scholarly journals Inhibition of ITK differentiates GVT and GVHD in allo-HSCT

Author(s):  
Mahinbanu Mammadli ◽  
Weishan Huang ◽  
Rebecca Harris ◽  
Aisha Sultana ◽  
Ying Cheng ◽  
...  

AbstractAllogeneic hematopoietic stem cell transplantation is a life-saving treatment for many malignant and nonmalignant diseases. Donor T cells contained within the graft prevent tumor recurrence via graft-versus-tumor (GVT) effects, however, also cause graft-versus-host disease (GVHD). Novel treatment strategies are therefore needed to allow maintenance of GVT while suppressing GVHD. Here we show using murine models, that targeting IL-2-inducible T cell kinase (ITK) in donor T cells reduces GVHD while preserving the beneficial GVT effects. Donor T cells from Itk-/- mice exhibit significantly reduced production of inflammatory cytokines and migration to GVHD target organs such as liver and small intestine, while maintaining GVT efficacy against primary B-ALL tumors. Itk-/- T cells exhibited reduced expression of IRF4 and decreased JAK/STAT signaling activity, but preserved cytotoxicity, which was accompanied by upregulation of Eomesodermin (Eomes), which was necessary for GVT function. A novel peptide inhibitor ITK signaling is also able to prevent GVHD. This novel peptide inhibitor also reduced cytokine production in mice and human T cells. Altogether, our data suggest that inhibiting ITK could be a therapeutic strategy to reduce GVHD while preserving the beneficial GVT effects following allo-HSCT treatment.Key PointsInhibiting ITK by a novel peptide significantly reduces GVHD but retains GVT.ITK deficient donor T cells exhibit minimal GVHD, but maintain GVT activity.ITK deficient donor T cells exhibit significantly reduced production of inflammatory cytokines and migration to GVHD target organs.Eomes is required for GVT effect.

2020 ◽  
Vol 11 ◽  
Author(s):  
Mahinbanu Mammadli ◽  
Weishan Huang ◽  
Rebecca Harris ◽  
Aisha Sultana ◽  
Ying Cheng ◽  
...  

Allogeneic hematopoietic stem cell transplantation is a potentially curative procedure for many malignant diseases. Donor T cells prevent disease recurrence via graft-versus-leukemia (GVL) effect. Donor T cells also contribute to graft-versus-host disease (GVHD), a debilitating and potentially fatal complication. Novel treatment strategies are needed which allow preservation of GVL effects without causing GVHD. Using murine models, we show that targeting IL-2-inducible T cell kinase (ITK) in donor T cells reduces GVHD while preserving GVL effects. Both CD8+ and CD4+ donor T cells from Itk-/- mice produce less inflammatory cytokines and show decrease migration to GVHD target organs such as the liver and small intestine, while maintaining GVL efficacy against primary B-cell acute lymphoblastic leukemia (B-ALL). Itk-/- T cells exhibit reduced expression of IRF4 and decreased JAK/STAT signaling activity but upregulating expression of Eomesodermin (Eomes) and preserve cytotoxicity, necessary for GVL effect. Transcriptome analysis indicates that ITK signaling controls chemokine receptor expression during alloactivation, which in turn affects the ability of donor T cells to migrate to GVHD target organs. Our data suggest that inhibiting ITK could be a therapeutic strategy to reduce GVHD while preserving the beneficial GVL effects following allo-HSCT treatment.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4011-4011
Author(s):  
Ji-Young Lim ◽  
Gyeongsin Park ◽  
Hyewon Youn ◽  
Eun-Young Choi ◽  
Dae-Chul Jeong ◽  
...  

Abstract Abstract 4011 Graft-versus-host disease (GVHD) is a common complication after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells with anti-inflammatory activity. MyD88 is a cytoplasmic adaptor molecule essential for integrating and transducing the signals generated by the toll-like receptor (TLR) family. Activation of inflammatory signaling through MyD88, presumably through ligation of multiple TLRs, plays a key role in the expansion of MDSCs. We therefore investigated how the MyD88-dependent expansion of MDSCs from donor bone marrow (BM) contributes to protection of acute GVHD. To test this, we employed an intestinal GVHD murine model, C57BL/6 (H-2b) → B6D2F1 (H-2b/d), which differs at major and minor histocompatibility loci. Lethally irradiated recipient mice were transplanted with wild-type (WT) or MyD88 knock out (KO) mice T cell-depleted (TCD)-BM together with WT spleen T cells. Morbidity and mortality of GVHD was significantly worse in recipients of MyD88 KO TCD-BM with higher intestinal pathologic grading. Animals that underwent syngeneic HSCT did not show early mortality regardless of presence of MyD88 in BM, which ruled out myelosuppression-associated toxicity. The expression of Gr-1+CD11b+ in blood, mesenteric lymph nodes and liver on day 13 was significantly reduced in the recipients of MyD88 KO TCD-BM compared with those of WT TCD-BM while the percentage of donor T cells infiltrating colon and liver was significantly higher. In parallel, the percentages of donor T cells to undergo apoptosis in response to alloantigens in vivo were significantly decreased in recipients of MyD88 KO TCD-BM. Injection of MDSCs from BM of non-tumor bearing donor markedly inhibited GVHD lethality in recipients of MyD88 KO TCD-BM. Moreover, in vivo administration of lipopolysaccharide (LPS), a TLR ligand, to donor mice expanded GR-1+CD11b+ in BM with enhanced expression of MyD88 mRNA. Recipients of TCD-BM from WT mice injected LPS showed attenuated GVHD severity as measured by weight loss and survival compared to those of TCD-BM from WT mice injected diluent. In summary, MyD88-dependent expansion of GR-1+CD11b+ population from donor TCD-BM appears to be critical for survival after allo-HSCT. Incomplete expansion of GR-1+CD11b+ population in target organs correlates with decreased apoptosis and increased infiltration of donor T cells into the target organs. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 455-455
Author(s):  
Jaebok Choi ◽  
Edward Dela Ziga ◽  
Julie Ritchey ◽  
Lynne Collins ◽  
Julie Prior ◽  
...  

Abstract Abstract 455 Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only curative treatment for patients with relapsed/refractory leukemia, and marrow failure states such as myelodysplasia and aplastic anemia. However, allo-HSCT is complicated by allogeneic donor T cell-mediated graft-versus-host disease (GvHD) which can be life-threatening especially in recipients of unrelated or HLA-mismatched hematopoietic stem cell products. These same alloreactive donor T cells also mediate a beneficial graft-versus-leukemia (GvL) effect. Thus, the clinical goal in allo-HSCT is to minimize GvHD while maintaining GvL. Recent studies have suggested that this might be achieved by infusing regulatory T cells (Tregs) which in some preclinical models suppress GvHD-causing alloreactive donor T cells but have only limited effects on GvL-promoting alloreactive donor T cells. Unfortunately, Tregs exist in low frequency in the peripheral blood, are costly to purify and expand, and after expansion are difficult to isolate due to the lack of cell surface markers, all of which prevent their routine use in the clinic. Thus, alternative therapeutic approaches that do not require Tregs are needed. We have found that interferon gamma receptor deficient (IFNγR−/−) allogeneic donor T cells induce significantly less GvHD in both a MHC fully-mismatched (B6 (H-2b) → Balb/c (H-2d)) and a minor-mismatched (B6 (H-2b) → B6×129(H-2b)) allo-HSCT models compared to WT T cells. In addition, IFNγR−/− donor T cells maintain a beneficial GvL effect, which has been examined in both systemic leukemia and solid tumor models using luciferase-expressing A20 cells derived from Balb/c. We find that IFNγR−/− T cells migrate primarily to the spleen while WT T cells to GI tract and peripheral lymph nodes (LNs) using bioluminescence imaging (BLI), suggesting that altered T cell trafficking of IFNγR−/− T cells to GvHD target organs might be the major reason for the reduced GvHD. We further demonstrate that the IFNγR-mediated signaling in alloreactive donor T cells is required for expression of CXCR3 which has been implicated in trafficking of T cells to areas of inflammation and target organs, commonly known to be the sites of GvHD. Indeed, CXCR3−/− T cells recapitulate the reduced GvHD potential of IFNγR−/− T cells. In addition, forced overexpression of CXCR3 in IFNγR−/− T cells via retroviral transduction partially rescues the GvHD defect observed in IFNγR−/− T cells. We next examine if inhibition of IFNγR signaling using a small molecule inhibitor can recapitulate the anti-GVHD effects seen in IFNγR−/− T cells. We find that INCB018424, an inhibitor of JAK1/JAK2 which are the mediators of IFNγR signaling, blocks CXCR3 expression in vitro. Most importantly, in vivo administration of INCB018424 after allo-HSCT alters T cell trafficking and significantly reduces GvHD. Thus, the IFNγR signaling pathway represents a promising therapeutic target for future efforts to mitigate GvHD while maintaining GvL after allo-HSCT. Moreover, this pathway can be exploited in other diseases besides GvHD such as those from organ transplantation, chronic inflammatory diseases and autoimmune diseases. Disclosures: DiPersio: genzyme: Honoraria.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2971-2971
Author(s):  
Jaebok Choi ◽  
Edward Dela Ziga ◽  
Julie Ritchey ◽  
Julie Prior ◽  
Lynne Collins ◽  
...  

Abstract Abstract 2971 Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only curative treatment for patients with relapsed/refractory leukemia, and marrow failure states such as myelodysplasia and aplastic anemia. However, allo-HSCT is complicated by allogeneic donor T cell-mediated graft-versus-host disease (GvHD) which can be life-threatening especially in recipients of unrelated or HLA-mismatched hematopoietic stem cell products. These same alloreactive donor T cells also mediate a beneficial graft-versus-leukemia (GvL) effect. Thus, the clinical goal in allo-HSCT is to minimize GvHD while maintaining GvL. Recent studies have suggested that this might be achieved by infusing regulatory T cells (Tregs) which in some preclinical models suppress GvHD-causing alloreactive donor T cells but have only limited effects on GvL-promoting alloreactive donor T cells. Unfortunately, Tregs exist in low frequency in the peripheral blood, are costly to purify and expand, and after expansion are difficult to isolate due to the lack of cell surface markers, all of which prevent their routine use in the clinic. Thus, alternative therapeutic approaches that do not require Tregs are needed. Using a MHC-mismatched GvHD model, B6 (H-2b) → Balb/c (H-2d), we demonstrated that infusion of IFN γR deficient allogeneic donor T cells induce significantly less GvHD, compared to WT T cells, determined by survival (74% vs. 0 % in overall survival; p =0.0004), weight and percentages of B220+ B cells (12.4% vs. 3.8%; p =0.0205), CD3+ T cells (14.3% vs. 4.3%; p =0.0025) in blood. Of note was that the IFN γR deficient donor T cells maintained a beneficial GvL effect, which was examined in both a systemic leukemia and a solid tumor model using luciferase-expressing A20 cells derived from Balb/c. We found that IFN γR deficient donor T cells responded normally to allogeneic antigens as measured by in vitro mixed lymphocyte reaction analyses, and express similar levels of granzyme B, compared to WT T cells. However, IFN γR deficient T cells trafficked predominantly to the spleen while WT T cells trafficked to gastrointestinal tract and peripheral lymph nodes, which are major GvHD target organs, based on in vivo bioluminescence imaging. All of these findings suggest that the reduced GvHD was not due to reduced function, altered subsets or relative deficiency of allogeneic donor T cells but from modification of in vivo trafficking of IFN γR deficient donor T cells compared to WT T cells. We further demonstrated that the IFN γR-mediated signaling in alloreactive donor T cells was required for expression of CXCR3 which has been implicated in trafficking of T cells to areas of inflammation and target organs, commonly known to be the sites of GvHD. CXCR3−/− T cells demonstrated a reduction in GvHD while maintenance of the same robust GvL effect using the same MHC mismatched transplant model. Thus, the IFN γR-CXCR3 axis represents a promising therapeutic target for future efforts to mitigate GvHD while maintaining GvL after allo-HSCT. Current studies are focused on 1) whether forced expression of CXCR3 rescues the GvHD-inducing potential of IFN γR deficient donor T cells and 2) if inhibition of IFN γR signaling (IFN γR, JAK1 and/or JAK2, CXCR3 and STAT1) using both neutralizing antibodies and small molecule inhibitors can recapitulate the anti-GvHD and pro-GvL effects seen in IFN γR−/− and CXCR3−/− T cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4464-4464
Author(s):  
Jaebok Choi ◽  
Matthew L Cooper ◽  
Julie Ritchey ◽  
Lynne Collins ◽  
Julie Prior ◽  
...  

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only curative treatment for patients with relapsed/refractory leukemia, and marrow failure states such as myelodysplasia and aplastic anemia. However, allo-HSCT is complicated by allogeneic donor T cell-mediated graft-versus-host disease (GvHD) which can be life-threatening especially in recipients of unrelated or HLA-mismatched hematopoietic stem cell products. These same alloreactive donor T cells also mediate a beneficial graft-versus-leukemia (GvL) effect. We have recently reported that interferon gamma receptor deficient (IFNγR-/-) allogeneic donor T cells induce significantly less GvHD in both a MHC fully-mismatched (B6 (H-2b) → Balb/c (H-2d)) (75% vs 0% overall survival) and a minor-mismatched (B6 (H-2b) → B6x129 (H-2b)) allo-HSCT models (100% vs 10% overall survival) compared to WT T cells (Choi et al Blood 2012). In addition, IFNγR-/- donor T cells maintain a beneficial GvL effect, which has been examined in both systemic leukemia and solid tumor models using luciferase-expressing A20 cells derived from Balb/c. We also found that IFNγR-/- T cells migrate primarily to the spleen while WT T cells to GI tract and peripheral lymph nodes (LNs) using bioluminescence imaging (BLI), suggesting that altered T cell trafficking of IFNγR-/- T cells to GvHD target organs might be the major reason for the reduced GvHD. We further demonstrated that the IFNγR-mediated signaling (via JAK1/2 - STAT pathway) in alloreactive donor T cells is required for expression of CXCR3 which has been implicated in trafficking of T cells to areas of inflammation and target organs, commonly known to be the sites of GvHD. Here, we examine if inhibition of IFNγR signaling using a small molecule inhibitor can recapitulate the reduced GVHD with potent anti-leukemia effects similarly to that seen with IFNγR-/- T cells. We find that INCB018424, an inhibitor of JAK1/JAK2 which mediate IFNγR signaling, blocks CXCR3 expression in vitro. Most importantly, in vivo administration of INCB018424 (100 ug, s.c., twice a day, day 1-31) after allo-HSCT alters T cell trafficking and significantly reduces GvHD (70% vs. 0% overall survival, n=10/group, p=0.0012). We also find that INCB018424 preserves the beneficial GvL effect, which has been examined in both systemic leukemia and solid tumor models using luciferase-expressing A20 cells derived from Balb/c (B6 to Balb/c model) and APL cells from B6x129 (B6 to B6x129 model). Of note is that INCB018424, when given after transplant, had no significant effect on neutrophil or platelet recovery compared to animals receiving placebo. Thus, the IFNγR signaling pathway represents a promising therapeutic target for future efforts to mitigate GvHD while maintaining GvL after allo-HSCT. Moreover, this pathway could be targeted and exploited in other diseases besides GvHD such as those from organ transplantation, chronic inflammatory diseases and autoimmune diseases. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3206-3206
Author(s):  
David Bastian ◽  
Hung Nguyen ◽  
Yongxia Wu ◽  
Steven Douglas Schutt ◽  
Mohammed Hanief Sofi ◽  
...  

Allogeneic hematopoietic stem cell transplantation (allo-HCT) is an effective means by which to treat a wide variety of diseases resulting from hematological dysfunction. However, the development of graft-versus-host disease (GVHD) remains a major cause of morbidity and mortality post transplantation. The IL-12 family of cytokines is comprised of IL-12, IL-23, IL-27, IL-35, and potentially IL-39. IL-12 family members are unique in that each cytokine and cognate receptor is comprised of heterodimers in which either the a or b subunit is shared among the others. IL-12 (p35+p40) and IL-23 (p19+p40) have well documented proinflammatory functions responsible for Th1 differentiation and Th17 stabilization, respectively, and play critical roles in GVHD development. IL-12R and IL-23R share a β-chain (IL-12Rβ1) yet use distinct α-chains to mediate their respective receptor signaling. While both IL-12R and IL-23R are widely implicated in inflammatory disorders, the role of IL-12Rβ1 in this context remains much less defined. We therefore studied the impact of eliminating the common IL-12Rβ1 chain or the unique IL-23Rα chain in T cells on GVHD using murine models of allogeneic bone marrow transplantation (BMT). In agreement with previous publications, we found a pathogenic role for IL-23Rα on donor T cells in aGVHD. Strikingly, a similar effect was not seen for IL-12Rβ1 (Figure 1A, B). These data suggest that that IL-23Rα contributes to GVHD pathogenesis via a pathway independent of IL-12Rβ1. To confirm that functional differences existed between T cells deficient for IL-23Rα or IL-12Rβ1 in GVHD, we assessed cytokine profiles of these T cells in target organs 14 days post-BMT. We found that, while production of IFNγ and IL-17 in the spleen was similarly decreased in both cohorts, GM-CSF production by CD4+ T cells was reduced exclusively in T cells deficient for IL-23Rα. Further, a significant reduction of IFNγ and GM-CSF in target organs, such as the liver and gut, was only observed in T cells deficient for IL-23Rα. The newest member of the IL-12 family, IL-39, was recently shown to contribute to SLE pathogenesis; this cytokine has been described to be composed of IL-23p19 and EBI3. Given the cognate receptor for IL-39 includes IL-23Rα and gp130, we hypothesized that IL-39 may play a role in aGVHD as this would explain why IL-12Rβ1 is dispensable. To validate that p19 and EBI3 can form a heterodimer, we transfected SV40 cells with vectors containing control, IL-23p19, EBI3 or both cDNAs. We detected IL-39 heterodimers only in the supernatant of cells transfected with both IL-23p19 and EBI3 via ELISA (Figure 1C). Furthermore, we observed significantly increased levels of IL-39 in allogeneic recipients at day 14 post BMT compared to naïve mice or recipients of BM alone in two models of aGVHD (Figure 1D). This may implicate IL-39 in the GVHD development. Taken together, our studies indicate that IL-23Rα plays an essential role, whereas IL-12Rβ1 is dispensable, for donor T cells to induce aGVHD. Our proposed model is that in the absence of IL-12Rβ1, IL-39 could transmit IL-23Rα signaling, hypothetically by forming a heterodimer with gp130. This new finding indicates that IL-23Rα and IL-39 are potential therapeutic targets for controlling aGVHD in the clinic. Figure 1. Effect of IL-12R 𝛃 1 in aGVHD and the potential role of IL-39 Lethally irradiated BALB/c mice were transplanted with 5x106 TCD-BM alone or plus 1x106 purified T cells from WT B6, IL-12R𝛃1KO, or IL-23RαKO mice. Survival (A) and body weight loss (B) are shown. Supernatant from SV40 cells transiently transfected with vectors containing control, IL-23p19, EBI3 or both cDNAs. p19 and EBI3 heterodimers were detected via ELISA (C). Serum was collected from naïve or lethally irradiated mice transplanted with 5x106 TCD-BM alone or plus 1x106 purified T cells. Formation of p19 and EBI3 heterodimers were tested in serum at 14 days post BMT via ELISA (D). Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 117 (16) ◽  
pp. 4181-4189 ◽  
Author(s):  
Olaf Penack ◽  
Gerard Socié ◽  
Marcel R. M. van den Brink

Abstract GVHD and tumor relapse are fundamental problems in allogeneic HSCT. Recent research has linked neovascularization to GVHD, tumor growth, and graft-versus-tumor (GVT) activity. Damage of the endothelium by the conditioning regimen provides the initiation stimulus for recruitment of donor-derived endothelial cells and their progenitors. During the early inflammatory phase of GVHD there is considerable neovascularization facilitating migration of inflammatory cells to target organs. In the course of GVHD, however, the vasculature itself becomes a target of alloreactive donor T cells. As a consequence, later stages of GVHD are characterized by fibrosis and rarefaction of blood vessels. Importantly, the inhibition of tumor-neovascularization by activated donor T cells that release antiangiogenic substances contributes to GVT and may be enhanced by pharmacologic inhibition of neovascularization. Furthermore, the therapeutic inhibition of neovascularization may improve immunotherapy for cancer by enhancing leukocyte infiltration in tumor tissue because of normalization of tumor vessels and stimulation of leukocyte–vessel wall interactions. These insights identify important mechanisms underlining the importance of neovascularization for allogeneic immune responses and move therapeutic approaches targeting neovascularization into the spotlight. This perspective covers current knowledge of the role of neovascularization during GVHD as well as GVT and its implications for HSCT.


Children ◽  
2021 ◽  
Vol 8 (6) ◽  
pp. 482
Author(s):  
Irene Paraboschi ◽  
Laura Privitera ◽  
Gabriela Kramer-Marek ◽  
John Anderson ◽  
Stefano Giuliani

Neuroblastoma (NB) is the most common extracranial solid tumour in childhood, accounting for approximately 15% of all cancer-related deaths in the paediatric population1. It is characterised by heterogeneous clinical behaviour in neonates and often adverse outcomes in toddlers. The overall survival of children with high-risk disease is around 40–50% despite the aggressive treatment protocols consisting of intensive chemotherapy, surgery, radiation therapy and hematopoietic stem cell transplantation2,3. There is an ongoing research effort to increase NB’s cellular and molecular biology knowledge to translate essential findings into novel treatment strategies. This review aims to address new therapeutic modalities emerging from preclinical studies offering a unique translational opportunity for NB treatment.


Blood ◽  
2011 ◽  
Vol 118 (22) ◽  
pp. 5965-5976 ◽  
Author(s):  
Ning Li ◽  
Catherine Matte-Martone ◽  
Hong Zheng ◽  
Weiguo Cui ◽  
Srividhya Venkatesan ◽  
...  

AbstractDonor T cells contribute to the success of allogeneic hematopoietic stem cell transplantation (alloSCT). Alloreactive donor T cells attack leukemia cells, mediating the GVL effect. Donor T cells, including the memory T cells (TM) that are generated after infection, also promote immune reconstitution. Nonetheless, leukemia relapse and infection are major sources of treatment failure. Efforts to augment GVL and immune reconstitution have been limited by GVHD, the attack by donor T cells on host tissues. One approach to augmenting GVL has been to infuse ex vivo–generated T cells with defined specificities; however, this requires expertise that is not widely available. In the present study, we tested an alternative approach, adoptive immunotherapy with CD8+ TM from donors vaccinated against a single minor histocompatibility antigen (miHA) expressed by leukemia cells. Vaccination against the miHA H60 greatly augmented TM-mediated GVL against mouse chronic-phase (CP-CML) and blast crisis chronic myeloid leukemia (BC-CML). TM-mediated GVL was antigen specific and was optimal when H60 expression was hematopoietically restricted. Even when H60 was ubiquitous, donor H60 vaccination had a minimal impact on GVHD. TM from lymphocytic choriomeningitis virus (LCMV)–immune and H60-vaccinated donors augmented GVL and protected recipients from LCMV. These data establish a strategy for augmenting GVL and immune reconstitution without elaborate T-cell manipulation.


2021 ◽  
Vol 12 ◽  
Author(s):  
Motoko Koyama ◽  
Geoffrey R. Hill

Allogeneic stem cell transplantation (alloSCT) is a curative therapy for hematopoietic malignancies. The therapeutic effect relies on donor T cells and NK cells to recognize and eliminate malignant cells, known as the graft-versus-leukemia (GVL) effect. However, off target immune pathology, known as graft-versus-host disease (GVHD) remains a major complication of alloSCT that limits the broad application of this therapy. The presentation of recipient-origin alloantigen to donor T cells is the primary process initiating GVHD and GVL. Therefore, the understanding of spatial and temporal characteristics of alloantigen presentation is pivotal to attempts to separate beneficial GVL effects from detrimental GVHD. In this review, we discuss mouse models and the tools therein, that permit the quantification of alloantigen presentation after alloSCT.


Sign in / Sign up

Export Citation Format

Share Document