Severe combined immunodeficiency (SCID) is a syndrome of diverse genetic cause characterized by profound deficiencies of T, B, and sometimes NK-cell function. Nonablative human leukocyte antigen–identical or rigorously T cell–depleted haploidentical parental bone marrow transplantation (BMT) results in thymus-dependent genetically donor T-cell development in the recipients, leading to long-term survival. We reported previously that normal T-cell numbers, function, and repertoire developed by 3 to 4 months after transplantation in SCID patients, and the repertoire remained highly diverse for the first 10 years after BMT. The T-cell receptor diversity positively correlated with T-cell receptor excision circle levels, a reflection of thymic output. However, the fate of thymic function in SCID patients beyond 10 to 12 years after BMT remained to be determined. In this greater than 25-year follow-up study of 128 patients with 11 different molecular types of SCID after nonconditioned BMT, we provide evidence that T-cell function, thymic output, and T-cell clonal diversity are maintained long-term.

Transplantation of human leukocyte antigen (HLA)–identical or haploidentical allogeneic-related, rigorously T cell–depleted bone marrow (BM) cells into humans with severe combined immunodeficiency (SCID) without pretransplantation chemotherapy or posttransplantation graft-versus-host disease (GVHD) prophylaxis results in immune reconstitution with a high survival rate.1,2  Within 3 to 4 months after transplantation, the number of genetically donor T cells and T-cell function gradually increase to normal levels in SCID recipients.1-3  Studies by this group have demonstrated that the T-cell reconstitution in SCID recipients of rigorously T cell–depleted allogeneic-related BM cells is the result of the development and maturation of donor stem cells in the infant's vestigial thymus.4,5  Within the SCID thymus, donor T-cell precursors undergo T-cell receptor (TCR) gene rearrangements by the junction of V(D)J gene segments and by the addition of N nucleotides. The process of TCR rearrangement generates extrachromosomal DNA episomes or TCR excision circles (TRECs), which can be detected in new T cells. The presence of TRECs in circulating T cells is an indication that rearrangement of their TCR genes has recently occurred in the thymus.6  Thymic function in normal persons (0-34 years of age), as measured by the number of TREC copies in peripheral blood mononuclear cells (PBMCs), ranges between 4 and 3 log10 TRECs/μg PBMC DNA.6  Infants with SCID who received HLA-identical or haploidentical T cell–depleted BM transplantations (BMTs) have the ability to generate T cells with newly rearranged antigen receptors.4,7  We reported 8 years ago that thymus-derived naive CD45RA+ T cells carrying TRECs become the predominant T cells in the circulation in SCID patients from 150 days after BMT.4,7  The number of CD45RA+ cells declined thereafter, but CD45RA+ T cells predominated over CD45RO+ memory phenotype T cells for 10 to 12 years. At the time of that publication, however, data were available on only a few patients who were more than 10 years after transplantation.

In other studies, we also demonstrated that maturation and selection of donor-derived precursor T cells from transplanted BM into SCID recipients result in the development of a diverse T-cell repertoire by 1 to 2 years after BMT, which correlates with the rise in the number of recent thymic emigrant (CD45RA+) T cells of donor origin and an increase in TRECs.5  We observed a direct correlation between TREC values and T-cell diversity that developed in the analyzed patients. A more skewed T-cell repertoire emerged 10 years after BMT5  that was predominantly found in the CD8 CD45RO+ T-cell subset, a physiologic phenomenon occurring with the development of immunologic memory and with aging in normal persons.

In our initial report,4  the fate of thymic function in SCID patients beyond 10 to 12 years after BMT remained to be determined. Some patients have now survived for nearly 26 years, have normal numbers of T, B, and NK cells, have normal T-cell function, and are generally healthy. In the present longitudinal study, we had the unique opportunity to perform a continued analysis beyond 10 to 12 years after transplantation of thymic reconstitution, T-cell clonal diversity, and T-cell function in 128 patients with 11 different molecular types of SCID.

Patients

We studied PBMCs from 128 SCID patients who were given unfractionated HLA-identical related BMT (N = 8), T cell–depleted HLA-identical related BMT (N = 8), or rigorously T cell–depleted HLA-haploidentical parental BMT (N = 112) between 1982 and 2007. None of the patients received pretransplantation chemotherapy or prophylactic drugs to prevent GVHD after transplantation. These 128 patients included all 123 of the survivors from a total of 158 SCID infants transplanted initially at Duke University Medical Center (DUMC) between May 19, 1982 and December 31, 2007, 2 more that had been transplanted elsewhere but given booster transplantations at DUMC and 3 that had been transplanted elsewhere but followed longitudinally at DUMC for most of their posttransplantation course. A majority of the 35 patients who died succumbed to viral infections present at the time of transplantation, but none died of GVHD. At the time of this report, patients ranged from 6 months to 25.6 years after transplantation, with 68 of them being more than 10 years after transplantation. Sixty of these surviving patients had X-SCID resulting from mutations in the gene encoding the common gamma-chain (γcDef),8  11 had mutations in the gene encoding Janus kinase 3 (Jak3Def),9  15 had IL-7R α-chain mutations (IL7RαDef),10  18 had ADA deficiency (ADADef), and 24 had mutations in other genes, including RAG-1 or RAG-2 (RAG1/2Def; N = 6),11  the genes encoding the various chains of CD3 (CD3Def; N = 4)12  the CD45 gene (N = 1),13  the Artemis gene (N = 1),14  autosomal recessive (AutoRec) SCID of unknown molecular type (N = 11), and one male SCID of unknown type (Table 1). Donor BM was depleted of T cells by agglutination with soybean lectin and 2 cycles of rosetting with sheep erythrocytes that had been treated with aminoethylisothiuronium bromide as previously described.3  The method of T-cell depletion was the same for all recipients over the more than 25 years of this study. Forty-three of these patients were treated with a nonablative booster BMT, and the outcome of the booster transplantations is described in a separate manuscript (R.H.B., manuscript in preparation). Four of the patients underwent gene therapy elsewhere (2 with ADA deficiency in Italy, 2 with γcDef SCID at the National Institutes of Health). Gene therapy was successful in 3 of the 4 but unsuccessful in one γcDef patient who subsequently received a matched unrelated donor (MUD) transplantation after reduced intensity conditioning. Four ADA-deficient patients are currently receiving polyethylene glycol-modified bovine adenosine-deaminase (PEG-ADA), and 1 received a MUD BMT elsewhere. One Artemis-deficient patient received a MUD BMT elsewhere.

Chimerism

Chimerism was detected by karyotyping; fluorescence in situ hybridization15 ; restriction fragment length polymorphism, HLA typing; flow cytometric detection of γc or CD45 on lymphocytes; or by the presence of ADA.

T-cell phenotype and mitogen/antigen stimulation

T-cell phenotypes were determined by 4-color immunofluorescent staining and flow cytometry on freshly isolated PBMCs or whole blood with labeled antibodies (Abs) to CD3, CD4, CD8, CD14, CD16, CD20, CD45, CD132, CD56 (clone NCAM16.2), TCRαβ (clone T10B9.1A-31), and TCRγδ (clone B1) purchased from BD Biosciences. Lymphocyte proliferation was assessed by measuring [3H] thymidine incorporation into PBMCs after culture with optimal concentrations of the indicated stimuli, as previously described.3 

Real-time quantitative PCR assay for TRECs

Polymerase chain reaction (PCR) analysis of TCR episomes was performed as described elsewhere.5,16  Genomic DNA from unfractionated PBMCs was extracted using TRIzol (Invitrogen). Signal joint TCR rearrangement excision circles (sjTRECs) in PBMCs were quantified using real-time quantitative PCR with the 5′-nuclease assay with iCycler (Bio-Rad). PCR was performed using primers (5′cacatccctttcaaccatgct and 5′gccagctgcagggtttagg) and the probe FAM-acacctctggtttttgtaaaggtgcccactBHQ1 (IDT Integrated DNA Technologies). PCRs contained 0.5 U Platinum Taq polymerase (Invitrogen), 3.5 mM MgCl2, 0.2 mM deoxyribonucleoside triphosphates, 500 nmol/L of each primer, and 150 nmol/L probe. Amplification conditions were 95°C for 5 minutes, followed by 40 cycles of 95°C for 30 seconds and 60°C for 1 minute. Samples were analyzed in duplicate and corrected to reflect TREC/μg of PBMC DNA. The sjTREC values were calculated using a standard curve of TCR sjTREC plasmid DNA (107 to 102 molecules, generously provided by Dr Daniel Douek, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health). For consistency with previously published data from our group, results are reported as sjTREC per μg of PBMC DNA. The lower limit of confidence is 100 molecules of sjTREC per μg of PBMC DNA.

CDR3 size spectratyping

Spectratype analysis of the TCR Vβ chain used in these studies was performed as published.5,17  Briefly, total RNA was extracted from fresh or cryopreserved PBMC samples (2-10 × 106 cells/sample) using TRIzol and reverse transcribed to single-stranded cDNA with reverse transcriptase from avian myeloblastosis virus (Roche Diagnostics) using an oligo (dT) primer according to the manufacturer's protocol (Promega). The newly synthesized cDNA was then used as a template for 23 PCRs with 23 different unlabeled TCR Vβ- and 1 Cβ-specific primers. PCR amplifications were conducted to saturation (40 cycles). The products were visualized by an elongation reaction using a nested fluorescent Cβ primer. The elongation products, which contained the CDR3, were separated on a capillary system.

The results were analyzed using Gene Scan software (Applied Biosystems) and are shown as graphic distributions of size peaks centered on a peak corresponding to a CDR3 of 10 amino acids. The peaks are spaced by 3 nucleotides. Normal distributions of size peaks were obtained in our laboratory using RNA extracted from as few as 5 × 104 normal donor PBMCs. Comparison with size standards shows that the peaks correspond to in-frame transcripts. Normal profiles have an average of 6 to 10 peaks/Vβ family. Previously, we classified TCR Vβ peak profiles as oligoclonal, polyclonal skewed, and polyclonal Gaussian.5  Here (supplemental Figure 1, available on the Blood website; see the Supplemental Materials link at the top of the online article), the Vβ density peak histograms were analyzed by a web-based automated program for the Kullback-Leibler divergence (DKL).18  The higher the DKL, the greater the divergence from a normal Gaussian profile and the more oligoclonal the distribution of peaks.19 

Statistical analysis

Individual comparisons of the mean number of T, B, and NK cells at the latest transplantation date between pairs of molecular types of SCIDs and with normal controls were performed using a 2-sided Satterthwaite test.20  Because there were 7 molecular types of SCID, a total of 21 pairwise comparisons were performed for each cell type. To maintain an overall type 1 error rate near 0.1, a conservative Bonferroni21  0.005 (∼ 0.1/21) α level was used for all the pairwise comparisons; that is, only those pairwise comparisons with P values less than .005 were considered statistically significant. For pairwise comparisons of the T-cell proliferation at the latest transplantation date between pairs of molecular types of SCIDs and with normal controls, 7 pairwise comparisons were performed. To maintain an overall type 1 error rate near 0.1, a conservative Bonferroni 0.015 (∼ 0.1/7) α level was used for all these pairwise comparisons. In the analysis of TREC values, proliferation values, and T, B, and NK-cell counts over time after transplantation, the longitudinal change of these values over time was estimated after the effects between patients and transplantations per patient were removed from the model. For the analysis of the TREC values, 95% confidence intervals were placed around the predicted TREC line over time for times more than 1 year after transplantation. The prediction line is the line through the mean TREC value/mean time point with a slope equal to the predicted change in TREC over the time after transplantation.

Chimerism

Table 1 shows the results of chimerism studies in the various molecular types of SCID after BMT. Ten patients did not achieve T-cell chimerism after transplantation. Seven of these were ADADef, 2 of whom later received successful gene therapy elsewhere, one received a BMT elsewhere, and 4 are currently on PEG-ADA. One was an AutoRec SCID, one a CD3ζ-chain–deficient SCID, and one was a γcDef SCID who received unsuccessful gene therapy and a subsequent MUD BMT elsewhere. Forty-one of the patients achieved B-cell chimerism, including one-third of the γcDef patients, although they did not receive pretransplantation chemotherapy. The clinically relevant data related to the patient population studied in this paper are reported in a parallel manuscript (M. D. Railey, Y. Lokhnygina, and R.H.B., manuscript submitted, January 2009), which also contains the Kaplan-Meier survival curve for the transplantation recipients.

Table 1

Characterization of SCID patients studied

SCID typeNumber of patients
Tested for TRECsPositive for TRECsT-cell chimerismB-cell chimerismOn IVIG
γc Def (N = 60) 57 47 59 20 40 
ADADef (N = 18) 13 11 11 10 
IL-7RαDef (N = 15) 15 14 15 
Jak3 Def (N = 11) 10 10 11 
Artemis (N = 1) 
RAG1/2Def (N = 6) 
Unknown (N = 1) 
CD3 Def (N = 4) 
CD45 Def (N = 1) 
AutoRec (N = 11) 10 
Totals (N = 128) 115 93 118 41 70 
SCID typeNumber of patients
Tested for TRECsPositive for TRECsT-cell chimerismB-cell chimerismOn IVIG
γc Def (N = 60) 57 47 59 20 40 
ADADef (N = 18) 13 11 11 10 
IL-7RαDef (N = 15) 15 14 15 
Jak3 Def (N = 11) 10 10 11 
Artemis (N = 1) 
RAG1/2Def (N = 6) 
Unknown (N = 1) 
CD3 Def (N = 4) 
CD45 Def (N = 1) 
AutoRec (N = 11) 10 
Totals (N = 128) 115 93 118 41 70 

Blood samples were obtained from healthy normal controls and from the patients before transplantation and at various intervals thereafter. Excess PBMCs were cryopreserved in RPMI 1640 medium containing dimethyl sulfoxide and 20% fetal bovine serum.3  All of the flow cytometric and proliferative assays were performed using fresh cells, whereas the TREC and spectratype assays, not available at the beginning of these studies, were performed using cryopreserved cells. All studies were performed with the approval of the DUMC Institutional Review Board and with the written informed consent of the patients or their parents in accordance with the Declaration of Helsinki.

Lymphocyte phenotypes and T-cell function before and at latest time tested after transplantation

Lymphocyte phenotypes.

Patients with the various genetic types of SCID had distinct lymphocyte phenotypes before transplantation (Figure 1A).2  All patients had a profound deficiency of T cells and, when T cells were present, they were usually transplacentally transferred maternal T cells. In one Jak3Def patient, there were 8268 circulating maternal T cells/mm3 at presentation. At the latest time tested after transplantation, T-cell values (Figure 1B) were significantly (P < .001) higher than pretransplantation values in all molecular types of SCID. Before transplantation (Figure 1A), B cells were elevated in all categories except ADA and RAG deficiencies but were most elevated in γcDef and Jak3Def SCIDs (P < .001). At the latest time evaluated after transplantation (Figure 1A-B), B-cell values were significantly (P < .001) smaller than pretransplantation values and posttransplantation NK-cell values were significantly (P < .001) smaller than pretransplantation values. Given the sample sizes and variances of the patients and control groups, the mean numbers of T, B, and NK cells at the latest date after transplantation were different between some of the 7 SCID types and normal control values (Figure 1B), as determined by a conservative Bonferroni α level analysis for all of the pairwise comparisons.

Figure 1

PBMC phenotype before and after BMT in SCID patients. Mean numbers (± SEM) of CD20 B, CD3 T, and CD16 NK cells were obtained by flow cytometric analysis of SCID PBMCs studied before (A; N = 151) and at the latest time after transplantation (B; N = 120). Results from 2 patients with Artemis deficiency (1 surviving), 1 with CD45 deficiency, 4 with SCID of unknown molecular type (1 surviving), and from the 5 patients who received transplants elsewhere for whom we do not have the original pretransplantation data, are not included in Figures 1 and 2. Values for normal infant (A) and pediatric to young adult (B) controls are shown for comparison. Using a conservative Bonferroni α level analysis for all of the pairwise comparisons, given the sample sizes and variances of the patients and control groups, differences in mean T-cell numbers (B) were found between γcDef, ADADef, IL7RαDef, RAG1/2Def, CD3Def, AutoRec, and the normal control range (P < .015); differences in mean B-cell numbers (B) were found between ADADef (P = .001) and RAG1/2Def (P < .001) and the normal control range; differences in mean NK-cell numbers (B) were found between γcDef (P < .001), ADADef (P < .001), IL7RαDef (P = .002), RAG1/2Def (P < .001), AutoRec (P = .001), and the normal control values.

Figure 1

PBMC phenotype before and after BMT in SCID patients. Mean numbers (± SEM) of CD20 B, CD3 T, and CD16 NK cells were obtained by flow cytometric analysis of SCID PBMCs studied before (A; N = 151) and at the latest time after transplantation (B; N = 120). Results from 2 patients with Artemis deficiency (1 surviving), 1 with CD45 deficiency, 4 with SCID of unknown molecular type (1 surviving), and from the 5 patients who received transplants elsewhere for whom we do not have the original pretransplantation data, are not included in Figures 1 and 2. Values for normal infant (A) and pediatric to young adult (B) controls are shown for comparison. Using a conservative Bonferroni α level analysis for all of the pairwise comparisons, given the sample sizes and variances of the patients and control groups, differences in mean T-cell numbers (B) were found between γcDef, ADADef, IL7RαDef, RAG1/2Def, CD3Def, AutoRec, and the normal control range (P < .015); differences in mean B-cell numbers (B) were found between ADADef (P = .001) and RAG1/2Def (P < .001) and the normal control range; differences in mean NK-cell numbers (B) were found between γcDef (P < .001), ADADef (P < .001), IL7RαDef (P = .002), RAG1/2Def (P < .001), AutoRec (P = .001), and the normal control values.

Close modal

T-cell function.

Figure 2 shows in vitro responses to stimulation with phytohemagglutinin (PHA), concanavalin A (ConA), and pokeweed mitogen (PWM) by PBMCs from SCID patients according to their various molecular types, analyzed before (Figure 2A) and at the latest time after (Figure 2B) transplantation, compared with responses from normal controls. For responses to all 3 mitogenic stimuli, there was a significant difference (P < .001) between pretransplantation and posttransplantation proliferation. When analyzing posttransplantation proliferative responses to PHA and ConA using a conservative Bonferroni α level analysis for all of the pairwise comparisons, given the sample sizes and variances of the patients and control groups, there was a significant difference between some of the SCID molecular types and normal control values (Figure 2B). Finally, whereas T cells from all SCID molecular types responded poorly to allogeneic cells before transplantation, posttransplantation T cells from all SCIDs responded normally to allogeneic cells, tetanus antigens (data not shown), and Candida (Figure 2D).

Figure 2

PBMC function before and after BMT in SCID patients. [3H]Thymidine incorporation by proliferating lymphocytes in response to PHA, ConA, and PWM was evaluated before (A; N = 151) and at latest time studied after transplantation (B; N = 120). Values are mean responses in counts per minute (cpm; ± SEM). Using a conservative Bonferroni α level analysis for all of the pairwise comparisons, given the sample sizes and variances of the patients and control groups at the latest transplantation date (B), the proliferative response to PHA of γcDef, ADADef, RAG1/2Def SCIDs was different (P < .015) from that of normal controls. Mean proliferation to ConA stimulus was different from normal controls only for ADADef (P = .005), and for the response to PWM only the IL-7RαDef was different from normal control values (P < .001).

Figure 2

PBMC function before and after BMT in SCID patients. [3H]Thymidine incorporation by proliferating lymphocytes in response to PHA, ConA, and PWM was evaluated before (A; N = 151) and at latest time studied after transplantation (B; N = 120). Values are mean responses in counts per minute (cpm; ± SEM). Using a conservative Bonferroni α level analysis for all of the pairwise comparisons, given the sample sizes and variances of the patients and control groups at the latest transplantation date (B), the proliferative response to PHA of γcDef, ADADef, RAG1/2Def SCIDs was different (P < .015) from that of normal controls. Mean proliferation to ConA stimulus was different from normal controls only for ADADef (P = .005), and for the response to PWM only the IL-7RαDef was different from normal control values (P < .001).

Close modal

Longitudinal analysis of T-cell function after BMT in all SCIDs

The data presented in Figure 2 represent a cross-sectional analysis of T-cell function in SCID PBMCs before and at the latest time after transplantation. In this section, PBMCs from 128 SCID patients who received BMT were also analyzed over time for their ability to proliferate to PHA, ConA, and PWM. A total of 6299 proliferative response results were collected on the 128 patients with the 3 stimuli over time (Figure 3). Because the longitudinal analysis was performed on each patient, it was necessary to remove patient-to-patient variation from the data before testing the changes in proliferation over time. In addition, changes over time were significantly different for proliferation observed before and 1 year after transplantation, when T-cell function is established. We analyzed the period 1 to 25.07 years after transplantation. The estimated change in the log10 proliferation value per year for the period 1 to 25.07 years after transplantation for the PHA stimulation was −0.0162 with an SD of 0.0043 with a P = .001 (Figure 3A), for the ConA response was −0.0187 with an SD of 0.0043 and a P < .001 (Figure 3B), for the Candida response the estimated change per year was −0.0136 with an SD of 0.0054 and a P = .011 (Figure 3D), whereas the change for the PWM response over time was not statistically significant with a P = .636 (Figure 3C).

Figure 3

Analysis of T-cell function over time after BMT in all SCIDs. Proliferation values (cpm; N = 6299) to PHA (A), ConA (B), PWM (C), and Candida (D) stimulations of PBMCs from 128 patients over time. Graphs represent the change in the log10 proliferation for PHA, ConA, and PWM stimulations, respectively, over time after the patient and transplantation effects were removed. Means for normal controls: PHA 187 073 cpm, SEM 2390 cpm; Con A 137 050 cpm, SEM 2289 cpm; PWM 92 191 cpm, SEM 2156 cpm; and Candida 48 793 cpm, SEM 2835 cpm.

Figure 3

Analysis of T-cell function over time after BMT in all SCIDs. Proliferation values (cpm; N = 6299) to PHA (A), ConA (B), PWM (C), and Candida (D) stimulations of PBMCs from 128 patients over time. Graphs represent the change in the log10 proliferation for PHA, ConA, and PWM stimulations, respectively, over time after the patient and transplantation effects were removed. Means for normal controls: PHA 187 073 cpm, SEM 2390 cpm; Con A 137 050 cpm, SEM 2289 cpm; PWM 92 191 cpm, SEM 2156 cpm; and Candida 48 793 cpm, SEM 2835 cpm.

Close modal

Analysis of thymic function over time after BMT in all SCIDs

TREC values were determined over time in 115 of 128 total subjects with 11 different molecular types of SCID as a measure of thymic function. A total of 597 TREC observations were collected on the 115 patients over time. Thirty-one of the 115 patients had more than one transplantation. Because the longitudinal analysis was performed on each patient for each transplantation, it was necessary to remove patient-to-patient and transplantation-to-transplantation variation from the data before analyzing the changes in TREC over time on each patient. In addition, changes over time were significantly different for TREC observations collected less than 1 year after transplantation and greater than 1 year after transplantation. The main emphasis was to examine long-term changes in TREC values over time, after T-cell function was fully established (after 1 year after transplantation). Therefore, only the results associated with the period from 1 to 25.07 years after transplantation were analyzed. Figure 4A provides a graph of the log10 change in TREC values with time after transplantation for all molecular types of SCID, after the patient/transplantation variation was removed from the model. The change in the log10 TREC value per year for the period from 1 to 25.07 years after transplantation was −0.03155 with an SD of 0.0086 and P = .001. The solid trend-line passes through the mean years after transplantation for the combined data for all SCID molecular types, with the estimated slope of −0.03155. This TREC value change over time is within the value change in the normal population for the period 1 to 25 years of age (−0.033).4 

Figure 4

Analysis of thymic function over time after BMT in all SCIDs. TREC values (N = 597) were observed over time in 115 patients for whom samples were available for analysis, each patient associated with 1 of 11 molecular types of SCID. (A) The combined results of all 115 patients. The points on the figure represent the predicted TREC value plus the residual after the patient and transplantation effects were removed. Lighter lines are then used to join points from the same patient before and 1 year after transplantation. The same analysis was performed for 15 IL7RαDef patients (B), 10 Jak3Def patients (C), and 57 γcDef patients (D); 95% confidence intervals were placed around the predicted TREC line over time for times more than 1 year after transplantation.

Figure 4

Analysis of thymic function over time after BMT in all SCIDs. TREC values (N = 597) were observed over time in 115 patients for whom samples were available for analysis, each patient associated with 1 of 11 molecular types of SCID. (A) The combined results of all 115 patients. The points on the figure represent the predicted TREC value plus the residual after the patient and transplantation effects were removed. Lighter lines are then used to join points from the same patient before and 1 year after transplantation. The same analysis was performed for 15 IL7RαDef patients (B), 10 Jak3Def patients (C), and 57 γcDef patients (D); 95% confidence intervals were placed around the predicted TREC line over time for times more than 1 year after transplantation.

Close modal

We were able to perform a follow-up study (Figure S2) on 3 of the same patients whose data we published previously.5  All 3 patients demonstrated a sustained thymic function (4-3 log10 TREC/μg DNA) beyond the time points previously reported.

Analysis of thymic function in 11 molecular types of SCIDs over time after BMT

Different cellular microenvironments and cell-cell interactions may develop in SCID of different molecular types, which could differentially affect the maintenance of thymic function over time. Therefore, we evaluated thymic function after BMT among the various molecular types of SCID. PBMCs from a total of 57 γcDef, 15 IL-7RαDef, 10 Jak3 Def, 13 ADADef, and 20 patients with other types of SCID (RAG1/2Def, CD3δDef, CD3ϵDef, CD3ζDef, CD45Def, AutoRec, and unknown) were subjected to TREC analysis. For all SCID molecular types except γcDef, TREC values measured within 20 years and beyond the first year of BMT remained constant. Figure 4 provides examples of these constant values over time, demonstrated by a horizontal trendline for IL7RαDef (Figure 4B) and Jak3Def SCID (Figure 4C) molecular types, respectively. For the γcDef SCIDs (Figure 4D), the change in the log10 TREC value per year for the period 1 to 21.91 years after transplantation was −0.0471 with an SD of 0.0099 and a P < .001. This significant negative TREC value change over time for γcDef is represented by a sloping trendline (Figure 4D), which was not statistically different (P = .078) from the slope of the trendline in the normal population for the period 1 to 25 years of age (−0.033).4 

Ten or more years after transplantation: thymic output, T-cell phenotype, and function in BMT recipients

The results reported so far refer to composite groups of patients from the same molecular type of SCID or from the same in vitro stimulation. We next evaluated 68 individual SCID patients (Table 2) who were more than 10 years after transplantation, to determine whether, at the latest date after transplantation, they generated peripheral T cells with normal phenotype and function. These patients were monitored for their TREC levels, their naive versus memory phenotype (percentage of CD3+ T cells that expressed the CD45RA marker vs those that expressed the CD45RO marker22 ), and/or their ability to proliferate in response to stimulation with mitogens. For the majority (83%) of the patients, thymic output correlated well with T-cell phenotype and function. Only 3 of 53 patients failed to demonstrate thymic output and had poor T-cell function (patients 5, 35, and 50 in Table 2). A fraction of the 53 patients (N = 9: patients 10, 11, 17, 22, 34, 38, 43, 56, and 57) had negative TREC results, but normal T-cell phenotype and function.

Table 2

SCID patients 10 or more years after transplantation

Patient no.SexRaceSCID typeTreatment typeNo. of treatmentsTime after treatment, yAge at treatment, dTRECALC*CD3+ CD45RA+CD3+ CD45RO+RA/RO ratioMediumPHACon APWM
ADADef Haplo 23.2 70 ND ND 13.3 69.1 0.1925 616 209 258 119 595 135 999 
ADADef Haplo 21 11 ND 1480 26.8 50.2 0.5339 4849 157 166 142 720 72 380 
ADADef Ident 20.1 127 224 1071 29.5 46.8 0.6303 427 61 016 48 611 72 426 
ADADef Haplo 19.5 112 ND 659 ND 50.6 ND 4460 40 447 48 784 81 243 
ADADef Ident 15.5 50 < 100 2015 22.7 39.1 0.5806 155 36 416 37 228 65 954 
ADADef Haplo 12.8 162 930 1334 29.5 32.3 0.9133 530 225 180 167 360 90 433 
ADADef Ident 12.5 97 409 1175 13.2 27.8 0.4748 464 25 957 69 912 52 802 
ADADef Ident 12.2 398 8180 1890 51.6 18.6 2.7742 549 265 898 224 603 168 812 
ADADef Haplo 11.1 182 ND 268 10 69.5 0.1439 113 50 021 32 754 33 224 
10 ADADef Haplo 10.8 97 < 100 2987 48.9 19.4 2.5206 385 126 886 87 006 45 768 
11 AutoRec Haplo 25.5 21 < 100 456 8.4 62.3 0.1348 242 80 334 104 525 87 007 
12 AutoRec Ident 25.4 303 ND 4000 54.4 33.7 1.6142 ND 232 340 197 013 134 957 
13 AutoRec Haplo 18.2 152 292 1802 49.6 26.5 1.8717 80 85 008 88 208 14 140 
14 AutoRec Haplo 16.3 127 ND 2220 ND ND ND 1017 136 469 197 957 203 740 
15 AutoRec Haplo 16.1 155 ND 1404 42.4 43.5 0.9747 738 232 237 152 777 128 081 
16 AutoRec Haplo 10.2 147 ND 960 ND ND ND 149 24 056 18 881 ND 
17 CD3eDef Haplo 19.4 171 < 100 1100 ND 5.9 ND 189 94 662 81 837 109 829 
18 CD3dDef Ident 11.9 222 450 1127 21.1 54 0.3907 169 84 202 48 974 35 092 
19 IL-7RDef Haplo 18.6 164 906 1224 53.1 23.6 2.25 327 242 178 143 809 129 342 
20 IL-7RDef Ident 16.2 285 3531 2214 68.7 19.4 3.5412 1607 182 517 221 894 125 406 
21 IL-7RDef Haplo 14.1 509 1750 1950 56.6 21.8 2.5963 255 162 613 130 055 92 337 
22 IL-7RDef Haplo 13.9 449 < 100 947 27.9 36.3 0.7686 420 108 680 96 460 158 800 
23 IL-7RDef Haplo 11.8 325 736 875 21.1 44.5 0.4742 63 157 564 142 408 100 806 
24 IL-7RDef Haplo 10.4 101 2120 1554 86.1 5.9 14.593 1298 123 639 110 257 35 732 
25 Jak3Def Haplo 24.8 177 ND ND 67 31.5 2.127 ND 243 707 420 456 138 425 
26 Jak3Def Haplo 23.1 238 126 1152 22.9 40.7 0.5627 785 260 860 186 940 192 166 
27 Jak3Def Haplo 18.4 263 828 1330 49.1 15.5 3.1677 942 76 229 48 674 27 226 
28 Jak3Def Ident 18.3 222 7360 4368 37.5 30.3 1.2376 307 170 674 125 659 91 353 
29 Jak3Def Haplo 16.8 25 485 896 20.4 36.1 0.5651 178 193 925 130 763 72 557 
30 Jak3Def Ident 16.2 290 326 2464 45.3 27.8 1.6295 669 151 044 112 650 104 931 
31 Jak3Def Haplo 14.3 21 1326 759 16 34.1 0.4692 531 133 790 135 770 114 156 
32 Jak3Def Haplo 12.7 334 2740 8195 77.6 9.8 7.9184 847 203 595 235 347 149 399 
33 Jak3Def Haplo 11.4 24 2722 2580 38.1 29.8 1.2785 251 115 436 85 833 27 530 
34 RAG1Def Ident 25.6 128 < 100 1180 21.2 37.2 0.5699 206 96 892 89 239 35 968 
35 RAG2Def Haplo 20 119 < 100 484 2.3 76.2 0.0302 166 23 457 20 513 ND 
36 Unknown Haplo 14.9 122 4060 ND 54.2 21.9 2.4749 305 207 770 144 453 107 274 
37 X-linked Haplo 24 175 100 957 4.9 18.4 0.2663 167 61 334 65 440 72 004 
38 X-linked Haplo 23.5 113 < 100 1311 30.9 72.3 0.4274 377 67 068 98 899 55 317 
39 X-linked Haplo 22.9 45 874 3356 9.8 82 0.1195 84 29 118 8223 10 121 
40 X-linked Haplo 22.5 20 292 2080 4.5 53.1 0.0847 303 124 335 175 970 109 327 
41 X-linked Haplo 21.4 105 4130 1806 37.4 28.2 1.3262 781 174 485 121 500 157 009 
42 X-linked Haplo 21.3 212 7550 2135 74.5 12.9 5.7752 497 258 225 89 614 84 634 
43 X-linked Haplo 20.7 217 < 100 1652 37.4 21.3 1.7559 124 46 406 90 702 66 304 
44 X-linked Haplo 19.6 516 3310 ND 67.7 13.7 4.9416 164 157 155 117 361 85 508 
45 X-linked Haplo 19.6 222 4699 ND 94.9 18 5.2722 ND 223 750 207 796 122 969 
46 X-linked Haplo 19.1 16 319 1085 34 20.6 1.6505 84 42 182 92 148 84 233 
47 X-linked Haplo 18.7 370 ND 1812 80.2 18.8 4.266 1293 260 082 265 320 149 143 
48 X-linked Ident 17 7050 2346 61.3 15.7 3.9045 128 137 196 122 384 80 470 
49 X-linked Ident 16.9 13 8970 1216 62.3 16.7 3.7305 68 122 949 124 230 62 856 
50 X-linked Haplo 16.1 175 < 100 3172 15.8 51.4 0.3074 84 13 794 4389 3458 
51 X-linked MUD 15.3 288 546 ND 32.1 34.4 0.9331 2592 109 762 120 866 80 358 
52 X-linked Haplo 14.9 289 ND 334 2.3 87.4 0.0263 266 22 024 17 387 9055 
53 X-linked Haplo 14.8 245 219 2112 55.4 14.5 3.8207 128 166 461 154 041 85 759 
54 X-linked Haplo 13.2 193 1366 2477 69.7 20.7 3.3671 501 287 040 224 342 11 685 
55 X-linked Haplo 13 367 ND 760 5.9 65.5 0.0901 81 13 541 20 565 25 583 
56 X-linked Haplo 12.7 351 < 100 1125 11.8 60.8 0.1941 1092 47 920 77 417 66 276 
57 X-linked Haplo 12.5 10 < 100 450 32.6 29.8 1.094 283 121 920 100 630 69 591 
58 X-linked Haplo 12.5 10 19 700 2580 65.4 14.5 4.5103 148 169 103 179 351 125 049 
59 X-linked Haplo 12.4 474 1170 58 12.7 4.5669 337 144 226 136 228 78 777 
60 X-linked Haplo 12.4 187 ND 2157 89.7 3.8 23.605 5091 225 611 270 960 241 576 
61 X-linked Haplo 12.1 163 7540 3276 61.9 8.6 7.1977 103 163 235 126 186 115 099 
62 X-linked Haplo 11.9 163 ND 3894 26.5 46.3 0.5724 ND ND ND ND 
63 X-linked Haplo 11.5 ND 2655 67 11.5 5.8261 3659 293 085 179 415 135 615 
64 X-linked Haplo 11.5 12 426 5472 86.8 3.8 22.842 654 229 229 231 768 88 773 
65 X-linked Haplo 11.4 112 560 1170 18.5 40.8 0.4534 1440 141 813 109 926 74 859 
66 X-linked Haplo 10.8 163 294 1768 13.8 33 0.4182 62 63 058 103 493 15 176 
67 X-linked Haplo 10.7 282 263 3354 27.5 33.6 0.8185 147 141 199 108 038 98 544 
68 X-linked Haplo 10 213 301 1040 39.5 27.6 1.4312 489 105 635 110 181 97 429 
Patient no.SexRaceSCID typeTreatment typeNo. of treatmentsTime after treatment, yAge at treatment, dTRECALC*CD3+ CD45RA+CD3+ CD45RO+RA/RO ratioMediumPHACon APWM
ADADef Haplo 23.2 70 ND ND 13.3 69.1 0.1925 616 209 258 119 595 135 999 
ADADef Haplo 21 11 ND 1480 26.8 50.2 0.5339 4849 157 166 142 720 72 380 
ADADef Ident 20.1 127 224 1071 29.5 46.8 0.6303 427 61 016 48 611 72 426 
ADADef Haplo 19.5 112 ND 659 ND 50.6 ND 4460 40 447 48 784 81 243 
ADADef Ident 15.5 50 < 100 2015 22.7 39.1 0.5806 155 36 416 37 228 65 954 
ADADef Haplo 12.8 162 930 1334 29.5 32.3 0.9133 530 225 180 167 360 90 433 
ADADef Ident 12.5 97 409 1175 13.2 27.8 0.4748 464 25 957 69 912 52 802 
ADADef Ident 12.2 398 8180 1890 51.6 18.6 2.7742 549 265 898 224 603 168 812 
ADADef Haplo 11.1 182 ND 268 10 69.5 0.1439 113 50 021 32 754 33 224 
10 ADADef Haplo 10.8 97 < 100 2987 48.9 19.4 2.5206 385 126 886 87 006 45 768 
11 AutoRec Haplo 25.5 21 < 100 456 8.4 62.3 0.1348 242 80 334 104 525 87 007 
12 AutoRec Ident 25.4 303 ND 4000 54.4 33.7 1.6142 ND 232 340 197 013 134 957 
13 AutoRec Haplo 18.2 152 292 1802 49.6 26.5 1.8717 80 85 008 88 208 14 140 
14 AutoRec Haplo 16.3 127 ND 2220 ND ND ND 1017 136 469 197 957 203 740 
15 AutoRec Haplo 16.1 155 ND 1404 42.4 43.5 0.9747 738 232 237 152 777 128 081 
16 AutoRec Haplo 10.2 147 ND 960 ND ND ND 149 24 056 18 881 ND 
17 CD3eDef Haplo 19.4 171 < 100 1100 ND 5.9 ND 189 94 662 81 837 109 829 
18 CD3dDef Ident 11.9 222 450 1127 21.1 54 0.3907 169 84 202 48 974 35 092 
19 IL-7RDef Haplo 18.6 164 906 1224 53.1 23.6 2.25 327 242 178 143 809 129 342 
20 IL-7RDef Ident 16.2 285 3531 2214 68.7 19.4 3.5412 1607 182 517 221 894 125 406 
21 IL-7RDef Haplo 14.1 509 1750 1950 56.6 21.8 2.5963 255 162 613 130 055 92 337 
22 IL-7RDef Haplo 13.9 449 < 100 947 27.9 36.3 0.7686 420 108 680 96 460 158 800 
23 IL-7RDef Haplo 11.8 325 736 875 21.1 44.5 0.4742 63 157 564 142 408 100 806 
24 IL-7RDef Haplo 10.4 101 2120 1554 86.1 5.9 14.593 1298 123 639 110 257 35 732 
25 Jak3Def Haplo 24.8 177 ND ND 67 31.5 2.127 ND 243 707 420 456 138 425 
26 Jak3Def Haplo 23.1 238 126 1152 22.9 40.7 0.5627 785 260 860 186 940 192 166 
27 Jak3Def Haplo 18.4 263 828 1330 49.1 15.5 3.1677 942 76 229 48 674 27 226 
28 Jak3Def Ident 18.3 222 7360 4368 37.5 30.3 1.2376 307 170 674 125 659 91 353 
29 Jak3Def Haplo 16.8 25 485 896 20.4 36.1 0.5651 178 193 925 130 763 72 557 
30 Jak3Def Ident 16.2 290 326 2464 45.3 27.8 1.6295 669 151 044 112 650 104 931 
31 Jak3Def Haplo 14.3 21 1326 759 16 34.1 0.4692 531 133 790 135 770 114 156 
32 Jak3Def Haplo 12.7 334 2740 8195 77.6 9.8 7.9184 847 203 595 235 347 149 399 
33 Jak3Def Haplo 11.4 24 2722 2580 38.1 29.8 1.2785 251 115 436 85 833 27 530 
34 RAG1Def Ident 25.6 128 < 100 1180 21.2 37.2 0.5699 206 96 892 89 239 35 968 
35 RAG2Def Haplo 20 119 < 100 484 2.3 76.2 0.0302 166 23 457 20 513 ND 
36 Unknown Haplo 14.9 122 4060 ND 54.2 21.9 2.4749 305 207 770 144 453 107 274 
37 X-linked Haplo 24 175 100 957 4.9 18.4 0.2663 167 61 334 65 440 72 004 
38 X-linked Haplo 23.5 113 < 100 1311 30.9 72.3 0.4274 377 67 068 98 899 55 317 
39 X-linked Haplo 22.9 45 874 3356 9.8 82 0.1195 84 29 118 8223 10 121 
40 X-linked Haplo 22.5 20 292 2080 4.5 53.1 0.0847 303 124 335 175 970 109 327 
41 X-linked Haplo 21.4 105 4130 1806 37.4 28.2 1.3262 781 174 485 121 500 157 009 
42 X-linked Haplo 21.3 212 7550 2135 74.5 12.9 5.7752 497 258 225 89 614 84 634 
43 X-linked Haplo 20.7 217 < 100 1652 37.4 21.3 1.7559 124 46 406 90 702 66 304 
44 X-linked Haplo 19.6 516 3310 ND 67.7 13.7 4.9416 164 157 155 117 361 85 508 
45 X-linked Haplo 19.6 222 4699 ND 94.9 18 5.2722 ND 223 750 207 796 122 969 
46 X-linked Haplo 19.1 16 319 1085 34 20.6 1.6505 84 42 182 92 148 84 233 
47 X-linked Haplo 18.7 370 ND 1812 80.2 18.8 4.266 1293 260 082 265 320 149 143 
48 X-linked Ident 17 7050 2346 61.3 15.7 3.9045 128 137 196 122 384 80 470 
49 X-linked Ident 16.9 13 8970 1216 62.3 16.7 3.7305 68 122 949 124 230 62 856 
50 X-linked Haplo 16.1 175 < 100 3172 15.8 51.4 0.3074 84 13 794 4389 3458 
51 X-linked MUD 15.3 288 546 ND 32.1 34.4 0.9331 2592 109 762 120 866 80 358 
52 X-linked Haplo 14.9 289 ND 334 2.3 87.4 0.0263 266 22 024 17 387 9055 
53 X-linked Haplo 14.8 245 219 2112 55.4 14.5 3.8207 128 166 461 154 041 85 759 
54 X-linked Haplo 13.2 193 1366 2477 69.7 20.7 3.3671 501 287 040 224 342 11 685 
55 X-linked Haplo 13 367 ND 760 5.9 65.5 0.0901 81 13 541 20 565 25 583 
56 X-linked Haplo 12.7 351 < 100 1125 11.8 60.8 0.1941 1092 47 920 77 417 66 276 
57 X-linked Haplo 12.5 10 < 100 450 32.6 29.8 1.094 283 121 920 100 630 69 591 
58 X-linked Haplo 12.5 10 19 700 2580 65.4 14.5 4.5103 148 169 103 179 351 125 049 
59 X-linked Haplo 12.4 474 1170 58 12.7 4.5669 337 144 226 136 228 78 777 
60 X-linked Haplo 12.4 187 ND 2157 89.7 3.8 23.605 5091 225 611 270 960 241 576 
61 X-linked Haplo 12.1 163 7540 3276 61.9 8.6 7.1977 103 163 235 126 186 115 099 
62 X-linked Haplo 11.9 163 ND 3894 26.5 46.3 0.5724 ND ND ND ND 
63 X-linked Haplo 11.5 ND 2655 67 11.5 5.8261 3659 293 085 179 415 135 615 
64 X-linked Haplo 11.5 12 426 5472 86.8 3.8 22.842 654 229 229 231 768 88 773 
65 X-linked Haplo 11.4 112 560 1170 18.5 40.8 0.4534 1440 141 813 109 926 74 859 
66 X-linked Haplo 10.8 163 294 1768 13.8 33 0.4182 62 63 058 103 493 15 176 
67 X-linked Haplo 10.7 282 263 3354 27.5 33.6 0.8185 147 141 199 108 038 98 544 
68 X-linked Haplo 10 213 301 1040 39.5 27.6 1.4312 489 105 635 110 181 97 429 

ND indicates not determined.

*

Values are expressed as cells/mm3.

Values are the percentage of total lymphocyte counts.

Values are cpm [3H]thymidine incorporation.

Analysis of the TCR repertoire in SCIDs followed long-term post-BMT

In previous published studies, we showed that a fully diverse TCR Vβ repertoire developed in BM-transplanted SCIDs at peak thymus output, whereas a more skewed repertoire emerged in the few patients studied more than 10 years after BMT.5  In this study, we analyzed the TCR Vβ repertoire of 36 SCID patients of various molecular types before and up to 25 years after transplantation. The DKL statistical program was used to assess the diversity of the TCR Vβ repertoire in these patients, as determined by spectratype analysis. Using this program, the higher the DKL, the greater the divergence from a normal Gaussian profile and the more oligoclonal the distribution of peaks (Figure S1).

As shown in Figure 5, the DKL values decreased from more than 0.3 within the first year of BMT, when the TCR repertoire is still oligoclonal, to a range between 0.1 and 0.03, indicating that patients developed and maintained a diverse TCR Vβ repertoire. The 4 patients who had higher than normal (> 0.08) DKL values beyond 10 years after BMT did not show evidence of thymic function.

Figure 5

Thymic output correlates with TCR diversity. Shown are dot plots of the DKL values compared with TREC levels of samples taken from 36 SCID patients at multiple time points after BMT. DKL values from normal controls were < 0.08. ○ represents patients whose TREC value was less than 100; ●, patients whose TREC value was > 100.

Figure 5

Thymic output correlates with TCR diversity. Shown are dot plots of the DKL values compared with TREC levels of samples taken from 36 SCID patients at multiple time points after BMT. DKL values from normal controls were < 0.08. ○ represents patients whose TREC value was less than 100; ●, patients whose TREC value was > 100.

Close modal

To examine the effect of DKL values on TREC values in these patients, we calculated the mean log10 TREC and DKL values per patient and analyzed these means through weighted least squares regression. Because of the small sample number, we did not evaluate the influence of DKL on TREC over time. The change in the log10 TREC value per DKL value was −4.728 with an SD of 1.607 with a P = .007, indicating a significant inverse correlation between DKL values and TREC values in these patients.

In this longitudinal study, we analyzed a total of 128 surviving SCID patients for thymic output and T-cell reconstitution over a period of up to more than 25 years after BMT. Our results show that T-cell reconstitution, including thymic output and T-cell diversity, are maintained long-term to levels similar or equal to normal control values.

T-cell chimerism was achieved in 92% of the transplanted patients, and 32% also developed B-cell chimerism, although these patients did not receive pretransplantation chemotherapy. The T-cell numbers after BMT were significantly greater than before transplantation, whereas the numbers of B and NK cells, which appeared elevated before BMT, significantly decreased to levels similar to normal controls at the latest time tested after transplantation. T-cell function, measured as proliferative responses to T-cell mitogens (PHA, ConA, PWM) was significantly increased in patients analyzed at their latest study after transplantation compared with responses before transplantation.

Ninety-three (81%) of the 115 patients analyzed (Table 1) developed detectable levels of TREC, and 14 of the patients who did not, received one or more booster BMT. The lack of thymic function in 5 SCIDs was attributed to the fact that they were tested at less than 1 year after BMT. Nine SCIDs who lacked TREC function are long-term survivors (> 10 years after BMT). One is RAG1/2Def, 4 are γcDef, 2 are ADADef, one is IL-7RαDef, and one is AutoRec recessive SCID of unknown molecular type, all of whom are in good health at times up to 25.6 years after transplantation. Those with ADA deficiency or with VDJ recombination defects have lower T-cell numbers, function and thymic output after transplantation than SCIDs of other molecular types. Whether this is the result of intrinsic thymic defects in these SCIDs or to postthymic influences is unknown. Defective double-negative thymocytes (in RAG1/2 Def SCIDs, for example) present in thymic stromal niches could compete for space with incoming progenitor T cells of donor origin derived from BMT as has been reported for RAG SCID mice.23  This could result in poor maturation of donor stem cells and insufficient peripheral T-cell reconstitution after BMT. By contrast, we did not see any evidence of defects in early or late thymopoiesis in the IL-7RαDef, γc-def SCIDs, or Jak3Def SCIDs, all of whose pretransplantation T-cell defects are caused by failure of IL-7 signaling. The ADA and RAG-deficient patients, although long-term survivors, are not as healthy as those with the other molecular types of SCID. It is possible that their inferior immune reconstitution or other consequences of their underlying molecular defects may result in even poorer general health over longer follow-up. Further investigation of this issue is forthcoming in our laboratory.

When an extensive TREC analysis was performed on 115 patients over time in the present study (Figure 4A), the estimated change in TREC values per year for the period of 1 year to more than 25 years after transplantation demonstrated a negative slope, which was within the level of change in slope for normal controls noted previously.4  TREC values change in the normal population between 4 and 3 log10 within the first 25 years of life. When the TREC analysis was performed separately for the various molecular types of SCID (Figure 4B,C), TREC values measured beyond the first year of BMT remained constant, except for γcDef, which demonstrated a significant negative slope (Figure 4D). However, this negative slope was not statistically different from the change in slope for normal controls noted previously.4 

The proliferative response to PWM over a period of 1 to more than 25 years of transplantation remained constant over time with responses to PHA and ConA producing a small but statistically significant deviation from a horizontal line. Studies by others have previously reported a decrease in proliferative responses of T cells to mitogens in the normal population over time.24 

Our group previously reported that thymic output appeared to decline at more than 10 years after BMT,4  a finding that has been frequently cited in the literature as a concern that nonablative marrow transplantations could not maintain sufficient T-cell function long-term to prevent infections.25,26  At the time of this earlier study,4  only a few of our patients were more than 10 years after transplantation. We now have many more (N = 68) patients beyond 10 years after transplantation; and, as shown in the present study, thymic output and T-cell function were retained in a majority (83%) of these patients. Patients were analyzed beyond 10 years of BMT for their thymic function, T-cell phenotype, and responsiveness. In 83% of the patients, the TREC values were positive and the CD45RA/RO ratio was more than 1 in 48% of the subjects, suggesting that many of the circulating T cells had a naive phenotype. Responses to mitogens were within or above normal ranges in 90% of the subjects. Interestingly, 3 of the patients who showed a decline in thymic function (J-1, J-2, and X-5) in our previous report5  were followed beyond 10 years of BMT in the present study and subsequently had improved thymic function. Therefore, the supposition that TREC function declines with time does not appear to be a general finding applicable to all SCIDs treated with nonablative BMT. The data in the present study also refute the hypothesis that there is an intrinsic inability of the SCID thymus to maintain thymic output beyond the first decade of life.

The TCR Vβ repertoire studies extended beyond 10 years of BMT also demonstrated that DKL values inversely correlated with TREC values (low DKL indicating polyclonality with high TREC values). The inverse association between TREC and DKL was statistically significant. For 4 patients who failed to develop TREC over time, the DKL values remained high, indicating oligoclonality. Failure to develop a diverse repertoire in those 4 patients was not associated with a specific molecular type.

Few studies have been reported to date on long-term T-cell reconstitution after hematopoietic stem cell transplantation (HSCT) in SCID patients.25-31  The findings described in the present study are in keeping with those of Borghans et al,27  who found that 11 of 19 SCID patients followed up to 25 years after conditioned BMT had no evidence for an accelerated decline of T-cell immunity or thymus output; however, those SCIDs who had low thymic output soon after transplantation continued to have decreased long-term T-cell reconstitution. Mazzolari et al25  reported on the long-term immune reconstitution and clinical outcome of 40 patients with severe T-cell immunodeficiency who survived for up to 16.3 years after HSCT. A total of 35% of the 40 patients had low TREC levels at their last follow-up, and oligoclonality of the T-cell repertoire was demonstrated at the last follow-up in 27.5% of the patients. Cavazzana-Calvo et al26  and Friedrich et al28  have reported findings similar to those of Mazzolari et al25  and speculated that the use of a conditioning regimen before HSCT for SCID was necessary for thymic output at greater than 10 years after transplantation. However, Patel et al30  have recently reported sustained T-cell function in 15 long-term SCID survivors (up to 26 years) of nonconditioned BMTs. Our data confirm that finding and provide evidence for continued thymic output at more than 10 years after transplantation in the absence of both pretransplantation conditioning and posttransplantation GVHD prophylaxis.

In conclusion, these results indicate that transplantation of HLA-identical or haploidentical allogeneic-related, T cell–depleted BM into SCID infants in the absence of pretransplantation chemotherapy or posttransplantation GVHD prophylaxis is successful at reconstituting long-term T-cell function. The ability of these patients to generate new T cells at these late time points is indirect evidence for stem cell engraftment.

An Inside Blood analysis of this article appears at the front of this issue.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors thank the patients and their families, the physicians who referred them and cared for them, the members of the Duke University DNA Analysis Facility, and the Duke Comprehensive Cancer Center Flow Cytometry Facility.

This work was supported by the National Institutes of Health (grants AI47605 and AI042951) and in part by the Duke Regional Biocontainment Laboratory (AI58607) and the Duke Center for Translational Research (AI51445).

National Institutes of Health

Contribution: M.S.-K. designed research, collected and analyzed data, and wrote the manuscript; C.M.W., R.E.P., and M.C. performed research and collected data; J.L.R. contributed research discussions and analyzed data; B.K.M. performed complete statistical analysis of data; G.D.S. contributed vital reagents and analytical tools; and R.H.B. designed research, collected and analyzed data, and wrote the manuscript.

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

Correspondence: Marcella Sarzotti-Kelsoe, Duke University Medical Center, 2812 Erwin Rd, Erwin Terrace II, Ste 301, Durham, NC 27710; e-mail: msarzott@duke.edu.

1
Buckley
 
RH
Schiff
 
SE
Schiff
 
RI
, et al. 
Hematopoietic stem cell transplantation for the treatment of severe combined immunodeficiency.
N Engl J Med
1999
, vol. 
340
 (pg. 
508
-
516
)
2
Buckley
 
RH
Molecular defects in human severe combined immunodeficiency and approaches to immune reconstitution.
Annu Rev Immunol
2004
, vol. 
22
 (pg. 
625
-
655
)
3
Buckley
 
RH
Schiff
 
SE
Sampson
 
HA
, et al. 
Development of immunity in human severe primary T cell deficiency following haploidentical bone marrow stem cell transplantation.
J Immunol
1986
, vol. 
136
 (pg. 
2398
-
2407
)
4
Patel
 
DD
Gooding
 
ME
Parrott
 
RE
, et al. 
Thymic function after hematopoietic stem-cell transplantation for the treatment of severe combined immunodeficiency.
N Engl J Med
2000
, vol. 
342
 (pg. 
1325
-
1332
)
5
Sarzotti
 
M
Patel
 
DD
Li
 
X
, et al. 
T cell repertoire development in humans with SCID after nonablative allogeneic marrow transplantation.
J Immunol
2003
, vol. 
170
 (pg. 
2711
-
2718
)
6
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
 (pg. 
690
-
695
)
7
Myers
 
LA
Patel
 
DD
Puck
 
JM
Buckley
 
RH
Hematopoietic stem cell transplantation for severe combined immunodeficiency in the neonatal period leads to superior thymic output and improved survival.
Blood
2002
, vol. 
99
 (pg. 
872
-
878
)
8
Noguchi
 
M
Yi
 
H
Rosenblatt
 
HM
, et al. 
Interleukin-2 receptor gamma chain mutation results in X-linked severe combined immunodeficiency in humans.
Cell
1993
, vol. 
73
 (pg. 
147
-
157
)
9
Roberts
 
JL
Lengi
 
A
Brown
 
SM
, et al. 
Janus kinase 3 (JAK3) deficiency: clinical, immunologic and molecular analyses of 10 patients and outcomes of stem cell transplantation.
Blood
2004
, vol. 
103
 (pg. 
209
-
218
)
10
Puel
 
A
Ziegler
 
SF
Buckley
 
RH
Leonard
 
WJ
Defective IL7R expression in T(-) B(+)NK(+) severe combined immunodeficiency.
Nat Genet
1998
, vol. 
20
 (pg. 
394
-
397
)
11
Schwarz
 
K
Gauss
 
GH
Ludwig
 
L
, et al. 
RAG mutations in human B cell-negative SCID.
Science
1996
, vol. 
274
 (pg. 
97
-
99
)
12
Buckley
 
RH
The multiple causes of human SCID.
J Clin Invest
2004
, vol. 
114
 (pg. 
1409
-
1411
)
13
Kung
 
C
Pingel
 
JT
Heikinheimo
 
M
, et al. 
Mutations in the tyrosine phosphatase CD45 gene in a child with severe combined immunodeficiency disease.
Nat Med
2000
, vol. 
6
 (pg. 
343
-
345
)
14
Moshous
 
D
Callebaut
 
I
de Chasseval
 
R
, et al. 
Artemis, a novel DNA double-strand break repair/V(D)J recombination protein, is mutated in human severe combined immune deficiency.
Cell
2001
, vol. 
105
 (pg. 
177
-
186
)
15
Van Den Berg
 
H
Vossen
 
JM
van den Bergh
 
RL
Bayer
 
J
van Tol
 
MJ
Detection of Y chromosome by in situ hybridization in combination with membrane antigens by two-color immunofluorescence.
Lab Invest
1994
, vol. 
64
 (pg. 
623
-
628
)
16
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
 (pg. 
1875
-
1881
)
17
Pannetier
 
C
Even
 
J
Kourilsky
 
P
T-cell repertoire diversity and clonal expansions in normal and clinical samples.
Immunol Today
1995
, vol. 
16
 (pg. 
176
-
181
)
18
He
 
M
Tomfohr
 
JK
Devlin
 
BH
, et al. 
SpA: web-accessible spectratype analysis: data management, statistical analysis and visualization.
Bioinformatics
2005
, vol. 
21
 (pg. 
3697
-
3699
)
19
Kepler
 
TB
He
 
M
Tomfohr
 
JK
, et al. 
Statistical analysis of antigen receptor spectratype data.
Bioinformatics
2005
, vol. 
21
 (pg. 
3394
-
3400
)
20
Moser
 
BK
Stevens
 
GR
Watts
 
CL
The two-sample T test versus Satterthwaite's approximate F test.
Commun Statistics Theory Methods
1989
, vol. 
18
 (pg. 
3963
-
3975
)
21
Bonferroni
 
CE
Il Calcolo Delle Assicurazioni su Gruppi di Teste.
Studi in Onore del Professore Salvatore Ortu Carboni Pubbl. Regio Ist. Sup. Sci. Econ. Comm. Firenze
1935
Italy
Rome
(pg. 
13
-
60
)
22
Shearer
 
WT
Rosenblatt
 
HM
Gelman
 
RS
, et al. 
Lymphocyte subsets in healthy children from birth through 18 years of age: the Pediatric AIDS Clinical Trials Group P1009 study.
J Allergy Clin Immunol
2003
, vol. 
112
 (pg. 
973
-
980
)
23
Prockop
 
SE
Petrie
 
HT
Regulation of thymus size by competition for stromal niches among early T cell progenitors.
J Immunol
2004
, vol. 
173
 (pg. 
1604
-
1611
)
24
Hefton
 
JM
Darlington
 
GJ
Casazza
 
BA
Weksler
 
ME
Immunologic studies of aging: V. Impaired proliferation of PHA responsive human lymphocytes in culture.
J Immunol
1980
, vol. 
125
 (pg. 
1007
-
1010
)
25
Mazzolari
 
E
Forino
 
C
Guerci
 
S
, et al. 
Long-term immune reconstitution and clinical outcome after stem cell transplantation for severe T-cell immunodeficiency.
J Allergy Clin Immunol
2007
, vol. 
120
 (pg. 
892
-
899
)
26
Cavazzana-Calvo
 
M
Carlier
 
F
Le Deist
 
F
, 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
, vol. 
109
 (pg. 
4575
-
4581
)
27
Borghans
 
JA
Bredius
 
RG
Hazenberg
 
MD
, et al. 
Early determinants of long-term T-cell reconstitution after hematopoietic stem cell transplantation for severe combined immunodeficiency.
Blood
2006
, vol. 
108
 (pg. 
763
-
769
)
28
Friedrich
 
W
Honig
 
M
Muller
 
SM
Long-term follow-up in patients with severe combined immunodeficiency treated by bone marrow transplantation.
Immunol Res
2007
, vol. 
38
 (pg. 
165
-
173
)
29
Slatter
 
MA
Brigham
 
K
Dickinson
 
AM
, et al. 
Long-term immune reconstitution after anti-CD52-treated or anti-CD34-treated hematopoietic stem cell transplantation for severe T-lymphocyte immunodeficiency.
J Allergy Clin Immunol
2008
, vol. 
121
 (pg. 
361
-
367
)
30
Patel
 
NC
Chinen
 
J
Rosenblatt
 
HM
, et al. 
Long-term outcomes of nonconditioned patients with severe combined immunodeficiency transplanted with HLA-identical or haploidentical bone marrow depleted of T cells with anti-CD6 mAb.
J Allergy Clin Immunol
2008
, vol. 
122
 (pg. 
1185
-
1193
)
31
Neven
 
B
Leroy
 
S
Decaluwe
 
H
, et al. 
Long-term outcome after hematopoietic stem cell transplantation of a single-center cohort of 90 patients with severe combined immunodeficiency.
Blood
2009
, vol. 
113
 (pg. 
4114
-
4124
)

Supplemental data

Sign in via your Institution