scholarly journals Functional analysis of clinical response to low-dose IL-2 in patients with refractory chronic graft-versus-host disease

2019 ◽  
Vol 3 (7) ◽  
pp. 984-994 ◽  
Author(s):  
Jennifer S. Whangbo ◽  
Haesook T. Kim ◽  
Sarah Nikiforow ◽  
John Koreth ◽  
Ana C. Alho ◽  
...  

Abstract Patients with chronic graft-versus-host disease (cGVHD) have a paucity of regulatory CD4 T cells (CD4Tregs) that mediate peripheral tolerance. In clinical trials, daily low-dose interleukin-2 (IL-2) has been administered safely for prolonged periods in patients with steroid-refractory cGVHD. Peripheral CD4Tregs expand dramatically in all patients during IL-2 therapy but clinical improvement was observed in ∼50% of patients. Here, we examined the impact of low-dose IL-2 therapy on functional T-cell markers and the T-cell repertoire within CD4Tregs, conventional CD4 T cells (CD4Tcons), and CD8+ T cells. IL-2 had profound effects on CD4Tregs homeostasis in both response groups including selective expansion of the naive subset, improved thymic output, and increased expression of Ki67, FOXP3, and B-cell lymphoma 2 within CD4Tregs. Similar changes were not seen in CD4Tcons or CD8 T cells. Functionally, low-dose IL-2 enhanced, in vitro, CD4Treg-suppressive activity in both response groups, and all patient CD4Tcons were similarly suppressed by healthy donor CD4Tregs. High-throughput sequencing of the T-cell receptor β (TCRβ) locus demonstrated that low-dose IL-2 therapy increased TCR repertoire diversity and decreased evenness within CD4Tregs without affecting CD4Tcons or CD8 T cells. Using clone-tracking analysis, we observed rapid turnover of highly prevalent clones in CD4Tregs as well as the conversion of CD4Tcons to CD4Tregs. After 12 weeks of daily IL-2, clinical responders had a greater influx of novel clones within the CD4Treg compartment compared with nonresponders. Further studies to define the function and specificity of these novel CD4Treg clones may help establish the mechanisms whereby low-dose IL-2 therapy promotes immune tolerance.

Blood ◽  
2020 ◽  
Author(s):  
Cheng Yin Yuan ◽  
Vivian Zhou ◽  
Garrett Sauber ◽  
Todd M Stollenwerk ◽  
Richard Komorowski ◽  
...  

Graft versus host disease (GVHD) pathophysiology is a complex interplay between cells that comprise the adaptive and innate arms of the immune system. Effective prophylactic strategies are therefore contingent upon approaches that address contributions from both immune cell compartments. In the current study, we examined the role of the type 2 cannabinoid receptor (CB2R) which is expressed on nearly all immune cells and demonstrated that absence of the CB2R on donor CD4+ or CD8+ T cells, or administration of a selective CB2R pharmacological antagonist, exacerbated acute GVHD lethality. This was accompanied primarily by the expansion of proinflammatory CD8+ T cells indicating that constitutive CB2R expression on T cells preferentially regulated CD8+ T cell alloreactivity. Using a novel CB2R-EGFP reporter mouse, we observed significant loss of CB2R expression on T cells, but not macrophages, during acute GVHD, indicative of differential alterations in receptor expression under inflammatory conditions. Therapeutic targeting of the CB2R with the agonists, tetrahydrocannabinol (THC) and JWH-133, revealed that only THC mitigated lethal T cell-mediated acute GVHD. Conversely, only JWH-133 was effective in a sclerodermatous chronic GVHD model where macrophages contribute to disease biology. In vitro, both THC and JWH-133 induced arrestin recruitment and ERK phosphorylation via CB2R, but THC had no effect on CB2R-mediated inhibition of adenylyl cyclase. These studies demonstrate that the CB2R plays a critical role in the regulation of GVHD and suggest that effective therapeutic targeting is dependent upon agonist signaling characteristics and receptor selectivity in conjunction with the composition of pathogenic immune effector cells.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3244-3244
Author(s):  
Jenny Zilberberg ◽  
Gichuru N. Loise ◽  
Thea M. Friedman

Abstract Lethal graft-versus-host disease (GVHD) can be induced between MHC-matched murine strains expressing multiple minor histocompatibility antigen (miHA) differences. In the C57BL6 (B6)->BALB.B strain combination, both CD4+ and CD8+ donor T cells can mediate severe lethal GVHD, whereas in the B6->CXB-2 model, in which the CXB-2 strain expresses a subset of the BALB.B miHA, only the CD8+ T cells directly potentiate lethality. We have previously used TCR Vβ CDR3-size spectratype analysis to examine the alloreactive B6 CD4+ and CD8+ T cells, isolated from the lymphohematopoietic compartment after transplantation into both BALB.B and CXB-2 recipients. However, since tissue-specific expression of miHA can potentially elicit differential T cell responses, we have extended our T cell repertoire analysis to examine the responses involved in target tissue damage. Infiltrating host-presensitized B6 CD4+ and CD8+ T cells were isolated post-transplant from the intestines, livers and spleens of lethally irradiated (9 Gy; split-dose) BALB.B and CXB-2 recipients. The results indicated some overlapping Vβ CDR3-size skewing in both the CD4+ and CD8+ T cell repertoires between the BALB.B and CXB-2 recipients within the tissues of each recipient strain. Most notably, spectratype analysis demonstrated tissue specific responses unique to each of the BALB.B and CXB-2 infiltrates. In situ observations of the tissue infiltrating alloreactive T cells were performed by fluorescent microscopy of transplanted B6 T cells constitutively expressing eGFP into BALB.B and CXB-2 recipients, in conjunction with immunohistochemical staining of skewed Vβ families. TUNEL staining was also performed to confirm apoptosis of tissue epithelium. These analyses confirmed the increased infiltration of skewed CD4+ and CD8+ Vβ families within the target tissues.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 231-231
Author(s):  
Abdulraouf Ramadan ◽  
Jilu Zhang ◽  
Mohammad Abu Zaid ◽  
Heather O'Leary ◽  
Reuben Kapur ◽  
...  

Abstract Treatment of acute myeloid leukemia (AML) has changed little over the last several decades and prognosis remains very poor. Allogeneic hematopoietic cell transplantation (allo-HCT) is one potentially curative option for relapsed or high-risk AML. The immunotherapeutic activity of allo-HCT is known as the graft-vs-leukemia (GVL) activity. However, GVL activity is often accompanied by T-cell reactivity to allo-antigens in normal host tissues, which leads to graft-versus-host disease (GVHD), another major cause of death after HCT with relapse. Therefore, there is a great unmet need to improve the current process of allo-HCT through increasing the GVL activity and decreasing GVHD. We have shown that an elevated plasma level of soluble (s)ST2 in HCT patients is a risk factor for severe GVHD (Vander Lugt et al, N Engl J Med, 2013) and that ST2 blockade reduces sST2-producing T cells while maintaining protective membrane (m)ST2-expressing T cells during GVHD (Zhang et al, Sci Transl Med, 2015). In addition, the interleukin (IL)-9-producing CD4 T helper (Th)9 and CD8 cytotoxic T (Tc)9 cell subsets (together T9 cells) have higher antitumor activity than Th1 and Tc1 cells in melanoma models (Lu et al, J Clin Invest, 2012 and Lu et al, ProcNatl Acad Sci, 2014). We hypothesized that activation of the ST2/IL-33 pathway in T9 cells will both alleviate GVHD and increase GVL. In our laboratory, we have shown that T9 cells express a high level of mST2 and that differentiation of total T cells into T9 cells in the presence of IL-33 (T9IL-33 cells) increases expression of mST2 (Figure 1A) and PU.1 (Figure 1B), a transcription factor that promotes IL-9 production on both CD4 and CD8 T cells. Adoptive transfer of T9IL-33 cells with bone marrow cells in a murine model of HCT resulted in less severe GVHD compared to transfer of T9IL33 cells generated from ST2-/- or IL-9-/- T cells (Figure 1C). Ex-vivo analysis of target organs such as gut showed a decrease in T9IL-33 interferon (IFN)g-producing T cells that was abolished in mice receiving T9IL-33 cells derived from ST2-/- or IL-9-/- T cells (not shown). Furthermore, T9IL-33 cells revealed higher anti-leukemic activity in vitro when cultured with a B cell lymphoma line (A20) or retrovirally transduced MLL-AF9 leukemic cells in cytolytic assays (not shown). In vivo GVL experiments with MLL-AF9 induced leukemia, and adoptive transfer of T9IL-33 cells resulted in increased survival compared to transfer of T9IL-33 cells generated from ST2-/- or IL-9-/- T cells (Figure 1D). Human T9 cells are poorly explored. We demonstrated that differentiation of human T9 cells in the presence of IL-33 enhanced IL-9 production by CD4 and CD8 T cells (Figure 2A). T9IL-33 cells also upregulated the expression of the cytolytic molecules granzymes A and B compared to T9 cells (Th9IL-33: 33.6%±4%, vs. Th9: 15.69%±2.53% p=0.021), (Tc9IL-33: 57.6%±4.7%, vs. Tc9: 34.61%±3.4% p=0.018), as well as demonstrated higher in vitro anti- leukemic cytolytic activity when incubated with MOLM14, an aggressive AML tumor cell line expressing FLT3/ITD mutations (Figure 2B). Transcriptome analysis of T9IL-33 cells from wild-type and ST2-/- T cells showed upregulation of molecules implicated in anti-leukemic activity (granzymes A and B, CD8α, IL-15, IL-15rα, IFNα, and IL-1α) on both CD4 and CD8 T cells (Figure 2C), and such upregulation was confirmed at the protein level (Figure 2D). Furthermore,investigations into the possible mechanism of activation using transwell assays revealed that both soluble factors and cell contact between Th9IL-33 and Tc9IL-33 T cells were required for maximum killing (not shown). We next investigated the possible mechanism of action and hypothesized that CD8α might be the contact-dependent component. CD8α blockade with neutralizing antibody during human T9IL-33 differentiation reduced the cytotoxicity of both murine T9IL-33 and human T9IL-33 cells (Figure 2E). Altogether, our observations suggest that adoptive transfer of T9IL-33 cells represents a promising cellular therapy following HCT. Figure 1. Figure 1. Figure 2. Figure 2. Disclosures Paczesny: Viracor laboratories: Patents & Royalties: "Methods of detection of graft-versus-host disease" (US- 13/573,766).


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 150-150
Author(s):  
Ludovic Belle ◽  
Kimberle A. Agle ◽  
Vivian Zhou ◽  
Vanessa Yuan ◽  
Jie Sun ◽  
...  

Abstract The interleukin-6 (IL-6) cytokine superfamily (i.e. IL-6, IL-12, and IL-23) plays a major role in the modulation of inflammatory and regulatory pathways during graft versus host disease (GVHD). IL-27, a recently discovered member of this family, is a heterodimeric cytokine that is composed of the p28 and EBI3 subunits and signals through a heterodimeric receptor composed of WSX-1 and gp130. Notably, IL-6 also uses gp130 as a signaling component which biologically links IL-27 and IL-6. IL-27 has been shown to have opposing proinflammatory and immunoregulatory effects, but its role in GVHD is not well understood. To define the functional significance of IL-27, lethally irradiated Balb/c (H-2d) mice were transplanted with C57BL/6J (H-2b) BM and spleen cells, and then treated with an anti-IL-27p28-specific antibody on days 0 and +6. p28 antibody-treated animals had significantly improved weight recovery and overall survival (47% versus 0% survival at day 60, p=0.002), as well as reduced numbers of proinflammatory CD4+ and CD8+ IFN-γ+ T cells in GVHD target organs, when compared to isotype control antibody-treated mice. A similar outcome was observed in an MHC-matched, minor antigen disparate model (B6→Balb.B), indicating that this was not a strain-specific phenomenon. Given the similarities between IL-6 and IL-27, we examined whether blockade of IL-27 promoted regulatory T cell (Treg) reconstitution as has been observed with inhibition of IL-6 signaling. Recipients transplanted with BM grafts from B6 Foxp3EGFP reporter animals and treated with p28 antibody had a significant increase in the number of CD4+ nTregs, CD4+ iTregs and CD8+ iTregs in GVHD target organs, indicating that blockade of IL-27 augmented global Treg reconstitution. In fact, inhibition of IL-27 was more effective at augmenting Treg reconstitution than comparable antibody blockade of IL-6. To further elucidate the role of IL-27, we employed transgenic IL-27−/− and IL-27R−/− animals to dissect the relevant contributions of donor and recipient populations. Paradoxically, we observed that transplantation with IL-27−/− donor grafts exacerbated GVHD mortality and augmented accumulation of proinflammatory T cells, whereas transplantation of recipient IL-27−/− mice with wild type grafts had no effect on transplant outcomes. This discordance between antibody-based and genetic studies was unexpected and led us to consider whether there were steady state alterations in T cells from IL-27−/− animals that biased these cells towards a proinflammatory phenotype. To that end, we observed that naive CD8+ T cells from IL-27−/− mice had greater IFN-γ production than wild type cells after in vitro polyclonal stimulation and CD4+ nTregs from these animals had diminished expression of CXCR3 which is critical for Treg trafficking into inflamed tissue sites. Thus, the lack of endogenous IL-27 resulted in intrinsic immune dysregulation which led to an exacerbation of GVHD after transfer of these T cells into recipients. To resolve this paradox, we employed IL-27R−/− (WSX-1−/−) mice and demonstrated that mice transplanted with IL-27R−/− grafts had enhanced weight recovery and survival providing confirmation that blockade of IL-27 signaling reduced GVHD. In addition, using IL-27R−/− Foxp3EGFP reporter mice, we observed increased frequencies and numbers of CD4+ and CD8+ Foxp3+ T cells in mice reconstituted with IL-27R−/− grafts, confirming results observed with p28 antibody blockade. Since IL-10 is a mechanism by which CD4+ Tregs suppress GVHD and IL-27 has been shown to enhance T cell-derived IL-10 secretion in nontransplant models, we examined whether IL-27 blockade adversely affected IL-10 production by Tregs. Recipients transplanted with marrow grafts from IL-10.BitFoxp3EGFP dual reporter animals and treated with p28 antibody had a significant reduction in the frequency of IL-10-producing conventional CD4+ and CD8+ T cells in GVHD target organs. Notably, however, there was no difference in the frequency of CD4+ Foxp3+ IL-10+ T cells, indicating that blockade of IL-27 signaling preferentially affected conventional T cells and had no adverse effect on CD4+ Foxp3+ T cell-derived IL-10 production. In summary, these studies demonstrate that blockade of IL-27 signaling potently augments Treg reconstitution leading to a reduction in the severity of GVHD and may therefore represent a novel strategy to reduce mortality from this disease in man. Disclosures No relevant conflicts of interest to declare.


2011 ◽  
Vol 186 (11) ◽  
pp. 6238-6254 ◽  
Author(s):  
Anthony D. Foster ◽  
Kateryna Soloviova ◽  
Irina Puliaeva ◽  
Maksym Puliaiev ◽  
Roman Puliaev ◽  
...  

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1300-1300
Author(s):  
Barry J. Kappel ◽  
Javier Pinilla-Ibarz ◽  
Adam A. Kochman ◽  
Jeffrey M. Eng ◽  
Vanessa M. Hubbard ◽  
...  

Abstract MHC molecules carrying selected peptides will bind specifically to their cognate T cell receptor on individual clones of reactive T cells. Fluorescently labeled, tetrameric MHC-peptide complexes have been widely used to detect and quantitate antigen specific T cell populations via flow cytometry. We hypothesized that such MHC-peptide tetramers could also be used to selectively deplete unique reactive T cell populations, while leaving the remaining T cell repertoire and immune response intact. We show in an MHC-matched, minor antigen disparate, murine BMT model (BALB.B → C57BL/6), MHC-peptide-tetramers can be used to deplete the T cells responsible for Graft-Versus-Host-Disease (GVHD), while leaving the remaining immune response intact, as demonstrated by the retention of Graft-Versus-Tumor (GVT) activity. Using PE-labeled tetramers, anti-PE microbeads and an autoMACs separation system, we successfully depleted donor splenocytes of alloantigen specific T cells prior to transplantation. We demonstrated the specificity of the depletion by showing loss of the tetramer reactivity after depletion, whereas no changes were observed in the Vβ repertoire and the percentage of T cells, B cells, NK cells, monocyte/macrophages and granulocytes between pre- and post-depletion samples. When analyzed 6 days after transplantation, mice receiving specifically-depleted splenocytes had <0.5% of their CD8+ T cells reactive against the alloantigen (tetramer +) as compared to >8.5% of the CD8+ T cells in mice that received control-depleted splenocytes. A nearly 50% decrease in in vivo proliferation of donor splenocytes, assessed by CFSE dilution, was seen 3 days after transplant in recipients of specifically-depleted splenocytes, as compared to mice receiving control-depleted splenocytes. However, pre- and post-depletion splenocytes (specific and control) were equally capable of mounting an immune response against third party cells as demonstrated by mixed lymphocyte reaction. In a series of bone marrow transplants designed to assess GVHD and GVT, mice receiving specifically-depleted splenocytes had a nearly 4-fold increased median survival due to significant decreases in GVHD morbidity and mortality compared to recipients of control-depleted splenocytes. All mice receiving splenocytes (tetramer-depleted or not) showed equal GVT activity. Finally, we were able to demonstrate the simultaneous abrogation of GVHD and the retention of GVT in a single bone marrow transplant. In recipients of specifically-depleted splenocytes, there was a 33% long-term survival and significant increases in median survival, as compared to recipients of non-depleted splenocytes, control-depleted splenocytes or bone marrow only; all of these latter groups succumbed to GVHD or tumor. This method also provides the proof-of-concept for similar strategies to selectively remove other unwanted T cell clones, which could result in novel therapies for certain autoimmune disorders, T cell malignancies and solid organ graft rejection.


Blood ◽  
2005 ◽  
Vol 106 (9) ◽  
pp. 3322-3330 ◽  
Author(s):  
Theis H. Terwey ◽  
Theo D. Kim ◽  
Adam A. Kochman ◽  
Vanessa M. Hubbard ◽  
Sydney Lu ◽  
...  

AbstractGraft-versus-host disease (GVHD) is a major complication of allogeneic hematopoietic stem cell transplantation (HSCT). Migration of donor-derived T cells into GVHD target organs plays a critical role in the development of GVHD and chemokines and their receptors are important molecules involved in this process. Here, we demonstrate in murine bone marrow transplantation models that the expression of the inflammatory CC chemokine receptor 2 (CCR2) on donor-derived CD8+ T cells is relevant for the control of CD8+ T-cell migration and development of GVHD. Recipients of CCR2-deficient (CCR2-/-) CD8+ T cells developed less damage of gut and liver than recipients of wild-type CD8+ T cells, which correlated with a reduction in overall GVHD morbidity and mortality. Assessment of donor CD8+ T-cell target organ infiltration revealed that CCR2-/- CD8+ T cells have an intrinsic migratory defect to the gut and liver. Other causes for the reduction in GVHD could be excluded, as alloreactive proliferation, activation, IFN-γ production and cytotoxicity of CCR2-/- CD8+ T cells were intact. Interestingly, the graft-versus-tumor effect mediated by CCR2-/- CD8+ T cells was preserved, which suggests that interference with T-cell migration by blockade of CCR2 signaling can separate GVHD from GVT activity.


Blood ◽  
2011 ◽  
Vol 117 (5) ◽  
pp. 1723-1733 ◽  
Author(s):  
Fang Zhao ◽  
Yi Zhang ◽  
Hao Wang ◽  
Min Jin ◽  
Shan He ◽  
...  

Abstract Graft-versus-host disease (GVHD), a life-threatening complication after allogeneic hematopoietic stem cell transplantation, is caused by alloreactive donor T cells that trigger host tissue damage. The inflammatory environment inside recipients is critical for GVHD pathogenesis, but the underpinning mechanisms remain elusive. Using mouse model of human GVHD, we demonstrate osteopontin (OPN), a potent proinflammatory cytokine, plays an important role in regulating activation, migration, and survival of alloreactive T cells during GVHD. OPN was significantly elevated after irradiation and persisted throughout the course of GVHD. Blockade of OPN attenuated GVHD with reduced accumulation of donor T cells in recipient organs. Amelioration was the result of migration and survival suppression caused by anti-OPN treatment on donor-derived T cells for 2 reasons. First, OPN promoted the migration and infiltration of naive and alloreactive CD8+ T cells into host organs. Second, it also facilitated activation and viability of donor-derived CD8+ T cells via synergizing with T-cell receptor/CD3 signaling. Finally, anti-OPN treatment retained graft-versus-leukemia effect of alloreactive CD8+ T cells. This study demonstrates, to our knowledge for the first time, the critical effect of OPN in the initiation and persistence of CD8+ T cell-mediated GVHD and validates OPN as a potential target in GVHD prevention.


1999 ◽  
Vol 189 (7) ◽  
pp. 1073-1081 ◽  
Author(s):  
Defu Zeng ◽  
David Lewis ◽  
Sussan Dejbakhsh-Jones ◽  
Fengshuo Lan ◽  
Marcos García-Ojeda ◽  
...  

Sorted CD4+ and CD8+ T cells from the peripheral blood or bone marrow of donor C57BL/6 (H-2b) mice were tested for their capacity to induce graft-versus-host disease (GVHD) by injecting the cells, along with stringently T cell–depleted donor marrow cells, into lethally irradiated BALB/c (H-2d) host mice. The peripheral blood T cells were at least 30 times more potent than the marrow T cells in inducing lethal GVHD. As NK1.1+ T cells represented <1% of all T cells in the blood and ∼30% of T cells in the marrow, the capacity of sorted marrow NK1.1− CD4+ and CD8+ T cells to induce GVHD was tested. The latter cells had markedly increased potency, and adding back marrow NK1.1+ T cells suppressed GVHD. The marrow NK1.1+ T cells secreted high levels of both interferon γ (IFN-γ) and interleukin 4 (IL-4), and the NK1.1− T cells secreted high levels of IFN-γ with little IL-4. Marrow NK1.1+ T cells obtained from IL-4−/− rather than wild-type C57BL/6 donors not only failed to prevent GVHD but actually increased its severity. Together, these results demonstrate that GVHD is reciprocally regulated by the NK1.1− and NK1.1+ T cell subsets via their differential production of cytokines.


Sign in / Sign up

Export Citation Format

Share Document