CD4 + CD25 - CD69 + t Cells Is a Novel Subset of Regulatory T Cells Involved In the Prevention of Acute Graft-Versus-Host Disease In Human

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1250-1250 ◽  
Author(s):  
Sheng-Ye Lu ◽  
Xiao Jun Huang ◽  
Kai-Yan Liu ◽  
Dai-Hong Liu ◽  
Lan-Ping Xu

Abstract Abstract 1250 Acute graft-versus-host disease (aGVHD) remains the major obstacle to a more favorable therapeutic outcome of allogeneic hematopoietic stem cell transplantation (HSCT). Regulatory T cells (Tregs) play an important role in aGVHD prevention and a variety of Treg populations with distinct suppressive mechanism have been described in mice or human so far. In this study, we sought to identify Tregs associated with aGVHD in unmanipulated allogeneic HSCT in human by examining the frequency of immunophenotypes of three subsets of Tregs in periphery blood including CD4+CD25+Foxp3+, CD4+CD25+CD62L+ and CD4+CD25-CD69+ Tregs in three clinical patterns: First, we monitored 17 aGVHD patients along the course of disease and all of them got complete response (CR) to aGVHD finally. All samples were divided into three groups according to clinical status of aGVHD: before aGVHD-specefic treatment; partial response (PR); and CR, only CD4+CD25-CD69+ subset of Tregs showed significant changes, and the frequency decreases with the onset of aGVHD and increases after aGVHD recovery in peripheral blood, (Fig.1) furthermore, decreased frequency is associated with severity of aGVHD. Compared with healthy donor (HD), frequency of CD4+CD25-CD69+ Treg increased significantly even at the onset of aGVHD (Fig. 1), while frequency of CD4+CD25+Foxp3+ Treg decreased significantly after HSCT. Second, we compared the frequency of Tregs at +30day-post-HSCT between 16 post-HSCT patients who developed aGVHD and 31 patients who did not develop, only CD4+CD25-CD69+Treg had significantly lower percentages in patients with aGVHD (p=0.042), the other two subsets showed no significant difference; Third, we detected the association between Tregs in allografts and the incidence of aGVHD in 50 post-HSCT patients in an unbiased fashion, there were no differences between the aGVHD-group and No-aGVHD-group in the aspect of patients’ age and sex, disease status, stem cell and donor source, while there were significant difference between the two in the aspect of donors’ age, conditoning regimen and patient/donor HLA compatibility. We compared both the frequency and absolute number of the allograft components of patients with aGVHD and those without aGVHD, and absolute number of CD3+ T cell and CD4+CD25-CD69+Treg were statistically different, however, only CD4+CD25-CD69+Treg showed significant difference in multivariate Cox proportional hazards models including patients’ factors with significant difference and other factors closely associated with aGVHD. Then, we chose the median of frequency and absolute number of CD4+CD25-CD69+Treg as the borderline (1.08% and 1.34*106/kg, respectively) to identify the association between this subset in allografts and aGVHD. Both higher frequency and higher absolute number of CD4+CD25-CD69+ Tregs in allografts correlate with lower incidence of aGVHD by Kaplan-Meier, and log-rank test (Fig. 2). Moreover, our results show that CD4+CD25-CD69+Tregs expand significantly early after HSCT, the frequency at +30day reach the summit and then decrease gradually, and a low percentage of CD4+CD25-CD69+Tregs at +30day-post-HSCT is associated with aGVHD. The reconstitution of this Tregs early after HSCT is strongly associated with its component in allograft by spearman correlation analysis (r=0.449, p=0.003). Taken together, our findings suggest that CD4+CD25-CD69+T cell is a novel subset of regulatory T cell providing protection against aGVHD.Figure 1Frequency of CD4+CD25-CD69+ changes with the clinical status of aGVHD. Before treatment vs. PR P=0.003; before treatment vs. CR P= 0.015; PR vs CR P=0.289. Before treatment vs HD P= 0.041; PR vs. HD P= 0.000; CR vs HD P= 0.000.Figure 1. Frequency of CD4+CD25-CD69+ changes with the clinical status of aGVHD. Before treatment vs. PR P=0.003; before treatment vs. CR P= 0.015; PR vs CR P=0.289. Before treatment vs HD P= 0.041; PR vs. HD P= 0.000; CR vs HD P= 0.000.Figure 2The incidence of aGVHD after allogeneic HSCT increased in patients infused percentage of CD4+CD25-CD69+ Treg less than 1.08%.Figure 2. The incidence of aGVHD after allogeneic HSCT increased in patients infused percentage of CD4+CD25-CD69+ Treg less than 1.08%. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2234-2234
Author(s):  
San San Min ◽  
Varun Mehra ◽  
Jennifer Clay ◽  
Gemma Cross ◽  
Abdel Douiri ◽  
...  

Abstract Background: Acute Graft-versus-host disease (aGVHD) is a leading cause of morbidity and mortality following allogeneic haematopoietic stem cell transplantation (HSCT). The diagnosis and management of aGVHD relies on clinical criteria and graded according to the number and extent of organ involvement. Clinical diagnosis of aGVHD is one of exclusion and usually requires tissue biopsy to aid diagnosis. The aim of this study was to evaluate the clinical utility of a biomarker panel to help identify individuals at risk of developing aGVHD and its ability to differentiate severity at onset of acute graft-versus-host disease (aGVHD) following T-cell depleted allogeneic HSCT. Methods: This retrospective study of T-cell depleted reduced intensity allogeneic HSCT patients in our centre included 12 biopsy confirmed aGVHD patients (Grade I-IV; Modified Glucksberg criteria) and 14 patients as negative controls without any evidence of aGVHD, matched for age, sex, underlying haematological disease, time since transplant and conditioning regimen, within 100 days of transplantation. Serum samples on Day 0, Day +7 and at onset of aGVHD were analysed by ELISA for Elafin, regenerating islet-derived 3-α(REG3α), soluble tumour necrosis factor receptor 1 (sTNFR1), soluble interleukin-2 receptor-α (sIL-2Rα) and hepatocyte growth factor (HGF). Biomarker data was combined as composite panels A-F (Table 1) using binary and ordinal logistic regression analysis as best fit model. Receiver Operating Characteristic curves (ROC) analysis was performed to study sensitivity and specificity of the composite panel for diagnosis of aGVHD. Results: Composite biomarker panel B (Elafin+ sIL-2Rα + sTNFR1) and panel D (Elafin+ HGF + REG3α) at Day 0 and at Day 7 post-transplant differentiated significantly between biopsy confirmed severe aGVHD (Grade III-IV) and patients with no GVHD respectively (p=0.02). Panel F (Elafin+ sIL-2Rα + REG3α) strongly differentiated between various grades of aGVHD (Grade I-IV) at time of aGVHD onset (p<0.01), potentially predictive of disease severity. Composite panels A (Elafin+ sIL-2R + sTNFR1) and C (Elafin+ HGF + REG3) analysed in samples at Day 0 and Day +7 could not differentiate between patients with aGVHD (all grades) and no GVHD (p=0.54 and p=0.84 respectively), however Panel E (Elafin+ sIL-2Rα + REG3α) significantly differentiated between patients with no GVHD and biopsy confirmed aGVHD at time of onset of clinical aGVHD (p=0.02). This is suggestive of diagnostic utility of this composite biomarker panel in aGVHD diagnosis. The area under the curve (AUC) for this panel at time of onset was 0.65 with specificity, sensitivity, positive predictive value and negative predictive value of 100%, 55.6%, 100% and 78.9% respectively (p=0.03) Conclusions: This pilot data supports the usefulness of these composite biomarker panels in the prediction of severity and diagnosis of acute GVHD in patients undergoing T-cell depleted reduced intensity allogeneic HSCT. Further larger studies are needed to validate their use in the clinical management of aGVHD. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2002 ◽  
Vol 100 (10) ◽  
pp. 3479-3482 ◽  
Author(s):  
Joseph H. Antin ◽  
Daniel Weisdorf ◽  
Donna Neuberg ◽  
Roberta Nicklow ◽  
Shawn Clouthier ◽  
...  

Acute graft-versus-host disease (GVHD) is thought to derive from direct T-cell injury of target tissues through perforin/granzyme, Fas/FasL interactions, and the effects of inflammatory cytokines. Animal models and some clinical trials support the notion that inhibition of inflammatory mediators such as interleukin-1 (IL-1), tumor necrosis factor α, and interferon γ may ameliorate or prevent GVHD. We hypothesized that blockade of IL-1 during the period of initial T-cell activation would reduce the risk of severe GVHD. We tested this hypothesis in a double-blind, placebo-controlled randomized trial of recombinant human IL-1 receptor antagonist (IL-1Ra) in 186 patients undergoing allogeneic stem cell transplantation. Randomization was stratified by degree of histocompatibility and stem cell source. All patients were conditioned with cyclophosphamide and total body irradiation. GVHD prevention consisted of cyclosporine and methotrexate in all patients. Recombinant human IL-1Ra or saline placebo was given from day −4 to day +10. Randomization was stratified according to GVHD risk. The 2 groups were well-matched for pretreatment characteristics. Moderate to severe GVHD (grades B-D) developed in 57 (61%) of 94 patients receiving IL-1Ra and in 51 (59%) of 86 patients on placebo (P = .88). There was no difference in hematologic recovery, transplantation-related toxicity, event-free survival, or overall survival. We conclude that blockade of IL-1 using IL-1Ra during conditioning and 10 days immediately after transplantation is not sufficient to reduce GVHD or toxicity or to improve survival.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3742-3742
Author(s):  
LeShara M Fulton ◽  
Michael J Carlson ◽  
James Coghill ◽  
Michelle L. West ◽  
Angela Panoskaltisis-Mortari ◽  
...  

Abstract Abstract 3742 CD4+ T helper (Th) cells play a critical role in the development of Graft-versus-Host Disease (GvHD). The relative contributions of particular Th subsets to GVHD pathogenesis, however, are incompletely understood. In order to clarify the contribution of the Th17 subset to GVHD induction, we made use of mice knocked out at the RORgt locus (RORgt−/−), a transcription factor crucial for Th17 polarization. Methods: Haplotype matched and complete MHC mismatched murine HSCT models were used. For the haploidentical model C57BL/6 (H-2b, B6) mice served as donors while C57BL/6 × DBA2 F1 (H-2bxd, B6D2) mice functioned as recipients. Effector T cells (Teffs) were isolated from the spleens of wild type (WT) B6 and RORgt knockout mice backcrossed 7–8 generations onto a B6 background. B6D2 mice were lethally irradiated with 900 rads on day -1 and injected intravenously with 4 × 106 Teffs from WT or RORgt−/− mice supplemented with 3 × 106 WT T cell depleted bone marrow cells (TCD BM) on day 0. For the completely MHC mismatched model, BALB/c mice (H-2d) were lethally irradiated with 800 rads on day -1 and administered 5 × 105 WT or RORgt−/− Teffs supplemented with 5 × 106 B6 TCD BM on day 0. Results: B6D2 mice that received RORgt−/− Teffs displayed significantly attenuated GvHD, recovering from weight loss by day +31 and demonstrating 100% survival on day +60. Conversely, mice that received WT Teffs showed intense disease progression with 100% mortality by day +31 (Figure A, p<0.0001 for survival comparison between WT and RORgt−/− recipients using Fisher's exact test). Similar results were seen using the completely MHC mismatched model, with superior overall survival noted in those animals receiving RORgt −/− Teffs (put in p value here). Recipients of RORgt −/− T cells demonstrated statistically significant decreased TNF in serum compared to WT recipients (Figure B, p=0.001 comparing WT and RORgt−/− recipients using student's t test). Interestingly, despite the decreased severity of GvHD, serum concentrations of IFN-g were increased in recipients transplanted with RORgt −/− T cells. Chimerism studies post-transplant revealed complete donor reconstitution in recipients of both RORgt−/− and WT Teffs. Donor Teffs isolated from recipient livers post-transplant consistently demonstrated an activated phenotype, with low L selectin and high CD25 expression. Conclusions: T cell expression of the Th17 transcription factor, RORgt, is critical for the development of lethal GvHD following allogeneic stem cell transplantation in both the haploidentical and MHC complete mismatch models. GvHD attenuation in the absence of RORgt is not the result of an inability for donor T cells to undergo activation or to engraft in vivo. Interestingly, the absence of RORgt from donor T cells led to enhanced IFN-g in serum. Thus, in vivo, the Th17 pathway is critical for the induction of GvHD. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2003 ◽  
Vol 102 (2) ◽  
pp. 734-739 ◽  
Author(s):  
Anke Franzke ◽  
Wenji Piao ◽  
Jörg Lauber ◽  
Patricia Gatzlaff ◽  
Christian Könecke ◽  
...  

Abstract Results from experimental models, in vitro studies, and clinical data indicate that granulocyte colony-stimulating factor (G-CSF) stimulation alters T-cell function and induces Th2 immune responses. The immune modulatory effect of G-CSF on T cells results in an unexpected low incidence of acute graft-versus-host disease in peripheral stem cell transplantation. However, the underlying mechanism for the reduced reactivity and/or alloreactivity of T cells upon G-CSF treatment is still unknown. In contrast to the general belief that G-CSF acts exclusively on T cells via monocytes and dendritic cells, our results clearly show the expression of the G-CSF receptor in class I– and II– restricted T cells at the single-cell level both in vivo and in vitro. Kinetic studies demonstrate the induction and functional activity of the G-CSF receptor in T cells upon G-CSF exposure. Expression profiling of T cells from G-CSF–treated stem cell donors allowed identification of several immune modulatory genes, which are regulated upon G-CSF administration in vivo (eg, LFA1-α, ISGF3-γ) and that are likely responsible for the reduced reactivity and/or alloreactivity. Most importantly, the induction of GATA-3, the master transcription factor for a Th2 immune response, could be demonstrated in T cells upon G-CSF treatment in vivo accompanied by an increase of spontaneous interleukin-4 secretion. Hence, G-CSF is a strong immune regulator of T cells and a promising therapeutic tool in acute graft-versus-host disease as well as in conditions associated with Th1/Th2 imbalance, such as bone marrow failure syndromes and autoimmune diseases.


Sign in / Sign up

Export Citation Format

Share Document