
YG
, et al. 
Cord blood CD4(+)CD25(+)-derived T regulatory cell lines express FoxP3 protein and manifest potent suppressor function.
Blood
2005
, vol. 
105
 
2
(pg. 
750
-
758
)
9
Brown
 
JA
Boussiotis
 
VA
Umbilical cord blood transplantation: basic biology and clinical challenges to immune reconstitution.
Clin Immunol
2008
, vol. 
127
 
3
(pg. 
286
-
297
)
10
Williams
 
KM
Hakim
 
FT
Gress
 
RE
T cell immune reconstitution following lymphodepletion.
Semin Immunol
2007
, vol. 
19
 
5
(pg. 
318
-
330
)
11
Garderet
 
L
Dulphy
 
N
Douay
 
C
, et al. 
The umbilical cord blood alphabeta T-cell repertoire: characteristics of a polyclonal and naive but completely formed repertoire.
Blood
1998
, vol. 
91
 
1
(pg. 
340
-
346
)
12
Szabolcs
 
P
Niedzwiecki
 
D
Immune reconstitution after unrelated cord blood transplantation.
Cytotherapy
2007
, vol. 
9
 
2
(pg. 
111
-
122
)
13
Komanduri
 
KV
St John
 
LS
de Lima
 
M
, et al. 
Delayed immune reconstitution after cord blood transplantation is characterized by impaired thymopoiesis and late memory T-cell skewing.
Blood
2007
, vol. 
110
 
13
(pg. 
4543
-
4551
)
14
Roux
 
E
Dumont-Girard
 
F
Starobinski
 
M
, et al. 
Recovery of immune reactivity after T-cell-depleted bone marrow transplantation depends on thymic activity.
Blood
2000
, vol. 
96
 
6
(pg. 
2299
-
2303
)
15
De Smedt
 
M
Reynvoet
 
K
Kerre
 
T
, et al. 
Active form of Notch imposes T cell fate in human progenitor cells.
J Immunol
2002
, vol. 
169
 
6
(pg. 
3021
-
3029
)
16
Gill
 
J
Malin
 
M
Sutherland
 
J
Gray
 
D
Hollander
 
G
Boyd
 
R
Thymic generation and regeneration.
Immunol Rev
2003
, vol. 
195
 (pg. 
28
-
50
)
17
Krenger
 
W
Holländer
 
GA
The immunopathology of thymic GVHD.
Semin Immunopathol
2008
, vol. 
30
 
4
(pg. 
439
-
456
)
18
Godthelp
 
BC
van Tol
 
MJ
Vossen
 
JM
van Den Elsen
 
PJ
T-Cell immune reconstitution in pediatric leukemia patients after allogeneic bone marrow transplantation with T-cell-depleted or unmanipulated grafts: evaluation of overall and antigen-specific T-cell repertoires.
Blood
1999
, vol. 
94
 
12
(pg. 
4358
-
4369
)
19
Eyrich
 
M
Leiler
 
C
Lang
 
P
, et al. 
A prospective comparison of immune reconstitution in pediatric recipients of positively selected CD34+ peripheral blood stem cells from unrelated donors vs recipients of unmanipulated bone marrow from related donors.
Bone Marrow Transplant
2003
, vol. 
32
 
4
(pg. 
379
-
390
)
20
Douek
 
DC
Koup
 
RA
Evidence for thymic function in the elderly.
Vaccine
2000
, vol. 
18
 
16
(pg. 
1638
-
1641
)
21
Michie
 
CA
McLean
 
A
Alcock
 
C
Beverley
 
PC
Lifespan of human lymphocyte subsets defined by CD45 isoforms.
Nature
1992
, vol. 
360
 
6401
(pg. 
264
-
265
)
22
McFarland
 
RD
Douek
 
DC
Koup
 
RA
Picker
 
LJ
Identification of a human recent thymic emigrant phenotype.
Proc Natl Acad Sci USA
2000
, vol. 
97
 
8
(pg. 
4215
-
4220
)
23
Kohler
 
S
Thiel
 
A
Life after the thymus: CD31+ and CD31- human naive CD4+ T-cell subsets.
Blood
2009
, vol. 
113
 
4
(pg. 
769
-
774
)
24
Douek
 
DC
McFarland
 
RD
Keiser
 
PH
, et al. 
Changes in thymic function with age and during the treatment of HIV infection.
Nature
1998
, vol. 
396
 
6712
(pg. 
690
-
695
)
25
Livak
 
F
Schatz
 
DG
T-cell receptor alpha locus V(D)J recombination by-products are abundant in thymocytes and mature T cells.
Mol Cell Biol
1996
, vol. 
16
 
2
(pg. 
609
-
618
)
26
Krenger
 
W
Blazar
 
BR
Holländer
 
GA
Thymic T-cell development in allogeneic stem cell transplantation.
Blood
2011
, vol. 
117
 
25
(pg. 
6768
-
6776
)
27
Dion
 
ML
Poulin
 
JF
Bordi
 
R
, et al. 
HIV infection rapidly induces and maintains a substantial suppression of thymocyte proliferation.
Immunity
2004
, vol. 
21
 
6
(pg. 
757
-
768
)
28
Douek
 
DC
Vescio
 
RA
Betts
 
MR
, et al. 
Assessment of thymic output in adults after haematopoietic stem-cell transplantation and prediction of T-cell reconstitution.
Lancet
2000
, vol. 
355
 
9218
(pg. 
1875
-
1881
)
29
Niehues
 
T
Rocha
 
V
Filipovich
 
AH
, et al. 
Factors affecting lymphocyte subset reconstitution after either related or unrelated cord blood transplantation in children — a Eurocord analysis.
Br J Haematol
2001
, vol. 
114
 
1
(pg. 
42
-
48
)
30
Moretta
 
A
Maccario
 
R
Fagioli
 
F
, et al. 
Analysis of immune reconstitution in children undergoing cord blood transplantation.
Exp Hematol
2001
, vol. 
29
 
3
(pg. 
371
-
379
)
31
Rénard
 
C
Barlogis
 
V
Mialou
 
V
, et al. 
Lymphocyte subset reconstitution after unrelated cord blood or bone marrow transplantation in children.
Br J Haematol
2011
, vol. 
152
 
3
(pg. 
322
-
330
)
32
Clave
 
E
Lisini
 
D
Douay
 
C
, et al. 
Thymic function recovery after unrelated donor cord blood or T-cell depleted HLA-haploidentical stem cell transplantation correlates with leukemia relapse.
Front Immunol
2013
, vol. 
4
 pg. 
54
 
33
Klein
 
AK
Patel
 
DD
Gooding
 
ME
, et al. 
T-cell recovery in adults and children following umbilical cord blood transplantation.
Biol Blood Marrow Transplant
2001
, vol. 
7
 
8
(pg. 
454
-
466
)
34
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
, vol. 
156
 
5
(pg. 
656
-
666
)
35
Talvensaari
 
K
Clave
 
E
Douay
 
C
, et al. 
A broad T-cell repertoire diversity and an efficient thymic function indicate a favorable long-term immune reconstitution after cord blood stem cell transplantation.
Blood
2002
, vol. 
99
 
4
(pg. 
1458
-
1464
)
36
Ballen
 
KK
Spitzer
 
TR
Yeap
 
BY
, et al. 
Double unrelated reduced-intensity umbilical cord blood transplantation in adults.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
1
(pg. 
82
-
89
)
37
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
, vol. 
105
 
3
(pg. 
1343
-
1347
)
38
Jacobson
 
CA
Turki
 
AT
McDonough
 
SM
, et al. 
Immune reconstitution after double umbilical cord blood stem cell transplantation: comparison with unrelated peripheral blood stem cell transplantation.
Biol Blood Marrow Transplant
2012
, vol. 
18
 
4
(pg. 
565
-
574
)
39
Ruggeri
 
A
Peffault de Latour
 
R
Carmagnat
 
M
, et al. 
Outcomes, infections, and immune reconstitution after double cord blood transplantation in patients with high-risk hematological diseases.
Transpl Infect Dis
2011
, vol. 
13
 
5
(pg. 
456
-
465
)
40
Kanda
 
J
Chiou
 
LW
Szabolcs
 
P
, et al. 
Immune recovery in adult patients after myeloablative dual umbilical cord blood, matched sibling, and matched unrelated donor hematopoietic cell transplantation.
Biol Blood Marrow Transplant
2012
, vol. 
18
 
11
(pg. 
1664
-
1676, e1
)
41
Brown
 
JA
Stevenson
 
K
Kim
 
HT
, et al. 
Clearance of CMV viremia and survival after double umbilical cord blood transplantation in adults depends on reconstitution of thymopoiesis.
Blood
2010
, vol. 
115
 
20
(pg. 
4111
-
4119
)
42
Ballen
 
K
Mendizabal
 
AM
Cutler
 
C
, et al. 
Phase II trial of parathyroid hormone after double umbilical cord blood transplantation.
Biol Blood Marrow Transplant
2012
, vol. 
18
 
12
(pg. 
1851
-
1858
)
43
van Heijst
 
JW
Ceberio
 
I
Lipuma
 
LB
, et al. 
Quantitative assessment of T cell repertoire recovery after hematopoietic stem cell transplantation.
Nat Med
2013
, vol. 
19
 
3
(pg. 
372
-
377
)
44
Brunstein
 
CG
Barker
 
JN
Weisdorf
 
DJ
, et al. 
Umbilical cord blood transplantation after nonmyeloablative conditioning: impact on transplantation outcomes in 110 adults with hematologic disease.
Blood
2007
, vol. 
110
 
8
(pg. 
3064
-
3070
)
45
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
, vol. 
113
 
25
(pg. 
6477
-
6484
)
46
Sairafi
 
D
Mattsson
 
J
Uhlin
 
M
Uzunel
 
M
Thymic function after allogeneic stem cell transplantation is dependent on graft source and predictive of long term survival.
Clin Immunol
2012
, vol. 
142
 
3
(pg. 
343
-
350
)
47
Uhlin
 
M
Sairafi
 
D
Berglund
 
S
, et al. 
Mesenchymal stem cells inhibit thymic reconstitution after allogeneic cord blood transplantation.
Stem Cells Dev
2012
, vol. 
21
 
9
(pg. 
1409
-
1417
)
48
Weinberg
 
K
Blazar
 
BR
Wagner
 
JE
, et al. 
Factors affecting thymic function after allogeneic hematopoietic stem cell transplantation.
Blood
2001
, vol. 
97
 
5
(pg. 
1458
-
1466
)
49
Kosugi
 
A
Zuniga-Pflucker
 
JC
Sharrow
 
SO
Kruisbeek
 
AM
Shearer
 
GM
Effect of cyclosporin A on lymphopoiesis. II. Developmental defects of immature and mature thymocytes in fetal thymus organ cultures treated with cyclosporin A.
J Immunol
1989
, vol. 
143
 
10
(pg. 
3134
-
3140
)
50
Hiramine
 
C
Hojo
 
K
Matsumoto
 
H
Abnormal distribution of T cell subsets in the thymus of cyclosporin A (CsA)-treated mice.
Thymus
1988
, vol. 
11
 
4
(pg. 
243
-
252
)
51
Mackall
 
CL
Fleisher
 
TA
Brown
 
MR
, et al. 
Age, thymopoiesis, and CD4+ T-lymphocyte regeneration after intensive chemotherapy.
N Engl J Med
1995
, vol. 
332
 
3
(pg. 
143
-
149
)
52
Chung
 
B
Barbara-Burnham
 
L
Barsky
 
L
Weinberg
 
K
Radiosensitivity of thymic interleukin-7 production and thymopoiesis after bone marrow transplantation.
Blood
2001
, vol. 
98
 
5
(pg. 
1601
-
1606
)
53
Jiménez
 
M
Martínez
 
C
Ercilla
 
G
, et al. 
Reduced-intensity conditioning regimen preserves thymic function in the early period after hematopoietic stem cell transplantation.
Exp Hematol
2005
, vol. 
33
 
10
(pg. 
1240
-
1248
)
54
Chao
 
NJ
Liu
 
CX
Rooney
 
B
, et al. 
Nonmyeloablative regimen preserves “niches” allowing for peripheral expansion of donor T-cells.
Biol Blood Marrow Transplant
2002
, vol. 
8
 
5
(pg. 
249
-
256
)
55
Clave
 
E
Rocha
 
V
Talvensaari
 
K
, et al. 
Prognostic value of pretransplantation host thymic function in HLA-identical sibling hematopoietic stem cell transplantation.
Blood
2005
, vol. 
105
 
6
(pg. 
2608
-
2613
)
56
Hill
 
JA
Koo
 
S
Guzman Suarez
 
BB
, et al. 
Cord-blood hematopoietic stem cell transplant confers an increased risk for human herpesvirus-6-associated acute limbic encephalitis: a cohort analysis.
Biol Blood Marrow Transplant
2012
, vol. 
18
 
11
(pg. 
1638
-
1648
)
57
Silva
 
LP
Patah
 
PA
Saliba
 
RM
, et al. 
Hemorrhagic cystitis after allogeneic hematopoietic stem cell transplants is the complex result of BK virus infection, preparative regimen intensity and donor type.
Haematologica
2010
, vol. 
95
 
7
(pg. 
1183
-
1190
)
58
Vandenbosch
 
K
Ovetchkine
 
P
Champagne
 
MA
Haddad
 
E
Alexandrov
 
L
Duval
 
M
Varicella-zoster virus disease is more frequent after cord blood than after bone marrow transplantation.
Biol Blood Marrow Transplant
2008
, vol. 
14
 
8
(pg. 
867
-
871
)
59
Beck
 
JC
Wagner
 
JE
DeFor
 
TE
, et al. 
Impact of cytomegalovirus (CMV) reactivation after umbilical cord blood transplantation.
Biol Blood Marrow Transplant
2010
, vol. 
16
 
2
(pg. 
215
-
222
)
60
Dumas
 
PY
Ruggeri
 
A
Robin
 
M
, et al. 
Incidence and risk factors of EBV reactivation after unrelated cord blood transplantation: a Eurocord and Société Française de Greffe de Moelle-Therapie Cellulaire collaborative study.
Bone Marrow Transplant
2013
, vol. 
48
 
2
(pg. 
253
-
256
)
61
Walker
 
CM
van Burik
 
JA
De For
 
TE
Weisdorf
 
DJ
Cytomegalovirus infection after allogeneic transplantation: comparison of cord blood with peripheral blood and marrow graft sources.
Biol Blood Marrow Transplant
2007
, vol. 
13
 
9
(pg. 
1106
-
1115
)
62
Albano
 
MS
Taylor
 
P
Pass
 
RF
, et al. 
Umbilical cord blood transplantation and cytomegalovirus: Posttransplantation infection and donor screening.
Blood
2006
, vol. 
108
 
13
(pg. 
4275
-
4282
)
63
Tomonari
 
A
Iseki
 
T
Ooi
 
J
, et al. 
Cytomegalovirus infection following unrelated cord blood transplantation for adult patients: a single institute experience in Japan.
Br J Haematol
2003
, vol. 
121
 
2
(pg. 
304
-
311
)
64
McGoldrick
 
SM
Bleakley
 
ME
Guerrero
 
A
, et al. 
Cytomegalovirus-specific T cells are primed early after cord blood transplant but fail to control virus in vivo.
Blood
2013
, vol. 
121
 
14
(pg. 
2796
-
2803
)
65
Shedlock
 
DJ
Shen
 
H
Requirement for CD4 T cell help in generating functional CD8 T cell memory.
Science
2003
, vol. 
300
 
5617
(pg. 
337
-
339
)
66
Parkman
 
R
Cohen
 
G
Carter
 
SL
, et al. 
Successful immune reconstitution decreases leukemic relapse and improves survival in recipients of unrelated cord blood transplantation.
Biol Blood Marrow Transplant
2006
, vol. 
12
 
9
(pg. 
919
-
927
)
67
Cohen
 
G
Carter
 
SL
Weinberg
 
KI
, et al. 
Antigen-specific T-lymphocyte function after cord blood transplantation.
Biol Blood Marrow Transplant
2006
, vol. 
12
 
12
(pg. 
1335
-
1342
)
68
Wils
 
EJ
van der Holt
 
B
Broers
 
AE
, et al. 
Insufficient recovery of thymopoiesis predicts for opportunistic infections in allogeneic hematopoietic stem cell transplant recipients.
Haematologica
2011
, vol. 
96
 
12
(pg. 
1846
-
1854
)
69
Lewin
 
SR
Heller
 
G
Zhang
 
L
, et al. 
Direct evidence for new T-cell generation by patients after either T-cell-depleted or unmodified allogeneic hematopoietic stem cell transplantations.
Blood
2002
, vol. 
100
 
6
(pg. 
2235
-
2242
)
70
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
, vol. 
114
 
19
(pg. 
4293
-
4299
)
71
Ponce
 
DM
Zheng
 
J
Gonzales
 
AM
, et al. 
Reduced late mortality risk contributes to similar survival after double-unit cord blood transplantation compared with related and unrelated donor hematopoietic stem cell transplantation.
Biol Blood Marrow Transplant
2011
, vol. 
17
 
9
(pg. 
1316
-
1326
)
72
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
, vol. 
116
 
22
(pg. 
4693
-
4699
)
73
Cutler
 
C
Stevenson
 
K
Kim
 
HT
, et al. 
Double umbilical cord blood transplantation with reduced intensity conditioning and sirolimus-based GVHD prophylaxis.
Bone Marrow Transplant
2011
, vol. 
46
 
5
(pg. 
659
-
667
)
74
Alpdogan
 
O
Hubbard
 
VM
Smith
 
OM
, et al. 
Keratinocyte growth factor (KGF) is required for postnatal thymic regeneration.
Blood
2006
, vol. 
107
 
6
(pg. 
2453
-
2460
)
75
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
, vol. 
99
 
12
(pg. 
4592
-
4600
)
76
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
, vol. 
86
 
10
(pg. 
879
-
882
)
77
Alpdogan
 
O
Schmaltz
 
C
Muriglan
 
SJ
, et al. 
Administration of interleukin-7 after allogeneic bone marrow transplantation improves immune reconstitution without aggravating graft-versus-host disease.
Blood
2001
, vol. 
98
 
7
(pg. 
2256
-
2265
)
78
Mackall
 
CL
Fry
 
TJ
Bare
 
C
Morgan
 
P
Galbraith
 
A
Gress
 
RE
IL-7 increases both thymic-dependent and thymic-independent T-cell regeneration after bone marrow transplantation.
Blood
2001
, vol. 
97
 
5
(pg. 
1491
-
1497
)
79
Mackall
 
CL
Fry
 
TJ
Gress
 
RE
Harnessing the biology of IL-7 for therapeutic application.
Nat Rev Immunol
2011
, vol. 
11
 
5
(pg. 
330
-
342
)
80
Sportès
 
C
Hakim
 
FT
Memon
 
SA
, et al. 
Administration of rhIL-7 in humans increases in vivo TCR repertoire diversity by preferential expansion of naive T cell subsets.
J Exp Med
2008
, vol. 
205
 
7
(pg. 
1701
-
1714
)
81
Lévy
 
Y
Sereti
 
I
Tambussi
 
G
, et al. 
Effects of recombinant human interleukin 7 on T-cell recovery and thymic output in HIV-infected patients receiving antiretroviral therapy: results of a phase I/IIa randomized, placebo-controlled, multicenter study.
Clin Infect Dis
2012
, vol. 
55
 
2
(pg. 
291
-
300
)
82
Chu
 
YW
Memon
 
SA
Sharrow
 
SO
, et al. 
Exogenous IL-7 increases recent thymic emigrants in peripheral lymphoid tissue without enhanced thymic function.
Blood
2004
, vol. 
104
 
4
(pg. 
1110
-
1119
)
83
Perales
 
MA
Goldberg
 
JD
Yuan
 
J
, et al. 
Recombinant human interleukin-7 (CYT107) promotes T-cell recovery after allogeneic stem cell transplantation.
Blood
2012
, vol. 
120
 
24
(pg. 
4882
-
4891
)
84
Fry
 
TJ
Sinha
 
M
Milliron
 
M
, et al. 
Flt3 ligand enhances thymic-dependent and thymic-independent immune reconstitution.
Blood
2004
, vol. 
104
 
9
(pg. 
2794
-
2800
)
85
Lyman
 
SD
James
 
L
Vanden Bos
 
T
, et al. 
Molecular cloning of a ligand for the flt3/flk-2 tyrosine kinase receptor: a proliferative factor for primitive hematopoietic cells.
Cell
1993
, vol. 
75
 
6
(pg. 
1157
-
1167
)
86
Goldberg
 
GL
King
 
CG
Nejat
 
RA
, et al. 
Luteinizing hormone-releasing hormone enhances T cell recovery following allogeneic bone marrow transplantation.
J Immunol
2009
, vol. 
182
 
9
(pg. 
5846
-
5854
)
87
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
, vol. 
14
 
4
(pg. 
1138
-
1149
)
88
Dudakov
 
JA
Hanash
 
AM
Jenq
 
RR
, et al. 
Interleukin-22 drives endogenous thymic regeneration in mice.
Science
2012
, vol. 
336
 
6077
(pg. 
91
-
95
)
89
Dahlberg
 
A
Delaney
 
C
Bernstein
 
ID
Ex vivo expansion of human hematopoietic stem and progenitor cells.
Blood
2011
, vol. 
117
 
23
(pg. 
6083
-
6090
)
90
Ohishi
 
K
Varnum-Finney
 
B
Bernstein
 
ID
Delta-1 enhances marrow and thymus repopulating ability of human CD34(+)CD38(-) cord blood cells.
J Clin Invest
2002
, vol. 
110
 
8
(pg. 
1165
-
1174
)
91
Delaney
 
C
Varnum-Finney
 
B
Aoyama
 
K
Brashem-Stein
 
C
Bernstein
 
ID
Dose-dependent effects of the Notch ligand Delta1 on ex vivo differentiation and in vivo marrow repopulating ability of cord blood cells.
Blood
2005
, vol. 
106
 
8
(pg. 
2693
-
2699
)
92
La Motte-Mohs
 
RN
Herer
 
E
Zúñiga-Pflücker
 
JC
Induction of T-cell development from human cord blood hematopoietic stem cells by Delta-like 1 in vitro.
Blood
2005
, vol. 
105
 
4
(pg. 
1431
-
1439
)
93
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
, vol. 
16
 
2
(pg. 
232
-
236
)
94
de Lima
 
M
McNiece
 
I
Robinson
 
SN
, et al. 
Cord-blood engraftment with ex vivo mesenchymal-cell coculture.
N Engl J Med
2012
, vol. 
367
 
24
(pg. 
2305
-
2315
)
95
Awong
 
G
Singh
 
J
Mohtashami
 
M
, et al. 
Human proT-cells generated in vitro facilitate hematopoietic stem cell-derived T-lymphopoiesis in vivo and restore thymic architecture.
Blood
2013
, vol. 
122
 
26
(pg. 
4210
-
4219
)
96
North
 
TE
Goessling
 
W
Walkley
 
CR
, et al. 
Prostaglandin E2 regulates vertebrate haematopoietic stem cell homeostasis.
Nature
2007
, vol. 
447
 
7147
(pg. 
1007
-
1011
)
97
Cutler
 
C
Multani
 
P
Robbins
 
D
, et al. 
Prostaglandin-modulated umbilical cord blood hematopoietic stem cell transplantation.
Blood
2013
, vol. 
122
 
17
(pg. 
3074
-
3081
)
98
Li
 
L
Kim
 
HT
Nellore
 
A
, et al. 
Prostaglandin E2 promotes survival of naive UCB T cells via the Wnt/β-catenin pathway and alters immune reconstitution after UCBT.
Blood Cancer J
2014
, vol. 
4
 pg. 
e178
 
99
Staal
 
FJ
Weerkamp
 
F
Baert
 
MR
, et al. 
Wnt target genes identified by DNA microarrays in immature CD34+ thymocytes regulate proliferation and cell adhesion.
J Immunol
2004
, vol. 
172
 
2
(pg. 
1099
-
1108
)
100
Nikiforow
 
S
McDonough
 
SM
Emerson
 
RO
, et al. 
Evolution of T cell repertoire diversity after reduced-intensity conditioning and double umbilical cord blood transplantation with or without exposure to FT1050 (16,16-dimethyl prostaglandin E2).
Biol Blood Marrow Transplant
2013
, vol. 
19
 
2
(pg. 
S206
-
S207
Abstract 187
Sign in via your Institution