ORIGINAL RESEARCH REPORT

STEM CELLS AND DEVELOPMENT Volume 00, Number 00, 2014  Mary Ann Liebert, Inc. DOI: 10.1089/scd.2013.0588

T-Cell Receptor Excision Circle Levels After Allogeneic Stem Cell Transplantation Are Predictive of Relapse in Patients with AML and MDS Mehmet Uzunel,1,2 Darius Sairafi,1,2 Mats Remberger,1,2 Jonas Mattsson,1,2 and Michael Uhlin1,2

In this retrospective study, 209 patients with malignant disease were analyzed for levels of T-cell receptor excision circles (TRECs) for the first 24 months after allogeneic stem cell transplantation. CD3 + cells were separated by direct antibody-coupled magnetic beads, followed by DNA extraction according to a standard protocol. The dReccJa signal joint TREC was measured with real-time quantitative PCR. Patients were grouped based on malignant disease: chronic myeloid leukemia, chronic lymphatic leukemia, acute myeloid leukemia (AML), acute lymphocytic leukemia (ALL), and myelodysplastic syndrome (MDS). Patients were further subdivided based on TREC levels below (low-TREC) or above (high-TREC) median at each time point. TREC levels were then correlated to relapse incidence and relapse-free survival (RFS). For patients with AML, low TREC levels 2 months post-transplantation were correlated to high relapse incidence at 5 years (P < 0.05). In patients with chronic leukemia, high TREC levels were correlated with improved RFS (P < 0.05). For patients with MDS, high TREC levels at 9 months post-transplantation were associated with higher RFS at 5 years (P < 0.02) and lower relapse incidence (P < 0.02). This study shows the potential use of TREC measurement in blood to predict relapse in patients with AML and MDS after allogeneic hematopoietic stem cell transplantation.

division. Due to this, the levels of TREC give an indirect measurement of thymic output. Several reports have described parameters that affect thymic activity after ASCT [9,11–13]. However, less is known about how TREC levels post-ASCT affect clinical outcome. Low TREC levels have been associated with an increased incidence of CMV reactivation [14]. This finding is supported by another report that also demonstrated a correlation between low TREC levels and higher transplantrelated mortality (TRM) [9]. Decreased TREC levels after ASCT have further been associated with lower overall survival (OS) [9,15]. To date, only small studies have shown an association between TREC levels post-ASCT and relapse of malignant disease [16,17]. In this study, we analyzed the impact of TREC levels after ASCT on disease relapse and survival in patients with hematological malignancies. Due to the difference in treatment protocols and disease etiology, the patient cohort was split into subgroups based on disease type. We show that a low TREC count after ASCT was associated with an increased relapse rate in patients with acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). Lower TREC levels were also associated with decreased OS in individuals with chronic leukemia and MDS.

Introduction

A

llogeneic hematopoietic stem cell transplantation (ASCT) is an efficient treatment modality for a variety of malignant diseases of the hematopoietic system [1]. The curative potential of ASCT results from the combined effect of chemotherapy and radiotherapy included in the conditioning regimen and the graft-versus-leukemia (GVL) effect exerted by the new donor immune system [2]. T lymphocytes are the main cell type responsible for the GVL effect [3]. The initial leukopenic period after ASCT is associated with a phase of reduced immune function [4]. Hence, an efficient immune reconstitution is of key importance to limit the risk of infections and malignant relapse [5]. The reconstitution of T cells post-ASCT depends on the peripheral expansion of T cells originating from the graft as well as differentiation of T cells from donor hematopoietic stem cells (HSC) [6,7]. The thymic output of de novo, differentiated T cells can be estimated by quantification of T-cell receptor excision circles (TRECs) in peripheral blood [8–10]. TRECs are formed as a circular DNA by-product after the rearrangement of the T-cell receptor genes. Since TRECs are not duplicated during mitosis, their concentration in peripheral circulation decreases after every T-cell 1 2

Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden. Division of Therapeutic Immunology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.

1

2

Materials and Methods

UZUNEL ET AL.

Table 1. Patient and Donor Characteristics

Patients This study included 209 patients with hematological relapse who had undergone ASCT at the Centre for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, between September 2002 and April 2009. Part of the patient material was also included in a study by Sairafi et al. [9]. All patients had a minimum follow-up time of 3 months. Leukemic relapse was defined as 20% or more blast cells in bone marrow (BM) aspirates on morphologic examination or as extramedullary occurrence of leukemic cells. In chronic myeloid leukemia (CML) patients with increasing levels of BCR-ABL, a BCRABL ratio exceeding 0.02% on three consecutive occasions or a ratio of 0.05% on two occasions was also defined as relapsed disease. Patients with MDS were classified according to both the French-American-British (FAB) Cooperative Group and the WHO classifications. All recipients and donors were typed for HLA class I and II by allele-level PCR sequence-specific primers [18]. Additional information about the patients, treatment procedures, and supportive care are given in Table 1. This study was approved by the regional ethics review board (approval No. 2011/549-31/1).

Conditioning Myeloablative conditioning regimens (MACs) consisted of cyclophosphamide (60 mg/kg daily) for 2 days combined with 12 Gy (4 · 3 Gy) of fractionated total body irradiation (TBI), busulfan (1 mg/kg daily) for 4 days, or a combination of busulfan and melfalan [19]. The reduced intensity conditioning regimens consisted of fludarabine in combination with TBI and/or other cytostatic agents [20]. All patients receiving grafts from matched, unrelated donors (MUD) were in vivo T cell depleted using antithymocyte globulin (2 mg/kg for 3–4 days before transplantation [21]).

Graft versus host disease prophylaxis The majority of patients (92%) were treated with cyclosporine A (CsA) combined with four doses of methotrexate (MTX). CsA was discontinued after 3 months in cases with a sibling donor or after 6 months for MUD transplants. Fifteen patients received a combination of tacrolimus and sirolimus (Table 1).

TREC analysis Patient blood samples were collected 2, 3, 9, 12, and 24 months after SCT. CD3 + cells were separated using direct antibody-coupled magnetic beads (Dynabead System; Invitrogen AS) followed by DNA extraction according to a standard protocol. DNA samples were stored at - 20C until use. The dRec-cJa signal joint TREC was measured using a TaqMan real-time quantitative PCR protocol with primers and probes directed to the signal joint region [22]. The amount of TRECs in CD3 + cells was expressed as the ratio between TRECs and the housekeeping gene glyceraldehyde3-phosphate dehydrogenase (GAPDH) and calculated using the delta-CT method, formula 2(Ct GAPDH - Ct TREC). TRECs and GAPDH were analyzed in multiplex. The methodology of TREC analysis is described in more detail elsewhere [9].

n = 209 Recipient age, median (range) Donor age, median (range) Recipient sex, n (%) Female Male Disease stage Low risk/high risk Diagnosis, n (%) CML CLL ALL AML MDS Donor, n (%) HLA-identical sibling Matched unrelated Allele mismatch unrelated Conditioning regimen Myeloablative (MAC) Cy + fTBI Cy + Bu Cy + BU + Melf Reduced intensity conditioning Bu + Flu + ATG Cy + fTBI + Flu + ATG Cy + low fTBI Flu + Cy Treo + Flu ATG GVHD prophylaxis, n (%) CsA + MTX CsA FK + MTX FK + RAPA Graft source, n (%) PBSCs Bone marrow Cell dose, median (range) Total NC ( · 108/kg) PBSCs BM CD34 + cells ( · 106/kg) PBSCs BM

41 (0.5–69) 36 (3–71) 93 (44) 116 (56) 108/101 26 8 50 93 32

(12) (4) (24) (45) (15)

96 (46) 98 (47) 15 (7) 128 (61%) 52 69 7 81 (39%) 64 5 1 3 8 138 (66%) 192 1 1 15

(92) (0.5) (0.5) (7)

166 (79) 43 (21) 12.9 (2.3–54.5) 2.9 (0.98–47) 83 (2.1–660) 38 (6–181)

Low risk, first remission or first chronic phase; high risk, beyond these stages; BM, bone marrow; CML, chronic myeloid leukemia; CLL, chronic lymphocytic leukemia; ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; MDS, myelodysplastic syndrome; MM, mismatch; Cy, cyclophosphamide; fTBI, fractionated total body irradiation; Bu, busulfan; Melf, melfalan, Flu, fludarabine; ATG, antiT-cell globuline; Treo, treosulfan; CsA, cyclosporine A; MTX, methotrexate; FK, tacrolimus; RAPA, sirolimus; PBSC, peripheral blood stem cell; NC, nucleated cell; GVHD, graft versus host disease.

Statistical analysis The probabilities of OS were estimated using the Kaplan– Meyer method and compared with the log-rank test. Relapse and TRM were estimated using an estimator of cumulative incidence curves taking competing events into consideration. The Mann–Whitney U test was used to compare

TREC LEVELS AS A PREDICTOR OF RELAPSE

continuous variables, and Fisher’s exact test was used to compare the distribution of categorical variables. Predictive analysis for OS was performed with the Cox proportional hazards regression. Predictive analyses for relapse, TRM, and graft versus host disease (GVHD) were based on the proportional hazards model for subdistribution of competing risks. Univariate and multivariate analyses were then performed using Gray’s test and the proportional subdistribution hazard regression model developed by Fine and Gray. Factors that were included in the statistical analysis were age, donor type, graft type, risk, cell dose, blood group and HLA matching, conditioning, ATG, GVHD, infections, T-cell chimerism, and the use of mesenchymal stromal cells and TREC levels. Factors that showed a P value of < 0.1 in the univariate analysis were included in the multivariate analysis.

Results Low TREC levels after ASCT are associated with increased risk for relapse in AML patients Patients with AML were analyzed separately for the correlation between TREC levels after ASCT and relapse incidence and relapse-free survival (RFS). Of the tested time points (2, 3, 6, 12, and 24 months), a significant correlation existed only at 2 months between lower TREC levels and a higher incidence of relapse (P < 0.05) (Fig. 1a). Only a trend (P = 0.15) could be observed at 2 months between RFS and low TREC levels (Fig. 1b). There was no correlation observed between TREC levels and OS or incidence of acute graft versus host disease (aGVHD) at any time points (data not shown). Multivariate analysis of risk factors associated with relapse in AML patients showed that both the use of peripheral blood stem cells (P < 0.05) and TREC levels above median at 2 months (P < 0.05) were associated with decreased relapse (Table 2). A major ABO bloodgroup mismatch (P < 0.01) between donor and recipient was associated with an increased relapse rate. Multivariate analysis for RFS only showed a disadvantage for patients with high-risk disease (P < 0.02) and bacteremia post-ASCT (P < 0.01).

3

Low levels of TREC correlate to increased GVHD incidence in acute lymphocytic leukemia patients In patients with acute lymphocytic leukemia (ALL), there was a significant correlation between aGVHD I–IV and TREC levels at 6 and 9 months post-transplantation. Individuals with a TREC value lower than the median had significantly higher incidence of aGHVD (6 months P < 0.001 and 9 months P < 0.02) (Fig. 2). No significant correlation was found between TREC levels and OS, RFS or incidence of relapse (data not shown).

TREC levels post-ASCT predict the survival rate in patients with chronic leukemia In individuals with chronic leukemia [CML and chronic lymphatic leukemia (CLL)], there was a significant correlation between TREC levels at 3 months and RFS (P < 0.05) and OS (P < 0.01) (Fig. 3a, b). No significant correlation was found between TREC levels and aGVHD or relapse incidence at any time point (data not shown). Multivariate analysis for RFS revealed that herpes simplex virus (HSV) reactivation post-HSC transplantation (HSCT) was associated with a decreased RFS (P < 0.05) (Table 2). TREC levels above median at 3 months postHSCT were significantly correlated to an increased RFS (P = 0.06). Multivariate analysis for OS for these patients revealed that reactivation of HSV (P < 0.05) and varicella zoster virus (VZV) (P < 0.05) was associated with a decreased OS. High TREC levels at 3 months post-HSCT could not be included in the multivariate analysis as all patients in that group were alive.

TREC levels post-ASCT in MDS patients predict relapse and long-term OS In patients with MDS, lower TREC levels at 9 months were significantly associated with an increased risk of relapse (P < 0.02) (Fig. 4a). At both 6 and 9 months post-ASCT, lower levels of TREC were associated with a decrease in RFS (P < 0.02) (Fig. 4b, c). For OS, there was a significant

FIG. 1. Acute myeloid leukemia (AML) patients. Incidence of relapse (a) and relapse-free survival (RFS) (b) in correlation to T-cell receptor excision circle (TREC) levels at 2 months post-allogeneic hematopoietic stem cell transplantation (ASCT). Patients were grouped according to TREC levels above or below median levels at 2 months.

4

UZUNEL ET AL.

Table 2. Multivariate Analysis

AML relapse ABO, major MM PBSC TREC > median 2 months AML RFS High risk SEPSIS 0–1 Chronic leukemia RFS HSV Chronic leukemia OS HSV VZV TREC > median 3 months MDS RFS NC dose TREC > median 6 months TREC > median 9 months MDS OS ABO, major MM TREC > median 9 months

P

Risk ratio

0.006 0.03 0.02

4.24 0.32 0.29

0.01 0.002

2.42 3.01

0.03

3.54

0.03 0.03

5.51 7.54

0.02 0.01

0.81 0.05

0.016

4.43

a

a

a

RFS, relapse-free survival; high risk, beyond first remission or first chronic phase; HSV, herpes simplex virus I and II; VZV, varicella zoster virus; TREC, T-cell receptor excision circles; OS, overall survival; a Multivariate was not possible to perform due to 0 individuals in one patient group.

advantage with higher TREC levels at 6 and 9 months postASCT (P < 0.05 and P < 0.02, respectively) (Fig. 4c, e). Most death in the low-TREC groups were due to relapse after 6 months. Multivariate analysis for RFS revealed that nucleated cell dose (P < 0.05) and TREC levels above median, 6 months post-HSCT (P < 0.02), were associated with favorable prognosis in this patient group (Table 2). TREC levels 9 months post-HSCT could not be included as no patients died in the

group containing patients with above median TREC levels. Additionally, in a multivariate analysis for OS in MDS/MPS patients, only a major ABO donor–recipient mismatch was associated with unfavorable prognosis (P < 0.05) (Table 2).

Effect of ATG on TREC levels We have previously shown that ATG significantly decreased TREC levels during the first 6 months post-ASCT [9]. Whereas ATG in itself is also associated with the risk of relapse [23], it is important to adjust the analysis for the patient with or without ATG separately to elucidate the direct effect on the correlation between TREC levels and relapse seen in this patient material. When analyzing only the patients receiving ATG separately for the different disease categories, TREC levels were still significantly associated with relapse and RFS as above (data not shown). For patients without ATG, the same analysis was performed with the same results, except for patients with chronic leukemia and ALL where the numbers were too few. Due to limited patient numbers in the patient subgroups, multivariate analysis was not possible to perform.

Discussion The critical role of rapid T-cell reconstitution postASCT and its links to GVHD and GVL are well known [3,24–27]. As peripheral blood TREC levels provide an easy, but indirect, measurement of T-cell thymic output, we speculated that their levels might be predictive of relapse after ASCT. Studies have shown that TREC is a more accurate marker for recent thymic emigrants compared with phenotypic markers such as CD45RA, CCR7, and CD62L [28,29]. In addition, the frequency of cell populations carrying the surface marker CD45RA changes over time due to the ongoing conversion of these cells to CD45RO+ memory [8]. Whereas several studies have associated TREC levels with CMV reactivation, GVHD, TRM, and OS [5,9,12,14,30–33],

FIG. 2. Acute lymphocytic leukemia (ALL) patients. The impact of acute graft versus host disease (aGVHD) on TREC levels at 3, 6, and 9 months post-ASCT. (a) Patients with any grade of GVHD are compared with patients without GVHD. (b) Patients with severe grade of GVHD (grades II–IV) are compared with patients without or grade I GVHD. Box plots show median values and 25–75 percentiles. The number of patients in each group is shown below the boxes.

TREC LEVELS AS A PREDICTOR OF RELAPSE

5

FIG. 3. Chronic leukemia patients. RFS (a) and overall survival (OS) (b) in correlation to TREC levels at 3 months postASCT. Patients were grouped according to TREC levels above or below median levels at 3 months.

no clear correlation has been shown between relapse and TREC levels after ASCT. An association between leukemic relapse and low TREC levels has been suggested, however, no statistical significance was reported possibly due to limited patient numbers [34]. Two recent studies do describe a significant correlation between recovery of thymic function and relapse after T-cell depleted HLAhaploidentical ASCT [16] and unrelated cord blood ASCT [17]. The studied populations consisted mainly of children with ALL and were relatively small. In this study, the kinetics of TREC levels after ASCT was highly variable and dependent on the primary disease (data not shown). This could be a consequence of the high variability between different diseases as relates to the conditioning regimen and median patient age, both known to significantly impact TREC levels [9,10]. In addition, the preference of different stem cell sources and doses might also influence the development [9,12,30,35,36]. Diseasedependent heterogeneity in thymic reconstitution could be a reason why earlier reports have not shown a convincing association between TREC levels and relapse of the underlying disease [12,13,32]. In an earlier study from our group with partly overlapping patient material, we failed to show any correlation between relapse and TREC levels, but in that study, no subdivision was done between disease indications in contrast to the present study [9]. Clearly, when dividing the patients with malignant disease into subgroups based on ASCT indication, several associations between TREC levels post-ASCT and relapse emerged. In earlier studies, it has been shown that quicker reconstitution of peripheral lymphocyte levels post-ASCT is associated with an increase in OS in patients with acute leukemia [37,38]. However, these studies do not distinguish between cells originating from the grafts and lymphocytes differentiated in the host from HSCs. TREC analysis was used as an estimate of de novo differentiation of naive T cells in the different patient groups. Higher TREC levels 2 months post-ASCT correlated to a lower relapse rate and a higher RFS in patients with AML (Fig. 1 and Table 2). This suggests that the presence of newly produced T cells early after ASCT plays an important

role in the GVL effect. The fact that low TREC levels in AML patients measured at 2 months are predictive for relapse is of clinical importance. Various treatment options exist at this stage to influence the survival chances in these patients, for example, through tapering of the immunosuppressive therapy or treatment with donor lymphocyte infusions (DLI). We could observe a strong association between a major ABO mismatch and an increased risk of relapse (Table 2). This is in contradiction to several larger studies [39,40]. In an earlier study from our center, we also failed to see this association [41]. Our observed results may be related to a small sample size and should be confirmed in a larger study on only AML patients. In patients with ALL, low TREC levels were associated with the incidence of aGVHD at 6 and 9 months (Fig. 2). Previous studies have demonstrated that aGVHD and chronic graft versus host disease (cGVHD) negatively affect TREC levels suggesting that low TREC levels are most likely a consequence, and not cause, of early aGVHD [11,12,14, 31,42]. Interestingly, patients diagnosed with ALL were significantly younger than those in other groups (median age < 20 compared to > 40, Table 1). We have previously reported an association between TREC levels and GVHD in younger ( < 30) individuals [9], in line with the current observations. Of interest is that only in ALL patients (due to the high proportion of children), TREC levels ever reach to levels comparable to normal healthy individuals. For all other patient categories in this cohort, TREC levels stay below normal levels up to 24 months post-HSCT (data not shown). In patients with chronic leukemia, there was a positive correlation between RFS and OS, and higher TREC levels 3 months post-ASCT (Fig. 3). These data are in line with a previous study on CML patients linking a higher relapse rate with no detectable TREC post-ASCT [43]. This suggests that effective thymic output is important to prevent relapse and improve survival in this patient group (Fig. 3). Furthermore, CML has been shown to be responsive to the GVL effect after DLI treatment [44,45]. This is also in line with our finding where a functional T-cell compartment and its putative GLV effect is of importance to inhibit the leukemic disease.

6

UZUNEL ET AL.

FIG. 4. Myelodysplastic syndrome patients. RFS (a, d), OS (b, e), and relapse incidence (c) in correlation to TREC levels at 6 and 9 months post-ASCT. Patients were grouped according to TREC levels above or below median levels at 6 (a, b) or 9 (c–e) months.

MDS consists of a heterogeneous group of clonal stem cell disorders of the hematopoietic lineage. Patients are often characterized by ineffective hematopoiesis with consequent cytopenias and a predisposition for transformation to acute leukemia [46]. Today, ASCT is the only potentially curative

treatment for patients with MDS, but it is associated with a high relapse risk [47]. Low TREC levels significantly correlated to decreased RFS and OS at 6 and 9 months (Fig. 4). Furthermore, the relapse incidence at 9 months correlated to low TREC levels. This is of interest as it is well known that

TREC LEVELS AS A PREDICTOR OF RELAPSE

these patients seldom respond to the added GVL effect of DLI [48]. That a GVL effect from T cells is still exerted by the allograft, can be seen through the fact that patients with low TREC levels have a lower RFS (Fig. 4). In addition, most deaths in the low TREC group are due to relapse (Table 2). These data suggest that reconstitution of a functional immunity after HSCT is vital for a sustainable control over malignant MDS clones. Whether this is a direct effect exerted by the thymus-derived T cells or a consequence of functional hematopoiesis inhibiting the growth of malignant clones through space limitation is unclear. In this study, we have shown that TREC monitoring can be a powerful tool to predict relapse after ASCT, especially in patients with AML and MDS. Low TREC levels may be a future indication for early immunological interventions, for example, DLI or tapering of immunosuppression to decrease the risk of relapse.

7

9.

10.

11.

12.

Acknowledgments This study was supported by the Swedish Childhood Cancer Foundation, The Swedish Research Council (VR), the Swedish Society for Medical Research (SSMF), the Stockholm County Council, and Karolinska Institutet.

13.

Author Disclosure Statement The authors have no conflict of interests to declare.

References 1. Remberger M, M Ackefors, S Berglund, O Blennow, G Dahllof, A Dlugosz, K Garming-Legert, J Gertow, B Gustafsson, et al. (2011). Improved survival after allogeneic hematopoietic stem cell transplantation in recent years. A single-center study. Biol Blood Marrow Transplant 17:1688–1697. 2. Ringden O, H Karlsson, R Olsson, B Omazic and M Uhlin. (2009). The allogeneic graft-versus-cancer effect. Br J Haematol 147:614–633. 3. Horowitz MM, RP Gale, PM Sondel, JM Goldman, J Kersey, HJ Kolb, AA Rimm, O Ringden, C Rozman, et al. (1990). Graft-versus-leukemia reactions after bone marrow transplantation. Blood 75:555–562. 4. Zakrzewski JL, GL Goldberg, OM Smith and MR van den Brink. (2008). Enhancing T cell reconstitution after hematopoietic stem cell transplantation: a brief update of the latest trends. Blood Cells Mol Dis 40:44–47. 5. Toubert A, S Glauzy, C Douay and E Clave. (2012). Thymus and immune reconstitution after allogeneic hematopoietic stem cell transplantation in humans: never say never again. Tissue Antigens 79:83–89. 6. Mackall CL, CV Bare, LA Granger, SO Sharrow, JA Titus and RE Gress. (1996). Thymic-independent T cell regeneration occurs via antigen-driven expansion of peripheral T cells resulting in a repertoire that is limited in diversity and prone to skewing. J Immunol 156:4609–4616. 7. de Gast GC, LF Verdonck, JM Middeldorp, TH The, A Hekker, JA vd Linden, HA Kreeft and BJ Bast. (1985). Recovery of T cell subsets after autologous bone marrow transplantation is mainly due to proliferation of mature T cells in the graft. Blood 66:428–431. 8. Mackall CL, JA Punt, P Morgan, AG Farr and RE Gress. (1998). Thymic function in young/old chimeras: substantial

14.

15.

16.

17.

18.

19.

thymic T cell regenerative capacity despite irreversible ageassociated thymic involution. Eur J Immunol 28:1886– 1893. Sairafi D, J Mattsson, M Uhlin and M Uzunel. (2012). Thymic function after allogeneic stem cell transplantation is dependent on graft source and predictive of long term survival. Clin Immunol 142:343–350. Uhlin M, D Sairafi, S Berglund, S Thunberg, J Gertow, O Ringden, M Uzunel, M Remberger and J Mattsson. (2012). Mesenchymal stem cells inhibit thymic reconstitution after allogeneic cord blood transplantation. Stem Cells Dev 21:1409–1417. Przybylski GK, KA Kreuzer, W Siegert and CA Schmidt. (2007). No recovery of T-cell receptor excision circles (TRECs) after non-myeloablative allogeneic hematopoietic stem cell transplantation is correlated with the onset of GvHD. J Appl Genet 48:397–404. Clave E, M Busson, C Douay, R Peffault de Latour, J Berrou, C Rabian, M Carmagnat, V Rocha, D Charron, G Socie and A Toubert. (2009). Acute graft-versus-host disease transiently impairs thymic output in young patients after allogeneic hematopoietic stem cell transplantation. Blood 113:6477–6484. Castermans E, M Hannon, J Dutrieux, S Humblet-Baron, L Seidel, R Cheynier, E Willems, A Gothot, JF Vanbellinghen, et al. (2011). Thymic recovery after allogeneic hematopoietic cell transplantation with non-myeloablative conditioning is limited to patients younger than 60 years of age. Haematologica 96:298–306. Jimenez M, C Martinez, G Ercilla, E Carreras, A UrbanoIspizua, M Aymerich, N Villamor, N Amezaga, M Rovira, F Fernandez-Aviles and E Montserrat. (2006). Clinical factors influencing T-cell receptor excision circle (TRECs) counts following allogeneic stem cell transplantation in adults. Transpl Immunol 16:52–59. Clave E, V Rocha, K Talvensaari, M Busson, C Douay, ML Appert, C Rabian, M Carmagnat, F Garnier, et al. (2005). Prognostic value of pretransplantation host thymic function in HLA-identical sibling hematopoietic stem cell transplantation. Blood 105:2608–2613. Clave E, D Lisini, C Douay, G Giorgiani, M Busson, M Zecca, D Charron, ME Bernardo, A Toubert and F Locatelli. (2012). A low thymic function is associated with leukemia relapse in children given T-cell-depleted HLAhaploidentical stem cell transplantation. Leukemia 26: 1886–1888. Clave E, D Lisini, C Douay, G Giorgiani, M Busson, M Zecca, F Moretta, G Acquafredda, LP Brescia, F Locatelli and A Toubert. (2013). Thymic function recovery after unrelated donor cord blood or T-cell depleted HLAhaploidentical stem cell transplantation correlates with leukemia relapse. Front Immunol 4:54. Olerup O and H Zetterquist. (1992). HLA-DR typing by PCR amplification with sequence-specific primers (PCRSSP) in 2 hours: an alternative to serological DR typing in clinical practice including donor-recipient matching in cadaveric transplantation. Tissue Antigens 39:225–235. Ringden O, T Ruutu, M Remberger, J Nikoskelainen, L Volin, L Vindelov, T Parkkali, S Lenhoff, B Sallerfors, et al. (1994). A randomized trial comparing busulfan with total body irradiation as conditioning in allogeneic marrow transplant recipients with leukemia: a report from the Nordic Bone Marrow Transplantation Group. Blood 83:2723–2730.

8

20. Uzunel M, M Remberger, D Sairafi, Z Hassan, J Mattsson, B Omazic, L Barkholt and O Ringden. (2006). Unrelated versus related allogeneic stem cell transplantation after reduced intensity conditioning. Transplantation 82:913– 919. 21. Ringden O, M Remberger, S Carlens, H Hagglund, J Mattsson, J Aschan, B Lonnqvist, S Klaesson, J Winiarski, et al. (1998). Low incidence of acute graft-versus-host disease, using unrelated HLA-A-, HLA-B-, and HLA-DRcompatible donors and conditioning, including anti-T-cell antibodies. Transplantation 66:620–625. 22. Hazenberg MD, MC Verschuren, D Hamann, F Miedema and JJ van Dongen. (2001). T cell receptor excision circles as markers for recent thymic emigrants: basic aspects, technical approach, and guidelines for interpretation. J Mol Med 79:631–640. 23. Remberger M, O Ringden, H Hagglund, BM Svahn, P Ljungman, M Uhlin and J Mattsson. (2013). A high antithymocyte globulin dose increases the risk of relapse after reduced intensity conditioning HSCT with unrelated donors. Clin Transplant 27:E368–E374. 24. Ferrara JL and P Reddy. (2006). Pathophysiology of graftversus-host disease. Semin Hematol 43:3–10. 25. Blazar BR, WJ Murphy and M Abedi. (2012). Advances in graft-versus-host disease biology and therapy. Nat Rev Immunol 12:443–458. 26. Kolb HJ, C Schmid, AJ Barrett and DJ Schendel. (2004). Graft-versus-leukemia reactions in allogeneic chimeras. Blood 103:767–776. 27. Molldrem JJ, PP Lee, C Wang, K Felio, HM Kantarjian, RE Champlin and MM Davis. (2000). Evidence that specific T lymphocytes may participate in the elimination of chronic myelogenous leukemia. Nat Med 6:1018–1023. 28. Picker LJ, JR Treer, B Ferguson-Darnell, PA Collins, D Buck and LW Terstappen. (1993). Control of lymphocyte recirculation in man. I. Differential regulation of the peripheral lymph node homing receptor L-selectin on T cells during the virgin to memory cell transition. J Immunol 150:1105–1121. 29. Zlotoff DA, A Sambandam, TD Logan, JJ Bell, BA Schwarz and A Bhandoola. (2010). CCR7 and CCR9 together recruit hematopoietic progenitors to the adult thymus. Blood 115:1897–1905. 30. Komanduri KV, LS St John, M de Lima, J McMannis, S Rosinski, I McNiece, SG Bryan, I Kaur, S Martin, et al. (2007). Delayed immune reconstitution after cord blood transplantation is characterized by impaired thymopoiesis and late memory T-cell skewing. Blood 110:4543– 4551. 31. Weinberg K, BR Blazar, JE Wagner, E Agura, BJ Hill, M Smogorzewska, RA Koup, MR Betts, RH Collins and DC Douek. (2001). Factors affecting thymic function after allogeneic hematopoietic stem cell transplantation. Blood 97:1458–1466. 32. Hazenberg MD, SA Otto, ES de Pauw, H Roelofs, WE Fibbe, D Hamann and F Miedema. (2002). T-cell receptor excision circle and T-cell dynamics after allogeneic stem cell transplantation are related to clinical events. Blood 99:3449–3453. 33. Fallen PR, L McGreavey, JA Madrigal, M Potter, M Ethell, HG Prentice, A Guimaraes and PJ Travers. (2003). Factors affecting reconstitution of the T cell compartment in allogeneic haematopoietic cell transplant recipients. Bone Marrow Transplant 32:1001–1014.

UZUNEL ET AL.

34. Hochberg EP, AC Chillemi, CJ Wu, D Neuberg, C Canning, K Hartman, EP Alyea, RJ Soiffer, SA Kalams and J Ritz. (2001). Quantitation of T-cell neogenesis in vivo after allogeneic bone marrow transplantation in adults. Blood 98:1116–1121. 35. Klein AK, DD Patel, ME Gooding, GD Sempowski, BJ Chen, C Liu, J Kurtzberg, BF Haynes and NJ Chao. (2001). T-Cell recovery in adults and children following umbilical cord blood transplantation. Biol Blood Marrow Transplant 7:454–466. 36. Brown JA, K Stevenson, HT Kim, C Cutler, K Ballen, S McDonough, C Reynolds, M Herrera, D Liney, et al. (2010). Clearance of CMV viremia and survival after double umbilical cord blood transplantation in adults depends on reconstitution of thymopoiesis. Blood 115: 4111–4119. 37. Ishaqi MK, S Afzal, A Dupuis, J Doyle and A Gassas. (2008). Early lymphocyte recovery post-allogeneic hematopoietic stem cell transplantation is associated with significant graft-versus-leukemia effect without increase in graft-versus-host disease in pediatric acute lymphoblastic leukemia. Bone Marrow Transplant 41:245–252. 38. Thoma MD, TJ Huneke, LJ DeCook, ND Johnson, RA Wiegand, MR Litzow, WJ Hogan, LF Porrata and SG Holtan. (2012). Peripheral blood lymphocyte and monocyte recovery and survival in acute leukemia postmyeloablative allogeneic hematopoietic stem cell transplant. Biol Blood Marrow Transplant 18:600–607. 39. Wang Z, ML Sorror, W Leisenring, G Schoch, DG Maloney, BM Sandmaier and R Storb. (2010). The impact of donor type and ABO incompatibility on transfusion requirements after nonmyeloablative haematopoietic cell transplantation. Br J Haematol 149:101–110. 40. Blin N, R Traineau, S Houssin, R Peffault de Latour, A Petropoulou, M Robin, J Larghero, P Ribaud and G Socie. (2010). Impact of donor-recipient major ABO mismatch on allogeneic transplantation outcome according to stem cell source. Biol Blood Marrow Transplant 16:1315–1323. 41. Remberger M, E Watz, O Ringden, J Mattsson, A Shanwell and A Wikman. (2007). Major ABO blood group mismatch increases the risk for graft failure after unrelated donor hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 13:675–682. 42. Dulude G, DC Roy and C Perreault. (1999). The effect of graft-versus-host disease on T cell production and homeostasis. J Exp Med 189:1329–1342. 43. Wysoczanska B, K Bogunia-Kubik, D Dlubek, E Jaskula, A Sok, D Drabczak-Skrzypek, M Sedzimirska and A Lange. (2007). Association with the presence of naive T cells in chronic myeloid leukemia patients after allogeneic human stem cell transplantation and the lower incidence of chronic graft-versus host disease and relapse. Transplant Proc 39:2898–2901. 44. Mackinnon S, EB Papadopoulos, MH Carabasi, L Reich, NH Collins, F Boulad, H Castro-Malaspina, BH Childs, AP Gillio, et al. (1995). Adoptive immunotherapy evaluating escalating doses of donor leukocytes for relapse of chronic myeloid leukemia after bone marrow transplantation: separation of graft-versus-leukemia responses from graftversus-host disease. Blood 86:1261–1268. 45. Falkenburg JH, AR Wafelman, P Joosten, WM Smit, CA van Bergen, R Bongaerts, E Lurvink, M van der Hoorn, P Kluck, et al. (1999). Complete remission of accelerated phase

TREC LEVELS AS A PREDICTOR OF RELAPSE

chronic myeloid leukemia by treatment with leukemiareactive cytotoxic T lymphocytes. Blood 94:1201–1208. 46. Klimek VM. (2013). Recent advances in the management of therapy-related myelodysplastic syndromes and acute myeloid leukemia. Curr Opin Hematol 20:137–143. 47. Tobiasson M, R Olsson, E Hellstrom-Lindberg and J Mattsson. (2011). Early detection of relapse in patients with myelodysplastic syndrome after allo-SCT. Bone Marrow Transplant 46:719–726. 48. Campregher PV, T Gooley, BL Scott, C Moravec, B Sandmaier, PJ Martin, HJ Deeg, EH Warren and ME Flowers. (2007). Results of donor lymphocyte infusions for relapsed myelodysplastic syndrome after hematopoietic cell transplantation. Bone Marrow Transplant 40:965–971.

9

Address correspondence to: Michael Uhlin Center for Allogeneic Stem Cell Transplantation, B87 Karolinska University Hospital Huddinge SE-141 86 Stockholm Sweden E-mail: [email protected] Received for publication December 3, 2013 Accepted after revision March 10, 2014 Prepublished on Liebert Instant Online XXXX XX, XXXX

T-cell receptor excision circle levels after allogeneic stem cell transplantation are predictive of relapse in patients with acute myeloid leukemia and myelodysplastic syndrome.

In this retrospective study, 209 patients with malignant disease were analyzed for levels of T-cell receptor excision circles (TRECs) for the first 24...
444KB Sizes 0 Downloads 3 Views