Although the impact of donor graft composition on clinical outcomes after hematopoietic stem cell transplantation (HSCT) has been studied, little is known about the role of intragraft γδ TCR repertoire on clinical outcomes following HSCT. Using a high-throughput sequencing platform, we sought to analyze the TCR γ-chain (TRG) repertoire of γδ T cells within donor stem cell grafts and address its potential impact on clinical response in the corresponding patients. A total of 20 peripheral blood stem cell grafts were analyzed, and donors were classified as CMV+/−. The respective acute myeloid leukemia recipients were followed for disease relapse and acute graft-versus-host disease (aGvHD) development post-HSCT. In all samples, TRG repertoire showed a reduced diversity and displayed overrepresented clones. This was more prominent in grafts from CMV+ donors, which presented a more private repertoire, lower diversity, skewed distribution, and reduced usage of the V9-JP pairing. Grafts given to nonrelapse patients presented a more public repertoire and increased presence of long sequence clonotypes. Variable-joining gene segment usage was not associated with aGvHD development, but a higher usage of V2-JP1 pairing and lower usage of V4-J2/V5-J2/V8-JP2 were observed in grafts given to nonrelapse patients. Our work identified five private overrepresented and one public CDR3 sequence (CATWDGPYYKKLF) associated with CMV infection, in addition to 12 highly frequent public sequences present exclusively in grafts given to nonrelapse patients. Our findings show that, despite CMV infection reshaping the TRG repertoire, TRG composition is not associated with aGvHD development, and several public sequences are associated with clinical remission.

Hematopoietic stem cell transplantation (HSCT) has been used for decades to treat a wide range of hematological malignancies, such as acute myeloid leukemia (AML) (1). Although its efficacy has evolved in the last years, the procedure is still associated with substantial transplant-related morbidity, mainly underlying disease relapse, graft-versus-host disease (GvHD), and infections (2). To improve transplantation outcomes, the impact of donor graft T cell subsets on the clinical outcome after HSCT has been studied. Although it has been clearly shown that αβ T cells present in the grafts are the main component behind GvHD development (36), little is known about γδ T cells’ influence on HSCT clinical outcomes.

Although comprising around 5% of total peripheral blood T cells, γδ T cells are present in substantially greater numbers in peripheral blood stem cell (PBSC) grafts and can impact clinical outcomes after transplant (7). In matched, unrelated (8, 9), partially mismatched T cell–depleted (1012) and haploidentical αβ-depleted (13, 14) HSCT settings, increased numbers of γδ T cells in peripheral blood after transplantation are associated with favorable outcomes. This could partly be due to the persistence of donor graft-derived γδ T cells in the peripheral blood of the patients that quickly reconstitute after transplantation (14, 15) and exert a potentially strong antileukemic effect (1618). This indicates that the number of γδ T cells within the graft and their distribution may help prevent disease recurrence and infections (7, 9). However, the role of these cells remains unclear, and whether HSCT outcomes are influenced by the TCR repertoire of γδ T cells has not been adequately addressed. This emphasizes the need to elucidate which donor graft γδ T cell clones have been infused to the patient during transplantation and how this impacts clinical outcome.

The role of γδ T cells in GvHD development remains controversial. Some studies in mice show that γδ T cells are not primary initiators of GvHD (19, 20) whereas others describe a role for these cells in GvHD pathogenesis (21, 22). In humans, most of the literature shows that the transient increase of γδ T cells after HSCT is not associated with acute GvHD (aGvHD) (10, 11, 2325), although there are contradictory results (26). In an earlier study, grafts with higher overall γδ T cell content were linked to increased cumulative incidence of aGvHD posttransplant (8). Given the functional plasticity and rich specificity of γδ T cells, in-depth analysis is required to fully unravel the role of these cells in GvHD, especially if specific clonotypes, repertoire characteristics, or usage of gene segments is associated with aGvHD development or protection.

The impact of CMV infection on T cell reconstitution after HSCT has been intensively investigated. CMV reactivation leads to posttransplant immune dysfunction (27) and γδ T cell expansion in the peripheral blood (16, 28, 29). γδ T cells exert anti-infectious activity (16), and CMV infection could be related to TCR γ-chain (TRG) distribution changes. It was recently shown that the peripheral blood TRG repertoire is dominated by the V-J segment V9-JP, presenting quick reconstitution after HSCT and adaptive clonal expansion in response to CMV infection (15). However, little is known about its distribution on PBSC grafts and how CMV can impact the TRG repertoire.

In this study, we used a next-generation sequencing (NGS) platform to analyze the TRG repertoire of γδ T cells within PBSC grafts given to 20 AML patients, evaluating its potential link to clinical outcome as well as to donor CMV status. We hypothesized that the γδ T cell composition of the grafts could be associated with clinical relapse or aGvHD development. Our findings show that the grafts from CMV+ donors presented a reshaped TRG repertoire, the intragraft TRG composition is not associated with aGvHD incidence, and several private and public sequences were associated with nonrelapse after HSCT.

Twenty AML patients (7 males and 13 females) who underwent HSCT from 2014 to 2016 at the Centre for Allogeneic Stem Cell Transplantation (Karolinska University Hospital, Huddinge, Sweden) and the respective donors (11 males and 9 females) were enrolled in this study. Patients’ and donors’ characteristics are shown in Table I. The median (range) recipient and donor age at time of enrollment were 59.5 (20–71) and 30 (19–71) y, respectively. All patients received unmanipulated PBSC grafts, comprising 17 grafts from matched, unrelated donors and 3 from matched siblings. All unrelated donors were fully matched for HLA-A, -B, -C, and -DRB1 loci. Patients were followed for a median period of 32.7 mo, and disease relapse was evaluated according to previously described criteria (30). Grading of aGvHD was performed according to the Glucksberg criteria over a 3 mo period post-HSCT. Donor and recipient CMV status was assessed following standard guidelines (31). The study was approved by the Regional Ethical Review Board in Stockholm, Sweden (2008/206-31, 2010/760-31/1, 2013/2215-32, 2017/469-32), and performed according to the Declaration of Helsinki.

PBSC samples were obtained during preparation of the graft for infusion and processed as previously described (3). Mononuclear cells were separated by density gradient centrifugation (800 g, 20 min, Lymphoprep; Fresenius Kabi, Oslo, Norway) and cryopreserved in liquid nitrogen in 10% DMSO in RPMI 1640 medium (HyClone; Thermo Fisher, Waltham, MA) enriched with 10% human AB-serum (Karolinska University Hospital), 100 IU/ml penicillin G (Life Technologies, Paisley, U.K.), and 100 mg/ml streptomycin (Life Technologies).

PBSC mononuclear cell staining was performed as described previously (3). Anti-CD3 (clone UCHT1; BD Biosciences, San Jose, CA) and anti-TCR pan γδ (clone REA591; Miltenyi Biotec, Teterow, Germany) Abs were used to assess the percentage of total T cells and total γδ T cells. Anti-TCR Vγ9 (clone B3; BioLegend), anti-TCR Vδ1 (clone TS8.2; Thermo Scientific), and anti-TCR Vδ2 (clone B6; BioLegend) Abs were used for further graft γδ T cell characterization. Samples were acquired by a FACSCanto instrument (BD Biosciences) using FACSDiva software (BD Biosciences). Gating strategy is provided in Supplemental Fig. 1.

γδ T cells were isolated from the grafts with the Anti-TCRγ/δ MicroBead Kit (Miltenyi Biotec) following the manufacturer’s instructions. Purity was checked by flow cytometry and was higher than 90% for all samples. Immediately after selection, genomic DNA was extracted using the EZ1 DNA Blood Kit and EZ1 instruments (QIAGEN, Hilden, Germany) and stored at −20°C. The extracted DNA (1 μg) was used for survey-level deep sequencing of the γ-chain using the ImmunoSEQ platform by Adaptive Biotechnologies (Seattle, WA). Briefly, bias-controlled V and J gene primers were used to amplify rearranged V-J segments for high-throughput sequencing (32, 33). After correcting sequencing errors via a clustering algorithm, CDR3 segments were annotated according to the International ImMunoGeneTics collaboration, identifying which V and J genes contributed to each rearrangement (34). NGS data are available at the Adaptive Biotechnology ImmunoSEQ site (https://clients.adaptivebiotech.com/immuneaccess, https://doi.org/10.21417/LA122018).

Sequencing data were initially analyzed using the ImmunoSEQ Analyzer (https://clients.adaptivebiotech.com/login). Postanalysis of TRG repertoire diversity, clonal space homeostasis, segment usage, spectratyping, and repertoire overlap were performed using the tcR package (35), VDJTools (36), and VDJviz browser (37).

Patient characteristics and the distributions of the continuous variables are described as median ± interquartile range. The Mann–Whitney nonparametric two-tailed test was used to assess differences between any two group comparisons. The p values <0.05 were considered statistically significant. Statistical analysis was performed using GraphPad Prism version 7 (San Diego, CA) or R Project Package in VDJTools and tcR environments.

HSCT patients were grouped into relapse (n = 8) or nonrelapse (n = 12) groups based on clinical follow-up. All grades of aGvHD (I–III) were grouped (aGvHD Yes, n = 12) and compared with patients who did not develop aGvHD (aGvHD No, n = 8). Donor grafts were divided into CMV+ (n = 7) and CMV (n = 13). Individual patient characteristics are shown in Table I.

Table I.
Patients’ characteristics
PatientRecipient Gender/AgeUnderlying DiseaseDisease StatusDonorDonor Gender/AgeConditioning RegimenGvHD ProphylaxisGraft SourceRecipient CMV StatusDonor CMV StatusCMV ReactivationRelapseaGvHD Grade
F/47 AML LR MUD M/23 MAC CsA+MTX PBSC − − No Yes 
F/69 AML LR MUD F/36 RIC CsA+MTX PBSC Yes No 
F/71 AML LR MUD F/28 RIC CsA+MTX PBSC − Yes Yes 
F/64 AML LR Sib M/61 RIC CsA+MTX PBSC No Yes 
F/65 AML LR MUD F/27 RIC CsA+MTX PBSC − Yes Yes 
F/54 AML LR MUD F/31 RIC CsA+MTX PBSC − Yes No 
F/61 AML LR Sib M/71 RIC CsA+MTX PBSC − No Yes 
M/63 AML HR MUD M/23 RIC CsA+MTX PBSC − Yes No 
M/38 AML HR MUD M/30 RIC CsA+MTX PBSC Yes No 
10 F/21 AML LR Sib M/22 MAC CsA+MTX PBSC − − No No 
11 F/40 AML LR MUD F/45 RIC CsA+MTX PBSC − No No 
12 F/66 AML LR MUD M/19 RIC CsA+MTX PBSC − Yes Yes 
13 F/21 AML LR MUD M/35 MAC CsA+MTX PBSC − − No Yes 
14 M/40 AML HR MUD F/20 RIC CsA+MTX PBSC Yes Yes 
15 M/20 AML HR MUD M/33 MAC CsA+MTX PBSC − Yes No 
16 M/30 AML LR MUD F/30 MAC CsA+MTX PBSC − − No No 
17 M/60 AML LR MUD M/21 RIC CsA+MTX PBSC − − No No 
18 M/65 AML LR MUD F/34 RIC CsA+MTX PBSC Yes No 
19 F/59 AML LR MUD F/37 RIC CsA+MTX PBSC Yes No 
20 F/64 AML LR MUD M/29 RIC CsA+MTX PBSC Yes No 
PatientRecipient Gender/AgeUnderlying DiseaseDisease StatusDonorDonor Gender/AgeConditioning RegimenGvHD ProphylaxisGraft SourceRecipient CMV StatusDonor CMV StatusCMV ReactivationRelapseaGvHD Grade
F/47 AML LR MUD M/23 MAC CsA+MTX PBSC − − No Yes 
F/69 AML LR MUD F/36 RIC CsA+MTX PBSC Yes No 
F/71 AML LR MUD F/28 RIC CsA+MTX PBSC − Yes Yes 
F/64 AML LR Sib M/61 RIC CsA+MTX PBSC No Yes 
F/65 AML LR MUD F/27 RIC CsA+MTX PBSC − Yes Yes 
F/54 AML LR MUD F/31 RIC CsA+MTX PBSC − Yes No 
F/61 AML LR Sib M/71 RIC CsA+MTX PBSC − No Yes 
M/63 AML HR MUD M/23 RIC CsA+MTX PBSC − Yes No 
M/38 AML HR MUD M/30 RIC CsA+MTX PBSC Yes No 
10 F/21 AML LR Sib M/22 MAC CsA+MTX PBSC − − No No 
11 F/40 AML LR MUD F/45 RIC CsA+MTX PBSC − No No 
12 F/66 AML LR MUD M/19 RIC CsA+MTX PBSC − Yes Yes 
13 F/21 AML LR MUD M/35 MAC CsA+MTX PBSC − − No Yes 
14 M/40 AML HR MUD F/20 RIC CsA+MTX PBSC Yes Yes 
15 M/20 AML HR MUD M/33 MAC CsA+MTX PBSC − Yes No 
16 M/30 AML LR MUD F/30 MAC CsA+MTX PBSC − − No No 
17 M/60 AML LR MUD M/21 RIC CsA+MTX PBSC − − No No 
18 M/65 AML LR MUD F/34 RIC CsA+MTX PBSC Yes No 
19 F/59 AML LR MUD F/37 RIC CsA+MTX PBSC Yes No 
20 F/64 AML LR MUD M/29 RIC CsA+MTX PBSC Yes No 

CsA, cyclosporine A; F, female; HR, high risk; LR, low risk; M, male; MAC, myeloablative conditioning; MTX, methotrexate; MUD, matched unrelated donor; RIC, reduced intensity conditioning; Sib, matched sibling.

The median total T cell frequency (percentage) was comparable between the relapse versus nonrelapse groups (30.5 versus 32.23%) and aGvHD Yes versus aGvHD No (33.55 versus 28.04%). CMV+ grafts presented a higher T cell percentage than CMV ones (43.68 versus 26.7%, p = 0.006). The same profile was observed for the percentage of γδ T cells, with no differences between relapsing or aGvHD incidence groups (9.03 versus 10.07% and 11.59 versus 7.89%, respectively) but a higher percentage in CMV+ donor grafts (12.3 versus 7.0%, p = 0.023). Regarding the major γδ T cell subfamilies Vγ9, Vδ1, and Vδ2, no differences were observed between the groups (Supplemental Fig. 1).

Deep sequencing showed similar median total productive templates (i.e., the sum of template counts for all productive rearrangements in the sample) in grafts given to patients experiencing a relapse or not (2.08 × 104 versus 1.06 × 104) and patients with or without aGvHD (1.07 × 104 versus 1.96 × 104, Fig. 1A). The same was observed for the number of unique reads (i.e., number of clonotypes) between the two sets of patient groups. However, grafts given to patients who later developed aGvHD trended toward fewer reads than those who did not develop aGvHD (respectively, median values 3.30 × 103 versus 2.91 × 103 and 2.33 × 103 versus 4.86 × 103, Fig. 1B).

FIGURE 1.

TRG diversity and clonality are not related to clinical response or aGvHD incidence. Sum of productive templates (A), unique reads (B), and unique/total reads ratio (C) of the TRG repertoire in the different groups. Unique CDR3 ratio ranges from 0 to 1, with 0 representing the lowest diversity and 1 representing the maximal diversity. (D) Quantile clonality plot of representative donor grafts from each group. The pie chart is divided into singletons (clonotypes represented by a single read), doubletons (two reads), and high-order clonotypes (three and more reads). High-order clonotypes are divided into five quantiles (top 20% of unique high-order clonotypes and so on). The top 10 clonotypes of the first quantile are explicitly shown. The size of each segment is the cumulative frequency of all clonotypes that fall into the corresponding frequency category. (E) Efron–Thisted estimator quantification in the different groups. (F) TRG diversity, calculated as inverse Simpson D index. (G) Donor graft clonality. Values for clonality range from 0 to 1. Values near 1 represent samples with one or a few predominant rearrangements (monoclonal or oligoclonal samples) dominating the observed repertoire. Clonality values near 0 represent more polyclonal samples. The boundaries of the boxes indicate the 25th and 75th percentiles, the lines within the boxes indicate the median, and the whiskers mark the range. Data were compared by two-tailed Mann–Whitney test.

FIGURE 1.

TRG diversity and clonality are not related to clinical response or aGvHD incidence. Sum of productive templates (A), unique reads (B), and unique/total reads ratio (C) of the TRG repertoire in the different groups. Unique CDR3 ratio ranges from 0 to 1, with 0 representing the lowest diversity and 1 representing the maximal diversity. (D) Quantile clonality plot of representative donor grafts from each group. The pie chart is divided into singletons (clonotypes represented by a single read), doubletons (two reads), and high-order clonotypes (three and more reads). High-order clonotypes are divided into five quantiles (top 20% of unique high-order clonotypes and so on). The top 10 clonotypes of the first quantile are explicitly shown. The size of each segment is the cumulative frequency of all clonotypes that fall into the corresponding frequency category. (E) Efron–Thisted estimator quantification in the different groups. (F) TRG diversity, calculated as inverse Simpson D index. (G) Donor graft clonality. Values for clonality range from 0 to 1. Values near 1 represent samples with one or a few predominant rearrangements (monoclonal or oligoclonal samples) dominating the observed repertoire. Clonality values near 0 represent more polyclonal samples. The boundaries of the boxes indicate the 25th and 75th percentiles, the lines within the boxes indicate the median, and the whiskers mark the range. Data were compared by two-tailed Mann–Whitney test.

Close modal

All groups presented similarly low unique CDR3 TRG ratios (i.e., TCR diversity; 0.004 versus 0.002 and 0.003 versus 0.004, Fig. 1C). The unique CDR3 ratio ranges from 0 to 1, with 0 representing the lowest diversity and 1 representing the maximal diversity. The median ratio found in all samples was 0.003, indicating that the γδ TCR repertoire in the grafts contains oligoclonal TRG sequences and low diversity. A closer TCR inspection by quantile plots revealed that the grafts presented multiple clonal overrepresentations, contributing to a skewed clonotype size distribution (Fig. 1D). To summarize the quantile plots, we assessed the TCR richness. We did not find any differences between the patient groups regarding the Efron–Thisted estimator (0.65 × 104 versus 0.66 × 104 and 0.58 × 104 versus 0.94 × 104, Fig. 1E), Simpson D index (72.58 versus 60.91 and 52.78 versus 75.70, Fig. 1F), or sample productive clonality (0.30 versus 0.29 and 0.29 versus 0.31, Fig. 1G). Altogether, our data suggest that CDR3 TRG diversity in PBSC grafts is overall reduced but does not seem to affect clinical relapse or aGvHD incidence.

We then analyzed the TRG clonal proportion and space homeostasis in PBSC samples (35). The top 10 most abundant clonotypes represented a large part of the repertoire (Fig. 2A), and almost all grafts presented a significant amount of hyperexpanded clones (i.e., clones with frequency ≥1%, Fig. 2B), leading to the reduced diversity. The median proportion of top 10 clones was similar between both relapse and nonrelapse patients (23.01 versus 25.67%) and whether patients experienced aGvHD or not (25% in both) (Fig. 2C).

FIGURE 2.

Clonal space homeostasis and TRG spectratyping. (A) Clonal proportion of the top n clonotypes. Red bars represent the TRG proportion taken by the 10 most abundant clones. (B) Proportion of homeostatic space occupied by clonotypes classified as hyperexpanded (0.01–1), large (0.001–0.01), medium (0.0001–0.001), small (0.00001–0.0001), and rare (0–0.00001). See Table I for clinical characteristics of individual patients. (C) Quantification of top 10 clone proportions in the different groups. The boundaries of the boxes indicate the 25th and 75th percentiles, the lines within the boxes indicate the median, and the whiskers mark the range. (D) Quantification of homeostatic space taken up by the clones in the different groups. Bars represent the median and interquartile range. (E) Frequency of unique CDR3 sequences with different nucleotide lengths. Bars represent the median and interquartile range. (F) Distribution patterns of unique CDR3 lengths are displayed with line graphs. Lines represent the nonlinear curve fitting (Gauss function) the frequency of TRG with different lengths in each group. The vertical line indicates the median length in each group. *p ≤ 0.05, two-tailed Mann–Whitney U test.

FIGURE 2.

Clonal space homeostasis and TRG spectratyping. (A) Clonal proportion of the top n clonotypes. Red bars represent the TRG proportion taken by the 10 most abundant clones. (B) Proportion of homeostatic space occupied by clonotypes classified as hyperexpanded (0.01–1), large (0.001–0.01), medium (0.0001–0.001), small (0.00001–0.0001), and rare (0–0.00001). See Table I for clinical characteristics of individual patients. (C) Quantification of top 10 clone proportions in the different groups. The boundaries of the boxes indicate the 25th and 75th percentiles, the lines within the boxes indicate the median, and the whiskers mark the range. (D) Quantification of homeostatic space taken up by the clones in the different groups. Bars represent the median and interquartile range. (E) Frequency of unique CDR3 sequences with different nucleotide lengths. Bars represent the median and interquartile range. (F) Distribution patterns of unique CDR3 lengths are displayed with line graphs. Lines represent the nonlinear curve fitting (Gauss function) the frequency of TRG with different lengths in each group. The vertical line indicates the median length in each group. *p ≤ 0.05, two-tailed Mann–Whitney U test.

Close modal

To address the impact of TRG clonal distribution on patient outcome, we classified clonotypes according to the TCR proportion taken up by clones measured as rare (0–0.001%), small (0.001–0.01%), medium (0.01–0.1%), large (0.1–1%), or hyperexpanded (1–100%). All patients showed few rare clonotypes, further explaining the reduced TRG diversity observed in all samples. Grafts given to patients who did not relapse after HSCT showed a contraction of clones classified as small (respective medians of 7.04 versus 18.86%, p = 0.01) and an expansion of clones classified as medium (32.61 versus 18.23%, p = 0.01) and large (29.38 versus 18.61%, p = 0.04) when compared with relapse patients (Fig. 2D). No differences were observed when looking at the occurrence or absence of aGvHD.

To investigate the profiles of CDR3 lengths in the TRG repertoire, the length of productive nucleotide CDR3 sequences was analyzed for every TRGV/TRGJ gene combination. The length of the CDR3 sequences ranged from 24 to 60 nt, with a median of 45 nt (Fig. 2E). Grafts received by the nonrelapse patient group had an increased proportion of CDR3 with long sequences (54 and 57 nt long) compared with relapse patients (median of 0.41 versus 0.27% [p = 0.02] and 0.11 versus 0.04% [p = 0.04], respectively). Patients who did not develop aGvHD received grafts with more enriched CDR3 sequences of 24 and 33 nt than the non-aGvHD patient group (0.80 versus 0.38% [p = 0.03] and 4.1 versus 3.3% [p = 0.04]). In general, these groups showed a bell-shaped pattern of the CDR3 region (Fig. 2F), characteristic of a Gaussian distribution associated with healthy TCR repertoire distribution. Our results indicate that γδ T cell clonal distribution and TRG CDR3 spectratyping could be related to disease relapse.

CMV infection is associated with γδ T cell expansion and TRG reshaping in the peripheral blood (28). We investigated whether TRG distribution in PBSC grafts was also affected by donor CMV status. The median total productive templates was similar between CMV+ and CMV graft donors (0.54 × 104 versus 1.85 × 104, Fig. 3A). CMV+ donor grafts showed a trend toward fewer unique reads than their counterparts, although the difference was not statistically significant (1.39 × 103 versus 4.03 × 103, Fig. 3B). The CDR3 TRG ratio was low and similar between the groups (0.003 versus 0.004, Fig. 3C). The quantile plots revealed that in CMV+ donor grafts, in addition to the multiple clonal expansions that has been shown in all grafts, a single dominant clone can occupy 21% of the TCR repertoire (Fig. 3D).

FIGURE 3.

CMV+ donors present reduced TRG diversity and increased clonality. Sum of productive templates (A), unique reads (B), and unique/total reads ratio (C) of the TRG repertoire in CMV and CMV+ donor grafts. (D) Quantile clonality plot of representative donor grafts from each group. (E) Efron–Thisted estimator quantification. (F) TRG diversity, calculated as inverse Simpson D index. (G) Donor graft clonality. (H) Quantification of top 10 clone proportions in the different groups. The boundaries of the boxes indicate the 25th and 75th percentiles, the lines within the boxes indicate the median, and the whiskers mark the range. (I) Quantification of homeostatic space taken up by the clones in the different groups. (J) Frequency of unique CDR3 sequences with different nucleotide lengths. Bars represent the median and interquartile range. See Table I for clinical characteristics and Fig. 2 for individual clonal space homeostasis. (K) Distribution patterns of unique CDR3 lengths are displayed with line graphs. Lines represent the nonlinear curve fitting (Gauss function) the frequency of TRG with different lengths in each group. The vertical line indicates the median length in each group. *p ≤ 0.05, **p < 0.01, two-tailed Mann–Whitney U test.

FIGURE 3.

CMV+ donors present reduced TRG diversity and increased clonality. Sum of productive templates (A), unique reads (B), and unique/total reads ratio (C) of the TRG repertoire in CMV and CMV+ donor grafts. (D) Quantile clonality plot of representative donor grafts from each group. (E) Efron–Thisted estimator quantification. (F) TRG diversity, calculated as inverse Simpson D index. (G) Donor graft clonality. (H) Quantification of top 10 clone proportions in the different groups. The boundaries of the boxes indicate the 25th and 75th percentiles, the lines within the boxes indicate the median, and the whiskers mark the range. (I) Quantification of homeostatic space taken up by the clones in the different groups. (J) Frequency of unique CDR3 sequences with different nucleotide lengths. Bars represent the median and interquartile range. See Table I for clinical characteristics and Fig. 2 for individual clonal space homeostasis. (K) Distribution patterns of unique CDR3 lengths are displayed with line graphs. Lines represent the nonlinear curve fitting (Gauss function) the frequency of TRG with different lengths in each group. The vertical line indicates the median length in each group. *p ≤ 0.05, **p < 0.01, two-tailed Mann–Whitney U test.

Close modal

Grafts from CMV+ donors presented significantly reduced diversity, as shown by having the lowest Efron–Thisted estimator (4.09 × 103 versus 7.71 × 103, p = 0.02, Fig. 3E) and inverse Simpson D index (30.70 versus 81.21, p = 0.02, Fig. 3F) and the highest clonality (0.42 versus 0.26, p = 0.006, Fig. 3G). The median proportion of top 10 clones was 2-fold higher in CMV+ grafts (51.48 versus 22.17%, p = 0.01, Fig. 3H). Corroborating these data, hyperexpanded clones took up 2.5 times more space in the CMV+ grafts (49.33 versus 19.38%, p = 0.007) at the expense of those present in small and medium quantities (4.95 versus 15.32% [p = 0.04] and 20.40 versus 40.00% [p = 0.04], respectively, Fig. 3I).

CDR3 spectratyping revealed that CMV+ grafts had a significantly decreased proportion of CDR3 sequences having 24, 27, 42, and 51 nt lengths (0.27 versus 0.61% [p = 0.01], 0.57 versus 1.12% [p = 0.02], 5.11 versus 7.54% [p = 0.007] and 0.61 versus 2.1% [p = 0.04], respectively). CMV+ grafts also had an increase of sequences with 30–39 nt, and the increase was significant for sequences having 36 nt compared with CMV grafts (5.32 versus 4.13%, p = 0.03, Fig. 3J). As a result, whereas CMV grafts present a Gaussian distribution of the CDR3 region, CMV+ grafts presented a non-Gaussian distribution (Fig. 3K), indicating that CMV positivity leads to dominant expansion of clones and subsequent reshaping of the grafts’ γδ TCR repertoire.

Taken together, the above data suggest that CDR3 TRG diversity is significantly reduced in the CMV+ PBSC grafts and that CMV may lead to TCR reshaping, with certain CDR3γ clonotypes undergoing CMV-driven clonal expansion.

To further explore the TRG repertoire composition, we performed hierarchical clustering of TRGV and TRGJ segment usage profiles (36). Despite observing two cluster formations based on J2 and JP expression, the grafts presented an overall similar V and J gene usage profile regardless of their clinical outcome after HSCT or donor graft CMV status (Fig. 4A).

FIGURE 4.

V/J gene usage and V-J combination pairing frequency. (A) TRGV (left) and TRGJ (right) usage profiles in the different groups. Hierarchical clustering of samples was based on the Euclidean distance between V (left) and J (right) segment frequency vectors. Lower x-axis labels indicate donor graft number (see Table I for clinical characteristics). Top x-axis color bars indicate patient groups. Note that there is no clear clustering for any of the groups. (B) Frequency usage of individual TRGV (left) and TRGJ (right) genes in the different groups. Bars represent median and interquartile range. (C) V-J segment pairing abundance in CDR3 junctions of representative donor grafts from each group. Chord diagrams are used for visualization; ribbons connecting segment pairs are scaled by corresponding V-J pair frequency. (D) Frequency of different TRGV-TRGJ rearrangements in the different groups. Bars represent median and interquartile range. *p ≤ 0.05, two-tailed Mann–Whitney U test.

FIGURE 4.

V/J gene usage and V-J combination pairing frequency. (A) TRGV (left) and TRGJ (right) usage profiles in the different groups. Hierarchical clustering of samples was based on the Euclidean distance between V (left) and J (right) segment frequency vectors. Lower x-axis labels indicate donor graft number (see Table I for clinical characteristics). Top x-axis color bars indicate patient groups. Note that there is no clear clustering for any of the groups. (B) Frequency usage of individual TRGV (left) and TRGJ (right) genes in the different groups. Bars represent median and interquartile range. (C) V-J segment pairing abundance in CDR3 junctions of representative donor grafts from each group. Chord diagrams are used for visualization; ribbons connecting segment pairs are scaled by corresponding V-J pair frequency. (D) Frequency of different TRGV-TRGJ rearrangements in the different groups. Bars represent median and interquartile range. *p ≤ 0.05, two-tailed Mann–Whitney U test.

Close modal

We also assessed the usage frequency of individual TRGV and TRGJ families. For all groups, the segments TRGV9 and TRGJP were the most frequent. Patients who did not relapse after HSCT received grafts with a bias toward lower usage of TRGV4, TRGV5, and TRGJP2 segments and higher usage of TRGJP1 compared with relapse patients (median 4.13 versus 7.46% [p = 0.04], 1.12 versus 4.77% [p = 0.02], 3.02 versus 5.52% [p = 0.02], and 6.62 versus 3.23% [p = 0.02], respectively). No differences were observed between the aGvHD and non-aGvHD groups (Fig. 4B). The TCR repertoire in CMV+ donor grafts presented a lower TRGV2 and TRGJP expression (6.29 versus 8.99%, p = 0.04, and 30.99 versus 60.63%, p = 0.01) as well as a higher TRGJP1 gene usage compared with CMV counterparts (11.04 versus 3.90%, p = 0.04).

Changes in V-J segment recombination in CDR3 junctions were then assessed (Fig. 4C) (36, 37). The V9-JP combination was the most frequent pairing in all patients. Nonrelapse patients received grafts with lower usage of the pairings V4-J2, V5-J2, and V8-JP2 (median 3.12 versus 6.57% [p = 0.04], 1.06 versus 4.38% [p = 0.02], and 1.33 versus 1.91% [p = 0.03], respectively) and higher usage of the V2-JP1 pairing (1.28 versus 0.36%, p = 0.03) than relapse patients. No differences were observed between aGvHD groups (Fig. 3D). The TCR usage of the sequence pairs V2-J2, V2-JP2, V4-JP2, V9-JP, and V9-JP2 was lower in CMV+ than in CMV grafts (median 4.62 versus 6.94% [p = 0.04], 0.59 versus 1.33% [p = 0.04], 0.31 versus 0.09% [p = 0.02], 30.75 versus 60.30% [p = 0.02], and 0.07 versus 0.29% [p = 0.01], respectively, Fig. 4D). In summary, we show that γδ T cells from the grafts mostly express TRGV9 and TRGJP segments. Additionally, we identified some significant variations in V and J gene usage and V-J pairing between the patient groups that did or did not relapse.

Next, we checked for TCR overlap and clonotype sharing (35) within the patient groups and whether these could be associated with the observed clinical outcome. Overall hierarchical clustering and multidimensional scaling did not show a clear clustering between the clinical groups (Fig. 5A). Actually, 36–1041 (median of 246) of all the 25,882 unique CDR3 amino acid sequences identified were shared between grafts (Fig. 5B). Therefore, we performed the combined rearrangement analysis of all grafts using the ImmunoSEQ Analyzer, searching for private (i.e., sequences present exclusively in one patient) and public sequences (i.e., sequences shared between at least two patients). The total number of amino acid public sequences was 3886, representing 15.02% of the total number of combined clones between samples. In fact, only one clone (CALWEVQELGKKIKVF) was shared between all samples, whereas the number of clones present exclusively in one sample was 21,996 (84.98%). This indicates that the TRG repertoire in PBSC is mostly composed of private sequences.

FIGURE 5.

The donor graft private repertoire is related to CMV positivity, whereas the public repertoire is associated with clinical response. (A) Left, Hierarchical clustering of donor grafts using the F pairwise similarity metric (the geometric mean of the total frequency of overlapping clonotypes). Right, Multidimensional scaling (MDS) plot. Samples were projected onto a two-dimensional plane based on pairwise similarities (F metric). See Table I for donor graft characteristics. Note that there is no clear coclustering within the different groups. (B) Clonotype sharing between the samples. (C) Morisita–Horn overlap indices for the clinical groups. All samples from the same group were overlapped with each other. The index ranges from 0 to 1, with 0 representing more private repertoires (samples do not share clonotypes; no similarity) and 1 indicating public repertoires (samples have identical clonotypes; complete similarity). Each dot represents one pairing between two samples from the same group, the boundaries of the boxes indicate the 25th and 75th percentiles, the lines within the boxes indicate the median, and the whiskers mark the range. ***p ≤ 0.001, ****p ≤ 0.0001, two-tailed Mann–Whitney U test. (D) Pie charts depicting the 10 most abundant clones in three CMV+ donor grafts. The most-expanded clones are highlighted in colors. Note that the samples do not share the same overrepresented clonotypes. Check Table II for clone identification. (E) Stacked clonotype frequency plot highlighting the details of overlap between CMV graft donors that were given to patients who did not relapse after HSCT. The top 100 clonotypes based on their average frequency in those samples are shown and colored by the peak position of their abundance profile, whereas other clonotypes that were observed in the samples are marked as “Not shown.” The frequency of remaining clonotypes is marked as “Not overlapping.” Check Table II for clone identification.

FIGURE 5.

The donor graft private repertoire is related to CMV positivity, whereas the public repertoire is associated with clinical response. (A) Left, Hierarchical clustering of donor grafts using the F pairwise similarity metric (the geometric mean of the total frequency of overlapping clonotypes). Right, Multidimensional scaling (MDS) plot. Samples were projected onto a two-dimensional plane based on pairwise similarities (F metric). See Table I for donor graft characteristics. Note that there is no clear coclustering within the different groups. (B) Clonotype sharing between the samples. (C) Morisita–Horn overlap indices for the clinical groups. All samples from the same group were overlapped with each other. The index ranges from 0 to 1, with 0 representing more private repertoires (samples do not share clonotypes; no similarity) and 1 indicating public repertoires (samples have identical clonotypes; complete similarity). Each dot represents one pairing between two samples from the same group, the boundaries of the boxes indicate the 25th and 75th percentiles, the lines within the boxes indicate the median, and the whiskers mark the range. ***p ≤ 0.001, ****p ≤ 0.0001, two-tailed Mann–Whitney U test. (D) Pie charts depicting the 10 most abundant clones in three CMV+ donor grafts. The most-expanded clones are highlighted in colors. Note that the samples do not share the same overrepresented clonotypes. Check Table II for clone identification. (E) Stacked clonotype frequency plot highlighting the details of overlap between CMV graft donors that were given to patients who did not relapse after HSCT. The top 100 clonotypes based on their average frequency in those samples are shown and colored by the peak position of their abundance profile, whereas other clonotypes that were observed in the samples are marked as “Not shown.” The frequency of remaining clonotypes is marked as “Not overlapping.” Check Table II for clone identification.

Close modal

Next, we calculated the overlap of full TRG sequences from each patient group pair by the Morisita–Horn similarity index (35). Nonrelapse patients presented a significantly higher Morisita–Horn index than relapse patients (0.67 versus 0.32, p < 0.001), indicating the occurrence of a higher number of public clonotypes shared between nonrelapse patients (Fig. 5C). There was no difference between patients who did or did not develop aGvHD (median 0.43 versus 0.45 respectively). A 4.7-fold lower index was observed in CMV+ donor grafts compared with CMV ones (0.13 versus 0.61, p < 0.0001). This indicates that donor graft CMV positivity leads to a significantly more private TCR repertoire compared with CMV grafts.

As highlighted in Fig. 5D, the CMV+ donor grafts have a notable presence of hyperexpanded clones that can take up to 41.61% of the entire repertoire. These overrepresented clonotypes are not shared between the CMV+ grafts and further corroborate the private TRG repertoires observed in these donors. After running a tracking clonotype routine (36) on CMV grafts, we observed that nonrelapse patients may present a clonotype sharing of up to 23.60% (Fig. 5E). This shows that CMV+ donor grafts are associated with private repertoires, whereas patients who did not relapse after HSCT received grafts with more public clonotypes.

We then checked which clonotypes are overrepresented and exclusively present in the different patient groups, as well as the clones shared between patients within the same patient group. We identified four private overrepresented sequences exclusively present in grafts given to nonrelapse patients taking from 2.00 to 6.23% of the TRG repertoire (Table II). All these sequences were longer than 45 nt, corroborating the spectratyping data shown above. Additionally, we identified 12 public clones shared exclusively between the grafts received by nonrelapsing patients.

Table II.
Analysis of donor graft TRG clonotypes
GroupCDR3 Amino Acid SequenceCDR3 Length (nt)Productive Reads (%)aTRGPatient
Private Overrepresented Clones 
 Nonrelapse CALWEVRIQELGKKIKVF 51 6.23 V9-JP 
CALWEVRTQELGKKIKVF 51 1.93 V9-JP 17 
CATWDERNYIVVGDWIKTF 54 2.47 V2-JP2 
CALWEVRGYQELGKKIKVF 54 2.00 V9-JP 15 
 aGvHD No CATWDMGELF 27 0.94 V8-J2 
CATWWGRWFKIF 33 9.82 V8-JP1 
 CATWWTGWFKIF 33 2.44 V8-JP1 
 CMV+ CAWGAGWFKIF 30 41.61 V9-JP 20 
CATWDIENYYKKLF 39 25.13 V8-J2 
CALWEMQELGKKIKVF 45 29.87 V9-JP 
CALWELAELGKKIKVF 45 13.72 V9-JP 19 
CALWEVLDELGKKIKVF 48 20.39 V9-JP 14 
Public Clones Shared by Same Group 
 Nonrelapse CALWEEELGKKIKVF 42 5.08   
CALWETQELGKKIKVF 45 3.47   
CALWEVSELGKKIKVF 45 3.02   
CALWEVELGKKIKVF 42 2.94   
CALWEVHGELGKKIKVF 48 2.85   
CALWELQELGKKIKVF 45 2.45   
CALWAQELGKKIKVF 42 1.48   
CALWEVGLGKKIKVF 42 1.45   
CATWDGRYKKLF 33 0.23   
CATWDGPKKLF 30 0.22   
CATWDGRYYKKLF 36 0.22   
CATWDRPNYYKKLF 39 0.19   
 aGvHD No CATWDRGYYKKLF 36 0.44   
CALWEVPYYKKLF 36 0.30   
CATWDRDYKKLF 33 0.25   
CATWDRRKKLF 30 0.24   
 CMV+ CATWDGPYYKKLF 36 0.51   
GroupCDR3 Amino Acid SequenceCDR3 Length (nt)Productive Reads (%)aTRGPatient
Private Overrepresented Clones 
 Nonrelapse CALWEVRIQELGKKIKVF 51 6.23 V9-JP 
CALWEVRTQELGKKIKVF 51 1.93 V9-JP 17 
CATWDERNYIVVGDWIKTF 54 2.47 V2-JP2 
CALWEVRGYQELGKKIKVF 54 2.00 V9-JP 15 
 aGvHD No CATWDMGELF 27 0.94 V8-J2 
CATWWGRWFKIF 33 9.82 V8-JP1 
 CATWWTGWFKIF 33 2.44 V8-JP1 
 CMV+ CAWGAGWFKIF 30 41.61 V9-JP 20 
CATWDIENYYKKLF 39 25.13 V8-J2 
CALWEMQELGKKIKVF 45 29.87 V9-JP 
CALWELAELGKKIKVF 45 13.72 V9-JP 19 
CALWEVLDELGKKIKVF 48 20.39 V9-JP 14 
Public Clones Shared by Same Group 
 Nonrelapse CALWEEELGKKIKVF 42 5.08   
CALWETQELGKKIKVF 45 3.47   
CALWEVSELGKKIKVF 45 3.02   
CALWEVELGKKIKVF 42 2.94   
CALWEVHGELGKKIKVF 48 2.85   
CALWELQELGKKIKVF 45 2.45   
CALWAQELGKKIKVF 42 1.48   
CALWEVGLGKKIKVF 42 1.45   
CATWDGRYKKLF 33 0.23   
CATWDGPKKLF 30 0.22   
CATWDGRYYKKLF 36 0.22   
CATWDRPNYYKKLF 39 0.19   
 aGvHD No CATWDRGYYKKLF 36 0.44   
CALWEVPYYKKLF 36 0.30   
CATWDRDYKKLF 33 0.25   
CATWDRRKKLF 30 0.24   
 CMV+ CATWDGPYYKKLF 36 0.51   

Analysis of overrepresented TRG clones exclusively detected in private repertoires (upper) and public clones shared between all patients with same clinical status (lower). Table shows CDR3 amino acid sequence, CDR3 length in nucleotides, productive reads (%), V-J segment pairing, and patient number (see Table I).

a

Given percentage of a particular patient or the sum of all productively rearranged CDR3 sequences per group.

For patients who did not have aGvHD, we found three preponderant clones in private repertoires and four public sequences shared exclusively between the patients from this group that could be associated with the clinical outcome (Table II), comprising 0.94–9.82% of the TRG repertoire. CMV+ grafts presented the highest percentage of repertoire taken by private overrepresented clones identified within the group, ranging from 13.72 to 41.61%. We found one public TCR sequence exclusively present in all patients from this group (CATWDGPYYKKLF) consisting of 39 nt. This indicates that some clones might play a role in clinical outcome after HSCT or in TRG repertoire reshaping in CMV+ donor grafts. The identified private and public clones may provide useful information to improve γδ T cell–based therapies in the future.

HSCT is widely applied in clinical practice to treat AML patients, being recognized as one of the most potent curative therapies for this condition (38). To improve transplantation safety, the impact of donor graft T cell composition has been studied and the role of αβ T cells on clinical outcome described (36). Research on γδ T cells remains hampered by limited information on their biology and potential role in HSCT (39). To our knowledge, we used for the first time a high-throughput analysis of the γδ T cell TRG repertoire to characterize γδ T cell distribution in the grafts received by AML patients. We show that basic TRG characteristics are not related to aGvHD development and identified public TRG CDR3 sequences that may play a role in AML remission after HSCT. In addition, we describe that CMV positivity is associated with graft TRG repertoire reshaping and overrepresentation of private sequences.

Initial evaluations of TRG repertoire by unique CDR3 ratio showed that CDR3 diversity is reduced overall in PBSC grafts, and there is high presence of multiple clonal expansions. This is in accordance with a recent study showing that γδ T cells from adult peripheral blood present much lower TRG diversity than cord blood samples, a result of age-driven clonal expansion (15). Additionally, a skewed peripheral blood TRG distribution is observed in adulthood (15, 40), indicating that the reduced diversity in the grafts might be related to expansion of TRG clones during life and harvested within the hematopoietic stem cells. Additional data are required to further show the reduced TRG diversity in grafts when compared with cord blood and adult samples. Furthermore, G-CSF used in hematopoietic stem cell mobilization may have affected the distribution and clonality of the TRG repertoire. Despite the fact that earlier literature has shown that G‐CSF mobilization does not alter the total amount of the γδ T cell fraction, which remains unchanged between peripheral blood of healthy donors and PBSC grafts (8), a previous work showed some TRG families expanding, presenting no expression, or having an oligoclonal distribution after mobilization (41). Which TRG clonotypes are expanded or depleted during mobilization should be addressed in future studies. These studies should be performed in the light of different mobilization regimens, such as G-CSF versus plerixafor, considering that different mechanisms of action can be associated with specific cells’ mobilization and potentially associated with posttransplant events.

The reduced diversity and high presence of overrepresented clones was more prominent in CMV+ graft donors. These grafts presented high frequency of hyperexpanded clones and non-Gaussian distribution of the TRG CDR3 length when compared with CMV counterparts, indicating TRG disturbance and reshaping. CMV infection leads to T cell inflation and clonal expansion, resulting in reduced TCR repertoire diversity (27, 42). CMV reactivation after HSCT impacts γδ T cell reconstitution, driving the proliferation of γδ T cell clones (15, 16, 29). There is no information about its influence on graft γδ T cell distribution. In this article, we show that donor CMV positivity is associated with a high clonality, increased frequency of hyperexpanded clones, and a reduced usage of the V9-JP pair, the most abundant rearrangement present in peripheral blood (15, 43). These changes are probably due to adaptive clonal expansion of distinct TRG clones in response to CMV (15). A recent study showed that few private CMV-induced TRG sequences underwent massive proliferation after CMV reactivation post-HSCT (15). Although it is not possible to compare the amino acid sequences with that study because of methodological differences, it did show specific clonotypes taking up more than 60% of the TRG repertoire after CMV reactivation (15). In the same way, we show private, hyperexpanded clonotypes taking up around 40% of the TRG repertoire. This indicates that donor graft CMV positivity is associated with the expansion of CMV-specific clones, reshaping the TRG repertoire.

After HSCT, an increased number of γδ T cells is associated with a better clinical outcome in terms of higher disease-free and event-free survivals (9, 10, 12, 14), making these cells promising candidates for cellular therapy (39). As γδ T cells exert a potent antileukemic effect (16) and can mediate a graft-versus-leukemia effect without causing GvHD (44), ongoing trials are investigating the benefits of grafts enriched in γδ T cell numbers (13, 14) and their role in donor lymphocyte infusion (45). The impact on outcome of γδ T cell donor graft composition is most likely not solely related to the absolute number of γδ T cells present in the graft, underlining the importance of a full qualitative characterization of these cells in the PBSC grafts. TRG NGS allowed us to show that basic characteristics of the repertoire, such as diversity, clonality, and number of reads, are not related to clinical relapse after HSCT. In contrast, we found substantial changes on the occupied homeostatic space, spectratyping, and V and J segment usage, as well as bias in V-J pairing frequencies on grafts given to nonrelapse patients that could be related to nonrelapse disease. Importantly, we also identified public TRG CDR3 amino acid sequences present exclusively in grafts given to patients who did experience clinical relapse after HSCT. Those clonotypes might play a role in clinical improvement after HSCT, given that previous works have shown that γδ T cells exert strong antileukemic effect against AML cell lines in vitro (1618) and possibly in vivo (46). Alternatively, these clonotypes might reflect hidden features of these grafts with a protective effect against relapse.

Although the selection of grafts with specific TRG clones might be difficult, the TRG sequences identified in this study might be used for the generation of a γδ TCR library that can be used to treat AML relapse. These libraries can be constructed by the rapid and efficient methods currently available for cloning and expression of specific TCRs by T cells, allowing prompt generation of leukemia/Ag-specific cells (47). These clonotypes can be transfected into donor lymphocyte infusion products and used to boost leukemia killing in cases of disease relapse after HSCT, allowing the production of leukemia-specific cellular products (48). In vitro studies are still required to show that the clonotypes described in this article exert high killing of AML blasts and that they can be used as a platform for TCR editing.

Although we did not evaluate the reconstitution of these clonotypes after HSCT, it is known that γδ T cells infused within the graft persist after transplantation and undergo fast immune reconstitution (14, 15). In particular, donor CMV-specific clones can be found in the peripheral blood of HSCT recipients for long times after transplant (49). Altogether, these studies show that clones identified in the graft persist after HSCT, indicating that the clones identified in this study might be related to AML remission. Future works comparing host and donor γδ T cell reconstitution and their role in clinical relapse or aGvHD development are still required.

A controversial study showed that patients receiving PBSC with high γδ T cell levels have both an increased cumulative incidence and higher grades of aGvHD (8). Additionally, G-CSF use in mobilization not only influences the distribution and expression levels of the γδ T cell repertoire but also changes γδ T cell clonality and might play a role in mediating aGvHD (41). Using deep sequencing and in-depth analysis, we show that TRG diversity metrics, V and J family usage, V-J pairing, clonal distribution, and spectratyping were not related to aGvHD development. Additionally, the clonality and TCR overlap were similar between patients who did or did not develop aGvHD, supporting the idea that γδ T cells are not involved in GvHD development (9, 10, 14). Future work to assess γδ T cell phenotype and functionality will help to further clarify their role in GvHD.

Ravens et al. (15) described that TRG repertoires in peripheral blood consist mostly of public sequences. In contrast, our work showed that around 15% of the TRG repertoire is composed of public sequences shared between the PBSC grafts, indicating that a large fraction of graft TRG repertoire consists of private sequences. This change from public TRG on peripheral blood to private TRG on grafts may occur during HSC mobilization by the use of G-CSF, once its use has been associated with TRG repertoire changes from a polyclonal distribution before mobilization to an oligoclonal distribution after mobilization (41). The most prevalent public Vγ9 clonotype (CALWEVQELGKKIKVF) (50), described to be persistent in fetus blood (51), cord blood (52, 53), and adults (34, 52, 54), was the only sequence found in all samples analyzed. This shows the presence of lifelong clonotypes in the graft, although their role is still unknown.

We found five private, overrepresented clones exclusively found in CMV+ donors that could be used in the future to develop CMV-specific cell therapies using γδ T cells. Additionally, public sequences were especially found in grafts given to patients who did not relapse after HSCT, sharing a large amount of their repertoire and presenting 12 exclusive sequences. CMV+ grafts showed a high presence of private clonotypes, probably due to TRG reshaping, indicating that the donor CMV status could be associated with AML outcome after HSCT. A beneficial effect of CMV reactivation after HSCT has been recently demonstrated, as several studies reported reduced risk of relapse in AML patients who develop CMV reactivation (5557). Additionally, donor CMV positivity alone was also associated with a lower relapse rate after HSCT (58). The immunological background underlying this complex interplay between CMV and relapse is still unclear and represents an interesting topic for ongoing research. γδ T cells can be an important player in this context. Identifying clones related to CMV infection and clinical remission of AML patients might aid in the choice of suitable γδ TCR sequences for cellular therapy (39, 47, 48). Larger validation cohorts are necessary to confirm our data.

Donor graft αβ T cell characterization has been extensively performed. In summary, grafts containing higher percentages of regulatory and CD4+ T cells and lower frequencies of CD27+, CD127+, CD28+, and of naive CD8+ T cells have been associated with reduced aGvHD incidence (36) and low relapse rates (4, 5). Oppositely, grafts enriched with γδ T cells were associated with higher aGvHD development but not relapse (8). These cells can interplay and influence clinical outcomes in a complementary or contrasting way (59). Future works evaluating the in-depth αβ TCR composition of the grafts and the detailed phenotype of graft γδ T cells are required to clarify these issues and their joint influence on clinical outcomes.

CMV reactivation, primary disease relapse, and GvHD represent the main causes of transplant-related morbidity and mortality following allogeneic HSCT. The immunological background underlying this complex interplay is still unclear, and γδ T cells can be an important player in this context. To our knowledge, our work provides the first deep sequencing results allowing characterization of γδ T cells infused within the grafts in a matched, unrelated HSCT setting. Using several complementary, in-depth evaluations, we show that none of the γδ T cell TRG repertoire characteristics evaluated (e.g., diversity, clonality, distribution, segment usage, and overlap) were associated with aGvHD development. We also identified public clones associated with nonrelapsing disease that may exert a graft-versus-leukemia effect. This opens the window for large-scale manipulation to enrich, isolate, and expand specific γδ T cell clones to explore the full biology of those cells or to assess their role in HSCT for AML patients. Additionally, we identified several private clones associated with donor graft CMV status. The clonotypes identified in this study could be used in the future to better understand the adaptive responses of γδ T cells and to improve the efficacy and safety of HSCT.

This work was supported by an Adaptive Biotechnologies Young Investigator Award (to L.C.M.A.).

The online version of this article contains supplemental material.

Abbreviations used in this article:

aGvHD

acute GvHD

AML

acute myeloid leukemia

GvHD

graft-versus-host disease

HSCT

hematopoietic stem cell transplantation

NGS

next-generation sequencing

PBSC

peripheral blood stem cell

TRG

TCR γ-chain.

1
Ringdén
,
O.
,
H.
Karlsson
,
R.
Olsson
,
B.
Omazic
,
M.
Uhlin
.
2009
.
The allogeneic graft-versus-cancer effect.
Br. J. Haematol.
147
:
614
633
.
2
Ghimire
,
S.
,
D.
Weber
,
E.
Mavin
,
X. N.
Wang
,
A. M.
Dickinson
,
E.
Holler
.
2017
.
Pathophysiology of GvHD and other HSCT-related major complications.
Front. Immunol.
8
:
79
.
3
Stikvoort
,
A.
,
A.
Gaballa
,
M.
Solders
,
I.
Nederlof
,
B.
Önfelt
,
B.
Sundberg
,
M.
Remberger
,
M.
Sundin
,
J.
Mattsson
,
M.
Uhlin
.
2018
.
Risk factors for severe acute graft-versus-host disease in donor graft composition.
Biol. Blood Marrow Transplant.
24
:
467
477
.
4
Impola
,
U.
,
A.
Larjo
,
U.
Salmenniemi
,
M.
Putkonen
,
M.
Itälä-Remes
,
J.
Partanen
.
2016
.
Graft immune cell composition associates with clinical outcome of allogeneic hematopoietic stem cell transplantation in patients with AML.
Front. Immunol.
7
:
523
.
5
Danby
,
R. D.
,
W.
Zhang
,
P.
Medd
,
T. J.
Littlewood
,
A.
Peniket
,
V.
Rocha
,
D. J.
Roberts
.
2016
.
High proportions of regulatory T cells in PBSC grafts predict improved survival after allogeneic haematopoietic SCT.
Bone Marrow Transplant.
51
:
110
118
.
6
Wikell
,
H.
,
S.
Ponandai-Srinivasan
,
J.
Mattsson
,
J.
Gertow
,
M.
Uhlin
.
2014
.
Cord blood graft composition impacts the clinical outcome of allogeneic stem cell transplantation.
Transpl. Infect. Dis.
16
:
203
212
.
7
Minculescu
,
L.
,
H.
Sengeløv
.
2015
.
The role of gamma delta T cells in haematopoietic stem cell transplantation.
Scand. J. Immunol.
81
:
459
468
.
8
Pabst
,
C.
,
H.
Schirutschke
,
G.
Ehninger
,
M.
Bornhäuser
,
U.
Platzbecker
.
2007
.
The graft content of donor T cells expressing gamma delta TCR+ and CD4+foxp3+ predicts the risk of acute graft versus host disease after transplantation of allogeneic peripheral blood stem cells from unrelated donors.
Clin. Cancer Res.
13
:
2916
2922
.
9
Perko
,
R.
,
G.
Kang
,
A.
Sunkara
,
W.
Leung
,
P. G.
Thomas
,
M. H.
Dallas
.
2015
.
Gamma delta T cell reconstitution is associated with fewer infections and improved event-free survival after hematopoietic stem cell transplantation for pediatric leukemia.
Biol. Blood Marrow Transplant.
21
:
130
136
.
10
Lamb
,
L. S.
 Jr.
,
P. J.
Henslee-Downey
,
R. S.
Parrish
,
K.
Godder
,
J.
Thompson
,
C.
Lee
,
A. P.
Gee
.
1996
.
Increased frequency of TCR γ δ + T cells in disease-free survivors following T cell-depleted, partially mismatched, related donor bone marrow transplantation for leukemia.
J. Hematother.
5
:
503
509
.
11
Lamb
,
L. S.
 Jr.
,
A. P.
Gee
,
L. J.
Hazlett
,
P.
Musk
,
R. S.
Parrish
,
T. P.
O’Hanlon
,
S. S.
Geier
,
R. S.
Folk
,
W. G.
Harris
,
K.
McPherson
, et al
.
1999
.
Influence of T cell depletion method on circulating gammadelta T cell reconstitution and potential role in the graft-versus-leukemia effect.
Cytotherapy
1
:
7
19
.
12
Godder
,
K. T.
,
P. J.
Henslee-Downey
,
J.
Mehta
,
B. S.
Park
,
K.-Y.
Chiang
,
S.
Abhyankar
,
L. S.
Lamb
.
2007
.
Long term disease-free survival in acute leukemia patients recovering with increased gammadelta T cells after partially mismatched related donor bone marrow transplantation.
Bone Marrow Transplant.
39
:
751
757
.
13
Locatelli
,
F.
,
P.
Merli
,
D.
Pagliara
,
G.
Li Pira
,
M.
Falco
,
D.
Pende
,
R.
Rondelli
,
B.
Lucarelli
,
L. P.
Brescia
,
R.
Masetti
, et al
.
2017
.
Outcome of children with acute leukemia given HLA-haploidentical HSCT after αβ T cell and B-cell depletion.
Blood
130
:
677
685
.
14
Airoldi
,
I.
,
A.
Bertaina
,
I.
Prigione
,
A.
Zorzoli
,
D.
Pagliara
,
C.
Cocco
,
R.
Meazza
,
F.
Loiacono
,
B.
Lucarelli
,
M. E.
Bernardo
, et al
.
2015
.
γδ T cell reconstitution after HLA-haploidentical hematopoietic transplantation depleted of TCR-αβ +/CD19+ lymphocytes. [Published erratum appears in 2016 Blood 127: 1620.]
Blood
125
:
2349
2358
.
15
Ravens
,
S.
,
C.
Schultze-Florey
,
S.
Raha
,
I.
Sandrock
,
M.
Drenker
,
L.
Oberdörfer
,
A.
Reinhardt
,
I.
Ravens
,
M.
Beck
,
R.
Geffers
, et al
.
2017
.
Human γδ T cells are quickly reconstituted after stem-cell transplantation and show adaptive clonal expansion in response to viral infection. [Published erratum appears in 2018 Nat. Immunol. 19: 1037.]
Nat. Immunol.
18
:
393
401
.
16
Scheper
,
W.
,
S.
van Dorp
,
S.
Kersting
,
F.
Pietersma
,
C.
Lindemans
,
S.
Hol
,
S.
Heijhuurs
,
Z.
Sebestyen
,
C.
Gründer
,
V.
Marcu-Malina
, et al
.
2013
.
γδT cells elicited by CMV reactivation after allo-SCT cross-recognize CMV and leukemia.
Leukemia
27
:
1328
1338
.
17
Dolstra
,
H.
,
H.
Fredrix
,
A.
van der Meer
,
T.
de Witte
,
C.
Figdor
,
E.
van de Wiel-van Kemenade
.
2001
.
TCR gamma delta cytotoxic T lymphocytes expressing the killer cell-inhibitory receptor p58.2 (CD158b) selectively lyse acute myeloid leukemia cells.
Bone Marrow Transplant.
27
:
1087
1093
.
18
Gertner-Dardenne
,
J.
,
R.
Castellano
,
E.
Mamessier
,
S.
Garbit
,
E.
Kochbati
,
A.
Etienne
,
A.
Charbonnier
,
Y.
Collette
,
N.
Vey
,
D.
Olive
.
2012
.
Human Vγ9Vδ2 T cells specifically recognize and kill acute myeloid leukemic blasts.
J. Immunol.
188
:
4701
4708
.
19
Drobyski
,
W. R.
,
D.
Majewski
,
G.
Hanson
.
1999
.
Graft-facilitating doses of ex vivo activated gammadelta T cells do not cause lethal murine graft-vs.-host disease.
Biol. Blood Marrow Transplant.
5
:
222
230
.
20
Drobyski
,
W. R.
,
S.
Vodanovic-Jankovic
,
J.
Klein
.
2000
.
Adoptively transferred gamma delta T cells indirectly regulate murine graft-versus-host reactivity following donor leukocyte infusion therapy in mice.
J. Immunol.
165
:
1634
1640
.
21
Blazar
,
B. R.
,
P. A.
Taylor
,
A.
Panoskaltsis-Mortari
,
T. A.
Barrett
,
J. A.
Bluestone
,
D. A.
Vallera
.
1996
.
Lethal murine graft-versus-host disease induced by donor gamma/delta expressing T cells with specificity for host nonclassical major histocompatibility complex class Ib antigens.
Blood
87
:
827
837
.
22
Ellison
,
C. A.
,
G. C.
MacDonald
,
E. S.
Rector
,
J. G.
Gartner
.
1995
.
Gamma delta T cells in the pathobiology of murine acute graft-versus-host disease. Evidence that gamma delta T cells mediate natural killer-like cytotoxicity in the host and that elimination of these cells from donors significantly reduces mortality.
J. Immunol.
155
:
4189
4198
.
23
Cela
,
M. E.
,
M. S.
Holladay
,
C. M.
Rooney
,
S.
Richardson
,
B.
Alexander
,
R. A.
Krance
,
M. K.
Brenner
,
H. E.
Heslop
.
1996
.
Gamma delta T lymphocyte regeneration after T lymphocyte-depleted bone marrow transplantation from mismatched family members or matched unrelated donors.
Bone Marrow Transplant.
17
:
243
247
.
24
Watanabe
,
N.
,
M.
Narita
,
T.
Furukawa
,
T.
Nakamura
,
A.
Yamahira
,
M.
Masuko
,
K.
Toba
,
I.
Fuse
,
Y.
Aizawa
,
M.
Takahashi
.
2011
.
Kinetics of pDCs, mDCs, γδT cells and regulatory T cells in association with graft versus host disease after hematopoietic stem cell transplantation.
Int. J. Lab. Hematol.
33
:
378
390
.
25
Yabe
,
M.
,
H.
Yabe
,
K.
Hattori
,
T.
Hinohara
,
T.
Morimoto
,
S.
Kato
,
A.
Kusunoki
.
1994
.
Transition of T cell receptor gamma/delta expressing double negative (CD4-/CD8-) lymphocytes after allogeneic bone marrow transplantation.
Bone Marrow Transplant.
14
:
741
746
.
26
Viale
,
M.
,
S.
Ferrini
,
A.
Bacigalupo
.
1992
.
TCR gamma/delta positive lymphocytes after allogeneic bone marrow transplantation.
Bone Marrow Transplant.
10
:
249
253
.
27
Suessmuth
,
Y.
,
R.
Mukherjee
,
B.
Watkins
,
D. T.
Koura
,
K.
Finstermeier
,
C.
Desmarais
,
L.
Stempora
,
J. T.
Horan
,
A.
Langston
,
M.
Qayed
, et al
.
2015
.
CMV reactivation drives posttransplant T cell reconstitution and results in defects in the underlying TCRβ repertoire.
Blood
125
:
3835
3850
.
28
Khairallah
,
C.
,
J.
Déchanet-Merville
,
M.
Capone
.
2017
.
γδ T cell-mediated immunity to cytomegalovirus infection.
Front. Immunol.
8
:
105
.
29
Knight
,
A.
,
A. J.
Madrigal
,
S.
Grace
,
J.
Sivakumaran
,
P.
Kottaridis
,
S.
Mackinnon
,
P. J.
Travers
,
M. W.
Lowdell
.
2010
.
The role of Vδ2-negative γδ T cells during cytomegalovirus reactivation in recipients of allogeneic stem cell transplantation.
Blood
116
:
2164
2172
.
30
Törlén
,
J.
,
O.
Ringdén
,
K.
Garming-Legert
,
P.
Ljungman
,
J.
Winiarski
,
K.
Remes
,
M.
Itälä-Remes
,
M.
Remberger
,
J.
Mattsson
.
2016
.
A prospective randomized trial comparing cyclosporine/methotrexate and tacrolimus/sirolimus as graft-versus-host disease prophylaxis after allogeneic hematopoietic stem cell transplantation.
Haematologica
101
:
1417
1425
.
31
Ljungman
,
P.
,
M.
Boeckh
,
H. H.
Hirsch
,
F.
Josephson
,
J.
Lundgren
,
G.
Nichols
,
A.
Pikis
,
R. R.
Razonable
,
V.
Miller
,
P. D.
Griffiths
;
Disease Definitions Working Group of the Cytomegalovirus Drug Development Forum
.
2017
.
Definitions of cytomegalovirus infection and disease in transplant patients for use in clinical trials.
Clin. Infect. Dis.
64
:
87
91
.
32
Carlson
,
C. S.
,
R. O.
Emerson
,
A. M.
Sherwood
,
C.
Desmarais
,
M.-W.
Chung
,
J. M.
Parsons
,
M. S.
Steen
,
M. A.
LaMadrid-Herrmannsfeldt
,
D. W.
Williamson
,
R. J.
Livingston
, et al
.
2013
.
Using synthetic templates to design an unbiased multiplex PCR assay.
Nat. Commun.
4
:
2680
.
33
Emerson
,
R. O.
,
W. S.
DeWitt
,
M.
Vignali
,
J.
Gravley
,
J. K.
Hu
,
E. J.
Osborne
,
C.
Desmarais
,
M.
Klinger
,
C. S.
Carlson
,
J. A.
Hansen
, et al
.
2017
.
Immunosequencing identifies signatures of cytomegalovirus exposure history and HLA-mediated effects on the T cell repertoire.
Nat. Genet.
49
:
659
665
.
34
Sherwood
,
A. M.
,
C.
Desmarais
,
R. J.
Livingston
,
J.
Andriesen
,
M.
Haussler
,
C. S.
Carlson
,
H.
Robins
.
2011
.
Deep sequencing of the human TCRγ and TCRβ repertoires suggests that TCRβ rearranges after αβ and γδ T cell commitment.
Sci. Transl. Med.
3
:
90ra61
.
35
Nazarov
,
V. I.
,
M. V.
Pogorelyy
,
E. A.
Komech
,
I. V.
Zvyagin
,
D. A.
Bolotin
,
M.
Shugay
,
D. M.
Chudakov
,
Y. B.
Lebedev
,
I. Z.
Mamedov
.
2015
.
tcR: an R package for T cell receptor repertoire advanced data analysis.
BMC Bioinformatics
16
:
175
.
36
Shugay
,
M.
,
D. V.
Bagaev
,
M. A.
Turchaninova
,
D. A.
Bolotin
,
O. V.
Britanova
,
E. V.
Putintseva
,
M. V.
Pogorelyy
,
V. I.
Nazarov
,
I. V.
Zvyagin
,
V. I.
Kirgizova
, et al
.
2015
.
VDJtools: unifying post-analysis of T cell receptor repertoires.
PLoS Comput. Biol.
11
:
e1004503
.
37
Bagaev
,
D. V.
,
I. V.
Zvyagin
,
E. V.
Putintseva
,
M.
Izraelson
,
O. V.
Britanova
,
D. M.
Chudakov
,
M.
Shugay
.
2016
.
VDJviz: a versatile browser for immunogenomics data.
BMC Genomics
17
:
453
.
38
Döhner
,
H.
,
E. H.
Estey
,
S.
Amadori
,
F. R.
Appelbaum
,
T.
Büchner
,
A. K.
Burnett
,
H.
Dombret
,
P.
Fenaux
,
D.
Grimwade
,
R. A.
Larson
, et al
European LeukemiaNet
.
2010
.
Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet.
Blood
115
:
453
474
.
39
Handgretinger
,
R.
,
K.
Schilbach
.
2018
.
The potential role of γδ T cells after allogeneic HCT for leukemia.
Blood
131
:
1063
1072
.
40
Kallemeijn
,
M. J.
,
F. G.
Kavelaars
,
M. Y.
van der Klift
,
I. L. M.
Wolvers-Tettero
,
P. J. M.
Valk
,
J. J. M.
van Dongen
,
A. W.
Langerak
.
2018
.
Next-generation sequencing analysis of the human TCRγδ+ T cell repertoire reveals shifts in Vγ- and Vδ-usage in memory populations upon aging.
Front. Immunol.
9
:
448
.
41
Xuan
,
L.
,
X.
Wu
,
Y.
Zhang
,
Z.
Fan
,
Y.
Ling
,
F.
Huang
,
F.
Zhang
,
X.
Zhai
,
Q.
Liu
.
2011
.
Granulocyte colony-stimulating factor affects the distribution and clonality of TRGV and TRDV repertoire of T cells and graft-versus-host disease.
J. Transl. Med.
9
:
215
.
42
Klenerman
,
P.
,
A.
Oxenius
.
2016
.
T cell responses to cytomegalovirus.
Nat. Rev. Immunol.
16
:
367
377
.
43
Pauza
,
C. D.
,
C.
Cairo
.
2015
.
Evolution and function of the TCR Vgamma9 chain repertoire: it’s good to be public.
Cell. Immunol.
296
:
22
30
.
44
Scheper
,
W.
,
C.
Gründer
,
T.
Straetemans
,
Z.
Sebestyen
,
J.
Kuball
.
2014
.
Hunting for clinical translation with innate-like immune cells and their receptors.
Leukemia
28
:
1181
1190
.
45
Rådestad
,
E.
,
H.
Wikell
,
M.
Engström
,
E.
Watz
,
B.
Sundberg
,
S.
Thunberg
,
M.
Uzunel
,
J.
Mattsson
,
M.
Uhlin
.
2014
.
Alpha/beta T cell depleted grafts as an immunological booster to treat graft failure after hematopoietic stem cell transplantation with HLA-matched related and unrelated donors.
J. Immunol. Res.
2014
:
578741
.
46
Otto
,
M.
,
R. C.
Barfield
,
R.
Iyengar
,
J.
Gatewood
,
I.
Müller
,
M. S.
Holladay
,
J.
Houston
,
W.
Leung
,
R.
Handgretinger
.
2005
.
Human gammadelta T cells from G-CSF-mobilized donors retain strong tumoricidal activity and produce immunomodulatory cytokines after clinical-scale isolation.
J. Immunother.
28
:
73
78
.
47
Guo
,
X. Z.
,
P.
Dash
,
M.
Calverley
,
S.
Tomchuck
,
M. H.
Dallas
,
P. G.
Thomas
.
2016
.
Rapid cloning, expression, and functional characterization of paired αβ and γδ T cell receptor chains from single-cell analysis.
Mol. Ther. Methods Clin. Dev.
3
:
15054
.
48
Chabannon
,
C.
,
J.
Kuball
,
A.
Bondanza
,
F.
Dazzi
,
P.
Pedrazzoli
,
A.
Toubert
,
A.
Ruggeri
,
K.
Fleischhauer
,
C.
Bonini
.
2018
.
Hematopoietic stem cell transplantation in its 60s: a platform for cellular therapies.
Sci. Transl. Med.
10
:
eaap9630
.
49
Terasako-Saito
,
K.
,
H.
Nakasone
,
Y.
Tanaka
,
R.
Yamazaki
,
M.
Sato
,
K.
Sakamoto
,
Y.
Ishihara
,
K.
Kawamura
,
Y.
Akahoshi
,
J.
Hayakawa
, et al
.
2014
.
Persistence of recipient-derived as well as donor-derived clones of cytomegalovirus pp65-specific cytotoxic T cells long after allogeneic hematopoietic stem cell transplantation.
Transpl. Infect. Dis.
16
:
930
940
.
50
Willcox
,
C. R.
,
M. S.
Davey
,
B. E.
Willcox
.
2018
.
Development and selection of the human Vγ9Vδ2+ T-cell repertoire.
Front. Immunol.
9
:
1501
.
51
Dimova
,
T.
,
M.
Brouwer
,
F.
Gosselin
,
J.
Tassignon
,
O.
Leo
,
C.
Donner
,
A.
Marchant
,
D.
Vermijlen
.
2015
.
Effector Vγ9Vδ2 T cells dominate the human fetal γδ T cell repertoire.
Proc. Natl. Acad. Sci. USA
112
:
E556
E565
.
52
Cairo
,
C.
,
B.
Sagnia
,
G.
Cappelli
,
V.
Colizzi
,
R. G. F.
Leke
,
R. J.
Leke
,
C. D.
Pauza
.
2013
.
Human cord blood γδ T cells expressing public Vγ2 chains dominate the response to bisphosphonate plus interleukin-15.
Immunology
138
:
346
360
.
53
Davey
,
M. S.
,
C. R.
Willcox
,
S.
Hunter
,
S. A.
Kasatskaya
,
E. B. M.
Remmerswaal
,
M.
Salim
,
F.
Mohammed
,
F. J.
Bemelman
,
D. M.
Chudakov
,
Y. H.
Oo
,
B. E.
Willcox
.
2018
.
The human Vδ2+ T cell compartment comprises distinct innate-like Vγ9+ and adaptive Vγ9- subsets.
Nat. Commun.
9
:
1760
.
54
Davey
,
M. S.
,
C. R.
Willcox
,
S. P.
Joyce
,
K.
Ladell
,
S. A.
Kasatskaya
,
J. E.
McLaren
,
S.
Hunter
,
M.
Salim
,
F.
Mohammed
,
D. A.
Price
, et al
.
2017
.
Clonal selection in the human Vδ1 T cell repertoire indicates γδ TCR-dependent adaptive immune surveillance.
Nat. Commun.
8
:
14760
.
55
Green
,
M. L.
,
W. M.
Leisenring
,
H.
Xie
,
R. B.
Walter
,
M.
Mielcarek
,
B. M.
Sandmaier
,
S. R.
Riddell
,
M.
Boeckh
.
2013
.
CMV reactivation after allogeneic HCT and relapse risk: evidence for early protection in acute myeloid leukemia.
Blood
122
:
1316
1324
.
56
Takenaka
,
K.
,
T.
Nishida
,
Y.
Asano-Mori
,
K.
Oshima
,
K.
Ohashi
,
T.
Mori
,
H.
Kanamori
,
K.
Miyamura
,
C.
Kato
,
N.
Kobayashi
, et al
.
2015
.
Cytomegalovirus reactivation after allogeneic hematopoietic stem cell transplantation is associated with a reduced risk of relapse in patients with acute myeloid leukemia who survived to day 100 after transplantation: the Japan society for hematopoietic cell transplantation transplantation-related complication working group.
Biol. Blood Marrow Transplant.
21
:
2008
2016
.
57
Inagaki
,
J.
,
M.
Noguchi
,
K.
Kurauchi
,
S.
Tanioka
,
R.
Fukano
,
J.
Okamura
.
2016
.
Effect of cytomegalovirus reactivation on relapse after allogeneic hematopoietic stem cell transplantation in pediatric acute leukemia.
Biol. Blood Marrow Transplant.
22
:
300
306
.
58
Nachbaur
,
D.
,
J.
Clausen
,
B.
Kircher
.
2006
.
Donor cytomegalovirus seropositivity and the risk of leukemic relapse after reduced-intensity transplants.
Eur. J. Haematol.
76
:
414
419
.
59
French
,
J. D.
,
C. L.
Roark
,
W. K.
Born
,
R. L.
O’Brien
.
2005
.
γδ T cell homeostasis is established in competition with αβ T cells and NK cells. [Published erratum appears in 2006 Proc. Natl. Acad. Sci. USA 103: 504.]
Proc. Natl. Acad. Sci. USA
102
:
14741
14746
.

The authors have no financial conflicts of interest.

Supplementary data