Emergence of Donor T Cells That Recognize Nonpolymorphic Antigens During Graft Versus Host Disease,

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4013-4013
Author(s):  
Hemalatha Rangarajan ◽  
Maryam Yassai ◽  
Xiao Chen ◽  
Richard Komorowski ◽  
Jack Gorski ◽  
...  

Abstract Abstract 4013 Chronic graft versus host disease (GVHD) is a major cause of morbidity and mortality in allogeneic stem cell transplant recipients and typically develops from antecedent acute GVHD. In contrast to acute GVHD which is characterized by a restricted set of organ involvement, chronic GVHD has more protean manifestations with much broader tissue involvement that can affect nearly all organ sites. Moreover, clinical manifestations in chronic GVHD often bear striking similarity to what is observed in autoimmune diseases. How GVHD evolves from an acute inflammatory syndrome in which donor T cells recognize polymorphic antigens expressed by host APCs to one that is characterized by autoimmune manifestations is not well understood. More specifically, it is not clear whether T cells that mediate autoimmune manifestations during chronic GVHD continue to respond to polymorphic antigens or whether T cells that recognize nonpolymorphic antigens emerge in these recipients. To address this issue, we performed a comprehensive analysis of the clonotypic T cell response in mice that developed autoimmune-mediated GVHD. To conduct these studies, lethally irradiated Balb/c [H-2d] mice were transplanted with C57BL/6 [H-2b] bone marrow and spleen cells to induce acute GVHD. At 19–21 days post transplantation, spleen cells or purified T cells from completely donor-engrafted mice were transferred into non irradiated syngeneic B6 Rag animals. In this model, animals develop pathological damage in the colon 60–70 days post transfer characterized by lamina propria inflammation, goblet cell depletion, and crypt cell destruction. This is attributed to the presence of autoreactive donor T cells in the original spleen cell inoculum that expand in syngeneic recipients due to loss of effective T cell regulation. To examine the clonotypic T cell response, we performed T cell receptor beta spectratyping on pathologically involved colonic tissue to identify over represented, skewed bands that were shared by replicate mice within the 21 Vβ families. These bands within a given Vβ family were sequenced to define the specific T cell clonotypes within colitic tissue. In the vast majority of families across multiple experiments, there were high frequency clonotypes that were present in all replicate mice and comprised 50–90% of all sequences. Notably, these shared clonotypes between replicate animals had the same CDR3 nucleotide sequence, indicating that they were the same T cell clones. The presence of high frequency clonotypes was also evidence that autoimmunity was characterized by antigen-driven expansion of a limited number of clones. During the progression from acute to chronic GVHD in humans, host APCs are eliminated and presentation of host peptides is by donor APCs through the indirect alloreactive pathway. To simulate this condition in our experimental model, we created chimeric animals by transplanting B6 Rag BM cells into lethally irradiated Balb/c Rag mice. Spleen cells from primary B6→Balb/c GVHD mice were then transferred into these fully donor-engrafted chimeric animals. Clonotypic analysis of T cells obtained from replicate mice with colitis revealed that dominant clonotypes were observed and comprised 65–80% of all sequences, similar to what was observed during conditions of autoimmunity. Given these similar results in models of autoimmunity and indirect alloimmunity, we then determined whether T cells that were capable of responding to B6 and Balb/c antigens when presented by B6 APCs could be identified. To address this question, the same pooled spleen cell suspension from primary (B6→Balb/c) GVHD mice was transferred into B6 Rag and chimeric (B6 Rag→Balb/c Rag) animals. We observed that T cell clonotypes with the same nucleotide sequences could be identified in pathologically involved colon tissue from both cohorts of mice indicating the emergence of T cells that were capable of recognizing both B6 and Balb/c antigens presented by B6 APCs. These results demonstrate that the loss of self tolerance which occurs during acute GVHD leads to the emergence of broadly reactive donor T cells that are capable of recognizing nonpolymorphic antigens that are shared between donor and host. These data also provide a mechanistic explanation for how autoimmunity develops as a consequence of GVHD and help explain the progression of tissue involvement that characterizes the transition from acute to chronic GVHD. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 598-598
Author(s):  
William R. Drobyski ◽  
Richard Komorowski ◽  
Elizabeth Tivol

Abstract The progression of acute to chronic graft versus host disease (GVHD) is characterized by the development of clinical manifestations that bear close resemblance to what is observed in autoimmune disorders. A longstanding unresolved issue is how acute GVHD which is initiated by alloreactive donor T cells evolves into chronic GVHD where autoreactive donor T cells appear to play a major role in the pathophysiology of this syndrome. We hypothesized that during the course of GVHD, there is breaking of tolerance such that donor T cells acquire the ability to recognize self antigens. To test this hypthesis, we examined the ability of donor T cells obtained from mice undergoing GVHD to respond to both donor and recipient antigens using an adoptive transfer model. Lethally irradiated Balb/c [H-2d] mice were transplanted with BM and spleen cells from MHC-mismatched C57BL/6 (B6)[H-2b] mice to induce severe GVHD. Spleen cells from fully donor T cell engrafted GVHD mice were then adoptively transferred into unirradiated B6 or Balb Rag animals. Adoptive transfer of GVHD spleen cells into B6-Rag animals resulted in either colitis or pulmonary inflammation in a >90% of recipients. Colitis was characterized by infiltration of mononuclear and granulocytic cells into the lamina propria with transmural extension and mucosal sloughing, while inflammation in the lung was characterized by pronounced perivascular and peribronchial mononuclear cell cuffing accompanied by areas of extensive pneumonitis.Immunohistochemical staining demonstrated significant T cell infiltration into damaged tissue in both the colon and lungs of affected animals. Neither Balb Rag nor third party C3H-Rag mice, however, exhibited pathological damage after adoptive transfer of the same spleen cell population. Administration of asialo GM1 to deplete host NK cells did not effect any histological abnormalities in Balb Rag mice, indicating that failure to observe pathology in these mice was not due to NK-mediated rejection of donor T cells. Autoreactivity could also be serially passaged through B6 Rag but not Balb Rag mice demonstrating that tolerance could not be reestablished by exposure to donor antigens in a different environmental milieu. To exclude the possibility that the observed pathological lesions were model dependent, similar transplant studies were performed in B6[H-2b]→B10.BR[H-2k] chimeras. Transfer of GVHD spleen cells into B6 Rag mice recapitulated the pathology observed in recipients of B6→Balb spleen cells. Independent experiments showed that transplantation of GVHD spleen cells into irradiated B6 as opposed to B6 Rag mice failed to induce any pathology, suggesting that a radiosensitive cell in host animals was necessary for propagation of autoreactivity. To determine if this cell was an antigen presenting cell (APC), Balb Rag mice were transplanted with B6 Rag BM to create chimeric animals where APCs were of donor origin. GVHD spleen cells were then transferred into chimeric mice where the same pathological lesions were observed as in B6 Rags. We conclude that GVHD results in the breaking of tolerance against self such that donor T cells acquire the ability to respond to both self and alloantigens but only within the context of donor APCs. These data suggest a new paradigm to GVHD pathophysiology whereby progression of GVHD is dependent upon donor APCs and provides an explanation for why chronic GVHD is characterized by autoimmune manifestations in man.


Blood ◽  
2010 ◽  
Vol 116 (2) ◽  
pp. 287-296 ◽  
Author(s):  
Geoffrey R. Hill ◽  
Rachel D. Kuns ◽  
Neil C. Raffelt ◽  
Alistair L. J. Don ◽  
Stuart D. Olver ◽  
...  

Abstract Suppressor of cytokine signaling-3 (SOCS3) is the main intracellular regulator of signaling by granulocyte colony-stimulating factor, an immune-modulatory cytokine used to mobilize stem cells for transplantation. We have therefore studied the contribution of SOCS3 to the spectrum of graft-versus-host disease (GVHD) after allogeneic stem cell transplantation (SCT). Grafts from SOCS3−/Δvav donor mice in which SOCS3 deficiency is restricted to the hematopoietic compartment had an augmented capacity to induce acute GVHD. With the use of SOCS3−/ΔLysM and SOCS3−/Δlck donors in which SOCS3 deficiency was restricted to the myeloid or T-cell lineage, respectively, we confirmed SOCS3 deficiency promoted acute GVHD mortality and histopathology within the gastrointestinal tract by effects solely within the donor T cell. SOCS3−/Δlck donor T cells underwent enhanced alloantigen-dependent proliferation and generation of interleukin-10 (IL-10), IL-17, and interferon-γ (IFNγ) after SCT. The enhanced capacity of the SOCS3−/Δlck donor T cell to induce acute GVHD was dependent on IFNγ but independent of IL-10 or IL-17. Surprisingly, SOCS3−/Δlck donor T cells also induced severe, transforming growth factor β– and IFNγ-dependent, sclerodermatous GVHD. Thus, the delivery of small molecule SOCS3 mimetics may prove to be useful for the inhibition of both acute and chronic GVHD.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 245-245
Author(s):  
Nicole Stauffer ◽  
Sayed Mehdi Hamadani ◽  
Catherine Heaphy ◽  
Ramasamy Santhanam ◽  
Sukhinder Sandhu ◽  
...  

Abstract Abstract 245 Acute Graft-versus-host disease (aGVHD) is a frequent and lethal complication of allogeneic hematopoietic stem cell transplantation (HSCT) in which donor T cells destroy HLA mismatched host tissues by secreting soluble inflammatory cytokines (TNF-α, IFN-γ) and/or inducing direct cytotoxic cellular responses. Despite recent advances, GVHD still remains a major clinical problem, underscoring the need to elucidate further its mechanisms to then develop novel therapeutic strategies. Recent studies indicate that microRNAs (miR) play critical roles in the development and function of the immune system. In particular, miR-155 is required for normal function of B and T lymphocytes. MiR-155 has been shown to be up-regulated upon B and T cell activation and mice deficient for miR-155 are immunodeficient, exhibiting T cells with attenuated INF-γ and TNF-α release in response to antigen stimulation. Based on these observations, we hypothesized that miR-155 is up-regulated in donor T cells during aGVHD and is involved in the modulation of this process. To test this hypothesis, we initially measured miR-155 expression from activated CD8 T cells obtained from an experimental animal model of aGVHD. For this, a major histocompatibility complex (MHC) mismatched HSCT model was used in which spleen cells and BM from C57BL/6 (B6) donors were transferred i.v. into lethally irradiated B6D2F1 recipient mice (n=4). Two additional groups were included as controls with one group receiving no cell infusion (radiation only, n=4); and a second group receiving T cell depleted bone marrow only (t-BM) (5×106) (n=4). GVHD scores were performed daily according to Cooke et al, Blood 1996;8:3230-9. Recipient mice were sacrificed and tissues harvested when GVHD scores were ≥7 or at the end of the experiment. After an average of 3–4 weeks, mice receiving donor spleen cells but not BM alone developed severe GVHD (scores >7) which was confirmed by histology. Activated CD8 T cells from the spleen were isolated using CD8+/CD44+ antibodies and after obtaining total RNA, miR-155 expression was measured by RT-PCR. Mice receiving donor BM plus spleen cells developed severe aGVHD and exhibited increased miR-155 expression with respect to the BM only group (fold change 5.2, t-test p<0.001). To confirm that a causal relationship exists between miR-155 and aGVHD severity, we repeated the above MHC mismatched murine experiment using BL/6 mice deficient for miR-155 expression as donors (Rodriguez et al, Science 2007;316;608-611). The groups were as follows; radiation alone (n=4), t-BM (n=8), BM + wild type (WT) spleen cells (n=8), and t-BM+ miR-155 KO spleen cells (n=8). We found that mice receiving donor spleen cells from miR-155 KO mice exhibited dramatically lower mean GVHD scores (3 Vs. 5.5, t-test p=0.0007) and improved survival compared to those receiving WT spleen cells(87% vs. 13% of mice alive at 70 days, respectively; log-rank test p<0.001). Our results showed that recipients from miR-155 KO spleen cells did not exhibit high GVHD histological scores (III-IV) in the spleen, liver or gut, while 80%, 60% and 40% of the WT did. Overall survival, GVHD scores and histological GHVD findings were similar between miR-155 KO and WT t-BM only group. Since high levels of soluble TNF-α is characteristic for aGVHD, we measured serum TNF-α by ELISA assay in mice at the time of harvest. Mice receiving miR-155 KO spleen cells had significantly lower TNF-α levels (mean 14 pg/ml) than WT controls (mean 48 pg/ml, t-test, p=0.005). Finally, to establish the relevance of this finding to the human system, we measured miR-155 expression using Locked Nucleic Acid in situ hybridization from histologically confirmed gut biopsies of aGVHD patients (n=5), and healthy controls (n=3). We found a strong up-regulation of miR-155 expression in all patients with gut aGVHD while miR-155 expression was absent in normal gut. In summary, we have shown that miR-155 expression is up-regulated in CD8 T cells from mice with aGVHD and showed that mice receiving donor lymphocyte cells deficient for miR-155 exhibited less GVHD and improved survival as compared to mice receiving WT donor lymphocytes. Finally, up-regulation of miR-155 was also found in clinical specimens from patients with gut aGVHD. Altogether our data indicate a role for miR-155 in the modulation of aGVHD, and thus point to miR-155 as a novel target for therapeutic intervention in aGVHD. Disclosures: Off Label Use: Decitabine and bortezomib in AML.


2001 ◽  
Vol 194 (10) ◽  
pp. 1433-1440 ◽  
Author(s):  
Pavan Reddy ◽  
Takanori Teshima ◽  
Mark Kukuruga ◽  
Rainer Ordemann ◽  
Chen Liu ◽  
...  

Interleukin (IL)-18 is a recently discovered cytokine that modulates both T helper type 1 (Th1) and Th2 responses. IL-18 is elevated during acute graft-versus-host disease (GVHD). We investigated the role of IL-18 in this disorder using a well characterized murine bone marrow transplantation (BMT) model (B6 → B6D2F1). Surprisingly, blockade of IL-18 accelerated acute GVHD-related mortality. In contrast, administration of IL-18 reduced serum tumor necrosis factor (TNF)-α and lipopolysaccharide (LPS) levels, decreased intestinal histopathology, and resulted in significantly improved survival (75 vs. 15%, P &lt; 0.001). Administration of IL-18 attenuated early donor T cell expansion and was associated with increased Fas expression and greater apoptosis of donor T cells. The administration of IL-18 no longer protected BMT recipients from GVHD when Fas deficient (lpr) mice were used as donors. IL-18 also lost its ability to protect against acute GVHD when interferon (IFN)-γ knockout mice were used as donors. Together, these results demonstrate that IL-18 regulates acute GVHD by inducing enhanced Fas-mediated apoptosis of donor T cells early after BMT, and donor IFN-γ is critical for this protective effect.


Blood ◽  
2021 ◽  
Author(s):  
Qingxiao Song ◽  
Xiaoning Wang ◽  
Xiwei Wu ◽  
Hanjun Qin ◽  
Yingfei Li ◽  
...  

Donor T cells mediate both graft-versus-leukemia (GVL) activity and graft-versus-host disease (GVHD) after allogeneic hematopoietic cell transplantation (Allo-HCT). Development of methods that preserve GVL activity while preventing GVHD remains a long-sought goal. Tolerogenic anti-IL-2 monoclonal antibody (mAb) (JES6-1) forms anti-IL-2/IL-2 complexes that block IL-2 binding to IL-2Rb and IL-2Rg on Tcon cells that have low expression of IL-2Rα. Here we show that administration of JES6 early after Allo-HCT in mice markedly attenuates acute GVHD while preserving GVL activity that is dramatically stronger than observed with tacrolimus (TAC) treatment. The anti-IL-2 treatment down-regulated activation of IL-2-Stat5 pathway and reduced production of GM-CSF. In GVHD target tissues, enhanced T cell PD-1 interaction with tissue-PD-L1 led to reduced activation of AKT-mTOR pathway and increased expression of Eomes and Blimp-1, increased T cell anergy/exhaustion, expansion of Foxp3-IL-10-producing Tr1 cells, and depletion of GM-CSF-producing Th1/Tc1 cells. In recipient lymphoid tissues, lack of donor T cell PD-1 interaction with tissue-PD-L1 preserved donor PD-1+TCF-1+Ly108+CD8+ T memory progenitors (Tmp) and functional effectors that have strong GVL activity. Anti-IL-2 and TAC treatments have qualitatively distinct effects on donor T cells in the lymphoid tissues, and CD8+ Tmp cells are enriched with the anti-IL-2 treatment compared to TAC treatment. We conclude that administration of tolerogenic anti-IL-2 mAb early after Allo-HCT represents a novel approach for preventing acute GVHD while preserving GVL activity.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 5237-5237 ◽  
Author(s):  
Guenther Richter ◽  
Andreas Mollweide ◽  
Esther Uhlmann ◽  
Stefan Burdach

Abstract Inducible costimulator (ICOS) interaction with its ligand (ICOSL) is involved in several T cell effector functions. The abundant expression of ICOSL in a variety of target tissues of acute graft versus host disease (GVHD) provided the rationale to investigate its role in acute GVHD development. C57BL/6 mice were lethally irradiated and reconstituted with allogeneic spleen cells in the absence or presence of ICOSL-blocking reagents. Mice reconstituted with allogeneic spleen cells experienced 12–16% weight loss around day 4 after transplantation and died untreated of acute GVHD within 7–10 days after transplantation. Mice treated from day 3 after transplantation with an anti-ICOSL mAb gained weight again and survived for additional 18 days, although the treatment was already stopped at day 11 after transplantation. Such mice revealed less T cells in spleen at day 4 after transplantation with reduced effector activity. In contrast, the anti-ICOSL treatment starting from day 0 did not prevent GVHD. The difference between therapeutic (day 3) and prophylactic (day 0) anti-ICOSL treatment was independent of CD25+CD4+ regulatory T cells since their depletion did not abrogate the therapeutic effect of ICOSL blockade. Similarly, depletion of NK cells did not improve prophylactic anti-ICOSL treatment. However, our results clearly show that delayed, i.e. therapeutic blockade of ICOS:ICOSL interaction can interfere with acute GVHD development and alleviates disease significantly in a polyclonal T cell setting.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 728-728 ◽  
Author(s):  
Yu Yu ◽  
Dapeng Wang ◽  
Kenrick Semple ◽  
Claudio Anasetti ◽  
Xue-Zhong Yu

Abstract Abstract 728 Background: Allogeneic hemopoietic stem cell transplantation (HCT) is an effective therapy with potential cure of hematological malignancies through T cell-mediated graft-versus-leukemia (GVL) effects. However, beneficial GVL effects are frequently offset by the development of destructive graft-versus-host disease (GVHD) also induced by donor T cells. Recent studies including ours have demonstrated that donor T cells differentiated into type 1 or type 17-subset contribute to GVHD. Thus, we hypothesize that blocking both Th1 and Th17 lineage via disrupting Th1-specific (T-bet) and Th17-specific (RORγt) transcription factors can significantly reduce GVHD after allo-HCT. Method: Two murine models of bone marrow transplantation (BMT) that represent clinical GVHD and GVL were used: C57BL/6 (B6)→BALB/c and B6→(B6 × DBA2)F1. To mimic clinical residual hematological malignant disease, B cell lymphoma (A20) and mastocytoma (p815) were infused into BALB/c and (B6 × DBA2)F1 mice, respectively. Results: We first compared the ability of WT, T-bet−/−, RORγt−/−, and T-bet−/−/RORγt−/− T cells in the induction of GVHD, and found that RORγt−/− T cells had a comparable ability to cause GVHD as WT T cells, whereas T-bet−/− T cells were less pathogenic. The T-bet−/−/RORγt−/− T cells failed to induce acute GVHD but caused minor to modest chronic GVHD in some of recipients at the doses tested. To investigate whether recipients of T-bet−/−/RORγt−/− T cells had less severe target organ GVHD damage, we analyzed GVHD associated organ damage in liver, lung and bowel. Fourteen days after adoptive transfer of WT, T-bet−/−, RORγt−/−, and T-bet−/−/RORγt−/− T cells, recipients which received T-bet−/−/RORγt−/− donor T cells showed markedly reduced T cell infiltration and tissue damage in liver, lung, and bowel. Mechanistic studies revealed that T-bet−/−/RORγt−/− T cells produced significantly less IFNγ (Th1 cytokine) and IL-17 (Th17-cytokine) but significantly more IL-4 and IL-5 (Th2-cytokines) as compared to WT T cells. In addition, T-bet−/−/RORγt −/− donor T-cells express significantly less CXCR3 and CCR6, chemokine receptors required for infiltration of alloreactive T cells into GVHD targeted organ, which could be the reason that significantly fewer T-bet−/−/RORγt−/− T cells were accumulated in recipient liver and lung than WT T cells. Furthermore, we tested the ability of WT and T-bet−/−/RORγt−/− T cells in mediating GVL effect. Although T-bet−/−/RORγt−/− T cells failed to induce acute GVHD, their ability to reject A20 or p815 cells was comparable to that of the WT T cells at the dose tested. Conclusions: These results indicate that blocking T-bet and RORγt prevents acute GVHD by suppressing donor T cell differentiation towards Th1 and Th17 and promoting differentiation towards Th2, and inhibiting donor T cell expansion and infiltration into GVHD target organs. Furthermore, blocking T-bet and RORγt could preserve GVL effect. Thus, the current study validates new targets for the separation of donor T cell–mediated GVHD and GVL activity, which could eventually be beneficial to patients with hematological malignancies. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3045-3045
Author(s):  
Rie Kuroda ◽  
Hideaki Maeba ◽  
Shintaro Mase ◽  
Raita Araki ◽  
Toshihiro Fujiki ◽  
...  

Abstract Abstract 3045 We have reported that measuring the activation markers and homing molecules on T-cells obtained from human peripheral blood (PB) samples provides useful information for predicting acute graft-versus-host disease (GVHD) severity in affected organs. Although T-cells are major player for developing GVHD, rodent GVHD studies have demonstrated that other immune cells such as monocytes/macrophages, B-cells, and mast cells involved in the pathogenesis of acute and chronic GVHD. We, therefore, evaluated activation markers and homing molecules regularly not only on T-cells but also on monocytes in PB obtained from 31 childhood patients (more than 600 samples at various time points) receiving hematopoietic stem cell transplantation (HSCT) by multicolor flow cytometry. The following markers were used: CD69, CD25, and HLA-DR for T cell activation, CCR4, CCR5, CXCR3, CCR9, and CLA for homing markers. Inflammatory monocytes were defined as CD14dimCD16+ cells or CD14+CD163+ cells. In addition we combined the data of cytokine profiles secreted mainly by T cells such as soluble interleukin 2 receptor, or monocytes such as neopterin, or both such as tumor necrosis factor-α (TNF-α), soluble TNF-αRI, and soluble TNF-αRII. In all cases showing acute GVHD, both T-cell and monocyte activation markers were elevated. Only either T-cell or monocyte activation was not observed in acute GVHD cases. However, we have some interesting results classified according to the status of T-cells and monocytes after day 100: 1) In the cases both T-cells and monocytes were highly activated as shown in Figure 1A, tapering of immunosuppressants led to exacerbation of GVHD, and prolonged administration of the drugs including steroids were needed. However steroid response itself was relatively good. 2) In the cases only T-cells, much less monocytes, were activated as shown in Figure 1B, calcineurin inhibitors were quite effective in improving GVHD. 3) In the cases of sustained chronic GVHD, neither T-cells nor mononytes were activated as shown in Figure 1C. Response to immunosuppressants was quite low. 4) When CD4 T-cell repertoire, not CD8, became normal, tapering of drugs was successful. In all cases successfully tapered immunosuppressive drugs, elevated activation markers of T-cells and monocytes completely returned to same levels of normal volunteers. In conclusion, evaluating the activation markers and homing molecules not only on T-cells but also on monocytes, combined with cytokine profiling, might provide useful information for management of patients with prolonged GVHD. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (1) ◽  
pp. 9-17 ◽  
Author(s):  
Ronjon Chakraverty ◽  
Megan Sykes

After allogeneic blood or bone marrow transplantation, donor T cells interact with a distorted antigen-presenting cell (APC) environment in which some, but not all, host APCs are replaced by APCs from the donor. Significantly, host APCs are required for the priming of acute graft-versus-host disease (GVHD). Donor APCs play a lesser role in the induction of acute GVHD despite their predicted capacity to cross-present host antigens. In contrast, donor APCs may play a role in perpetuating the tissue injury observed in chronic GVHD. Host APCs are also required for maximal graft-versus-leukemia responses. Recent studies have suggested potential strategies by which the continued presence of host APCs can be exploited to prime strong donor immunity to tumors without the induction of GVHD.


Blood ◽  
2000 ◽  
Vol 95 (12) ◽  
pp. 3693-3701 ◽  
Author(s):  
Ypke V. J. M. van Oosterhout ◽  
Liesbeth van Emst ◽  
Anton V. M. B. Schattenberg ◽  
Wil J. M. Tax ◽  
Dirk J. Ruiter ◽  
...  

Abstract This study evaluated the anti-graft versus host disease (GVHD) potential of a combination of immunotoxins (IT), consisting of a murine CD3 (SPV-T3a) and CD7 (WT1) monoclonal antibody both conjugated to deglycosylated ricin A. In vitro efficacy data demonstrated that these IT act synergistically, resulting in an approximately 99% elimination of activated T cells at 10−8 mol/L (about 1.8 μg/mL). Because most natural killer (NK) cells are CD7+, NK activity was inhibited as well. Apart from the killing mediated by ricin A, binding of SPV-T3a by itself impaired in vitro cytotoxic T-cell cytotoxicity. Flow cytometric analysis revealed that this was due to both modulation of the CD3/T-cell receptor complex and activation-induced cell death. These results warranted evaluation of the IT combination in patients with refractory acute GVHD in an ongoing pilot study. So far, 4 patients have been treated with 3 to 4 infusions of 2 or 4 mg/m2 IT combination, administered intravenously at 48-hour intervals. The T1/2 was 6.7 hours, and peak serum levels ranged from 258 to 3210 ng/mL. Drug-associated side effects were restricted to limited edema, fever, and a modest rise of creatine kinase levels. One patient developed low-titer antibodies against ricin A. Infusions were associated with an immediate drop of circulating T cells, followed by a more gradual but continuing elimination of T/NK cells. One patient mounted an extensive CD8 T-cell response directly after treatment, not accompanied with aggravating GVHD. Two patients showed nearly complete remission of GVHD, despite unresponsiveness to the extensive pretreatment. These findings justify further investigation of the IT combination for treatment of diseases mediated by T cells.


Sign in / Sign up

Export Citation Format

Share Document