scholarly journals Adverse Events Following Infusion of T Cells for Adoptive Immunotherapy: A 10 Year Experience.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3212-3212
Author(s):  
Conrad Russell Y. Cruz ◽  
Patrick Hanley ◽  
Hao Liu ◽  
Vicky Torrano ◽  
Yu-Feng Lin ◽  
...  

Abstract Abstract 3212 Poster Board III-149 Following administration of ex-vivo expanded T cells, the FDA currently recommends at least 4 hours of recipient monitoring to detect early infusion reactions. Recent catastrophic reactions to “first in man” biological agents have emphasized the importance of this rule for initial studies of new products, but its value for longer established agents is less evident. We have therefore reviewed the incidence and nature of infusion-related adverse events (AEs) associated with administration of ex-vivo expanded T cell products (antigen specific CTLs, allodepleted T cells, and genetically modified T cells) on Investigational New Drug (IND) studies in our center over the last decade. From 1998 to 2008, we infused 381 T cell products to 180 recipients who were enrolled on 18 such studies, receiving T cells targeting malignancies or post-transplant viral infections. The age of these recipients ranged from 9 months to 80 years. Cell doses were protocol specific and ranged from 104/kg up to 3 ×108/m2. Patients were premedicated with diphenhydramine (0.5-1mg/kg) and acetaminophen (10mg/kg up to a maximum of 625mg) prior to infusion. All cellular products were cryopreserved and administered intravenously over 1-15 minutes immediately after thawing. There were no Grade 3-4 infusion reactions during initial monitoring or 24 hour follow-up. Twenty four grade 1-2, non-severe adverse events (AEs) occurred in 21 infusions either during or immediately following infusion (up to 6 hours). The most common AEs were nausea and vomiting (10/24; 41.6%), most likely due to DMSO used in cryopreservation of T cell products, and hypotension (20.8%), attributable to diphenhydramine pre-medication. An additional 22 non-severe events within 24 hours of infusion were reported, the most common of which were fever (with negative blood cultures)/chills/constitutional symptoms (6/22; 27.3%) and nausea/vomiting (4/22; 18.2%) Overall, a total of 46 non-severe adverse events were noted within 24 hours of T cell product infusion (12.56%). A Fisher's exact analysis of all T cell product infusions grouped by patient age, patient ethnicity, or cell source revealed no association with increased risk. T cells from both allogeneic and autologous sources produced similar adverse events in terms of type, frequency, and severity, and allogeneic cells that were mismatched at > 2 HLA antigens had the same AEs as donor T cells matched at 5/6 or 6/6 HLA loci. We further analyzed the data using Poisson regression and the general estimating equation (GEE) model for correlated counts, to seek associations that may have been missed because AEs within a subject may not be statistically independent. By this analysis, we found decreased risks of adverse events in older patients (IRR 0.98; 95% CI 0.96-1.00; p=0.05), and increased risks of immediate (defined as occurring during the monitoring period) infusion-related events in patients reporting allergies (IRR 2.72; 95% CI 1.00-7.40; p=0.05). We thus conclude that infusion of the ex-vivo expanded T cell products used in these studies is a safe procedure associated with no severe reactions, that it is safe in the outpatient setting and that monitoring can be limited to an hour after infusion. As many of the AEs observed were due to diphenhydramine premedication, a lower dose (0.25mg/kg) of this agent may be preferred. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1715-1715
Author(s):  
Martijn H.A. van Attekum ◽  
Sanne Terpstra ◽  
Emilie Reinen ◽  
Marieke Von Lindern ◽  
Erik Slinger ◽  
...  

Abstract Survival of CLL cells critically depends on heterotypic communication with benign bystanders cells in micro-environmental niches such as lymph node (LN) tissue. Here, mesenchymal stromal cells and macrophages, in concert with CD40L expressing T cells, are thought to participate in the dialog with the neoplastic B cells, but the mechanisms of this intricate interplay remain largely unknown. Moreover, whether CLL cells actively participate in shaping their prosurvival niche is poorly understood. We aimed to study 1) whether CD40 stimulation initiates active recruitment of monocytes by CLL cells, 2) whether CLL cells are able to differentiate these monocytes towards a supporting phenotype and 3) by which mechanism macrophages induce CLL survival. We first studied the chemokinome of CLL cells after T cell stimulation using both microarray and Luminex techniques. Co-culture of autologous activated T cells with CLL cells resulted in induction of mRNA expression of CCL2,3,4,5,22 and IL10, which are known chemo-attractants for monocytes. These effects could be mimicked by CD40 activation of CLL cells. Protein screens of supernatants of CD40 activated CLL cells by Luminex assays confirmed increased protein expression of these chemo-attractants. Indeed, transwell assays showed enhanced migration of primary monocytes towards supernatants of CD40L stimulated CLL cells. Inhibitor experiments furthermore showed that the migratory effects of these chemokines was largely governed via the CCR2 and CCR3 receptors. We next examined and compared polarization patterns of monocytes after differentiation with serum derived from CLL patients (N=25) or pooled healthy donor serum and found that CLL serum was able to differentiate macrophages towards a tumor supporting M2 phenotype. This finding was confirmed ex vivo by IHC, as M2 marker CD206 co-localizes with CD68 cells in CLL LNs, while the majority of macrophages in non-CLL derived LNs are CD80+ (M1 type). Lastly, we examined how these macrophages exert their pro-survival effect on CLL. From a variety of Bcl-2 family proteins investigated, only Mcl-1 protein expression levels increased after interaction with macrophages. The relevance of Mcl-1 upregulation was verified by MCL-1 siRNA interference studies. The mechanism of induction of Mcl-1 was independent on NF-κB signaling, Mcl-1 mRNA transcription levels or protein stability, but rather unexpectedly appeared as a result of recruitment of polysomes to Mcl-1 mRNA, resulting in an increase in translation. This increase was accompanied by an increased phosphorylation of the rate-limiting translation initiation factor 4E-BP1 and ribosomal protein S6. The increase in Mcl-1 translation could be attributed to macrophage-induced Akt signaling. In conclusions, these studies shed light on reciprocal cellular interactions in the CLL LN that shape pro-tumor differentiation of supporting cells, that in turn cause survival by changing the apoptotic balance. These interactions can be targeted at different levels, creating new treatment venues for this still incurable disease. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3154-3154 ◽  
Author(s):  
Weijun Fu ◽  
Juan Du ◽  
Hua Jiang ◽  
Zhi CHENG ◽  
Runhong Wei ◽  
...  

Background: Encouraging results are seen from several early phase clinical trials on the cellular immunotherapy based on chimeric antigen receptor (CAR)-engineered T (CAR-T) targeting B cell maturation antigen (BCMA) for the treatment of relapsed/refractory (RR) multiple myeloma (MM). We developed an anti-BCMA CAR-T cell product manufactured via gamma-retrovirus-mediated transduction of activated T cells to express a second-generation CAR with the 4-1BB costimulatory domain along with a truncated epidermal growth factor receptor (tEGFR) as a safety switch. The preclinical study confirmed its high reactivity against MM cells. Methods: A phase 1 clinical trial (NCT03093168) has been launched to evaluate the safety and feasibility of this BCMA CAR-T cell product for treating RRMM. The enrolled RRMM patients had received at least 2 prior treatment regimens, including a proteasome inhibitor and an immunomodulatory agent, or are double-refractory, and have over 5% BCMA expression on plasma cells (Nine patient with extramedullary plasmacytoma does not express BCMA). Patients were subjected to a lymphodepleting regimen with Cy (300 mg/m2, d-5 to d-3) and Flu daily for 3 days (25 mg/m2, d-5 to d-3) prior to the CAR-T infusion (d0) at a dose of 9×106CAR+ cells/kg. The efficacy was assessed by the International Uniform Response Criteria for Multiple Myeloma, and the toxicity is graded by CTCAE 4.03. Results: As of March 1th, 2019, 46 patients had been infused with this intended dose of the autologous BCMA CAR-T cells, and 44 patients had reached at least 1 month of follow-up. As of this data cut-off, the overall response rate (ORR) for the 44 evaluable patients was 79.6%, including 2sCRs, 16CRs, 8VGPRs and 8PRs, and 16 patients reached MRD-negative response. The CAR-T cell expansion and persistence were consistently observed throughout these patients. The medianPFS is 15mon, and the median OS result has not been reached (49.16% progression-free survival, and 53.95% overall survival at 24 months). Among the 44 infused patients, 22.7% had grade 1-2 Cytokine release syndrome (CRS ) and 6.8% (3 patients) had grade 3 CRS. No grade 4 CRS reactions developed and all toxicities were fully reversible. Conclusions: Our result demonstrates the high potential of this single CAR-T infusion therapy for RRMM, including 2sCRs, 16CRs and ongoing clinical responses for more than 26 months, with manageable CRS to date. These initial data provide strong evidence to support the further development of this anti-myeloma cellular immunotherapy. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5771-5771
Author(s):  
Giulia Barbarito ◽  
Irma Airoldi ◽  
Alessia Zorzoli ◽  
Alice Bertaina ◽  
Andrea Petretto ◽  
...  

Abstract A new method of graft manipulation based on physical removal of αβ+ T cells and CD19+ B cells, leaving mature NK cells and γδ T cells in the graft, has been recently developed for HLA-haploidentical HSCT. We demonstrated that γδ T cells collected from transplanted patients are endowed with capacity of killing leukemia cells after ex vivo treatment with zoledronic acid (ZOL). Thus, we hypothesized that infusion of ZOL in patients receiving this type of graft, may boost γδ T cell cytotoxic activity against leukemia cells. Thirty-three patients were treated with ZOL every 28 days at least twice. γδ T cells before and after ZOL treatments were studied till at least 7 months after HSCT by high-resolution mass spectrometry, flow-cytometry, and degranulation assay. Proteomic analysis of γd T cells purified from patients showed that, starting from the first infusion, ZOL caused up-regulation of proteins involved in activation processes and immune response, paralleled by down-regulation of proteins involved in proliferation. These findings are consistent with an induction of Vδ2 cell differentiation, paralleled by increased cytotoxicity of both Vδ1 and Vδ2 cells against primary leukemia blasts. Furthermore, a proteomic signature was identified for each individual ZOL treatment. Patients given 3 or more ZOL infusions had a better probability of survival in comparison to those given 1 or 2 treatments. In conclusion,ZOL influences Vδ2 cell activity, determines a specific proteomic signature and enhances anti-leukemia activity, this potentially resulting into an increased anti-tumor effect. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4465-4465
Author(s):  
Margot J Pont ◽  
Willemijn Hobo ◽  
M. Willy Honders ◽  
Simone A.P. van Luxemburg-Heijs ◽  
Michel G.D. Kester ◽  
...  

Patients with hematological malignancies can be successfully treated with allogeneic hematopoietic stem cell transplantation (alloSCT). Beneficial Graft-versus-Leukemia (GvL) reactivity, however, is often accompanied with undesired Graft-versus-Host Disease (GvHD). In HLA-matched alloSCT, donor T-cells can mediate GvL reactivity by recognition of minor histocompatibility antigens (MiHA) on malignant cells, but also GvHD when MiHA are recognized on non-hematopoietic tissues. To decrease the incidence and severity of GvHD, T-cells can be (partially) depleted from the graft and re-administered later as donor lymphocyte infusion (DLI). MiHA are polymorphic peptides that are presented in HLA-molecules and can be recognized as non-self by donor-derived T-cells. Only a minority of MiHA have therapeutic relevance based on hematopoietic-restricted expression and only 25% and 40% of recipients transplanted with sibling and unrelated donors, respectively, are eligible for T-cell therapies in which one of the known hematopoietic-restricted MiHA is targeted. Therefore, to increase the efficacy and limit the toxicity of T-cell therapy, more therapeutic MiHA are needed. Recently, we identified a MiHA encoded by ARHGDIB, a gene that has been described to be highly expressed on hematopoietic cells, and we here studied the therapeutic relevance of LB-ARHGDIB-1R in detail. First, we confirmed hematopoietic-restricted expression of ARHGDIB by microarray gene expression analysis and demonstrated >10-fold overexpression in the majority of malignant and healthy hematopoietic versus non-hematopoietic cells. In line with its hematopoietic-restricted gene expression profile, LB-ARHGDIB-1R in the context of HLA-B* 07:02 was specifically recognized on different hematological malignancies, but not on non-hematopoietic fibroblasts and keratinocytes cultured in the absence or presence of IFN-γ, which was added to mimick the pro-inflammatory cytokine milieu of the early post-transplantation period. Next, we investigated the cytolytic capacity of LB-ARHGDIB-1R specific T-cells, and demonstrated specific lysis of patient, but not donor, EBV-B cells and specific lysis of an ALL sample in a 10 hrs 51Cr-release assay. Specific lysis of additional ALL and AML samples could be measured after 48 hrs of co-incubation in a flowcytometry-based cytotoxicity assay. To determine the in vivo immunogenicity of LB-ARHGDIB-1R, 11 MiHA-disparate HLA-B* 07:02 positive patient-donor pairs were screened for specific CD8+ T-cells by dual color tetramer analysis. All patients received partial T-cell depleted alloSCT and sampling was done at different time points after alloSCT (and DLI). In 4 out of 11 patients, LB-ARHGDIB-1R-specific T-cells (>0.01%) could be detected directly ex vivo and in 4 additional patients after 7 days of in vitro peptide stimulation, indicating that LB-ARHGDIB-1R is highly immunogenic. High frequencies of LB-ARHGDIB-1R specific T-cells were measured ex vivo in a patient whose hematological relapse was successfully treated with DLI, without development of GvHD, further supporting the therapeutic relevance of LB-ARHGDIB-1R. In summary, we confirmed hematopoietic-restricted expression of LB-ARHGDIB-1R and demonstrated T-cell mediated lysis of primary leukemic cells in long-term co-incubation assays. Furthermore, we showed that LB-ARHGDIB-1R is highly immunogenic and that specific T-cells could be detected in a patient who responded to DLI in the absence of GvHD. Altogether, our data support the clinical relevance of LB-ARHGDIB-1R as therapeutic MiHA with the potential to shift the delicate balance between GvL and GvHD in favor of a desired anti-tumor effect. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3748-3748 ◽  
Author(s):  
David C. Fisher ◽  
Marianne Tawa ◽  
Michele Walsh ◽  
Rachael A Clark ◽  
Thomas S. Kupper

Abstract Abstract 3748 Poster Board III-684 Sezary Syndrome and other Cutaneous T Cell Lymphomas with peripheral blood involvement (collectively termed leukemic CTCL, or L-CTCL) are often refractory to multiple therapies. Alemtuzumab, an antibody directed against the pan-lymphocyte antigen CD52, has been used to treat multiple hematologic malignancies, including L-CTCL. However, conventional treatment regimens are associated with an increased risk of infections, and this is of particular concern in CTCL patients with compromised skin integrity. We have treated six L-CTCL patients with a modified regimen of alemtuzumab: 10mg/kg, subcutaneously, three times weekly for six weeks (1/3 of the conventional dose). All patients had had disease for >3 years duration, skin biopsies consistent with CTCL, elevated absolute CD4 counts, CD4/CD8 ratios of >10 and were refractory to at least two prior therapies, including extracorporeal photochemotherapy, interferon, and single and combination agent chemotherapy. In five of six patients, the malignant clone was identifiable using a Vb family specific monoclonal antibody; all cells of the malignant clonotype strongly expressed CD52. Within three weeks, all six patients achieved clearance of detectable T and B cells in peripheral blood, including loss of the malignant clone. In parallel, all patients developed clearing of erythroderma, relief of pruritus, and by the end of the six week treatment period, all patients had complete to near complete resolution of all clinical signs of disease. Isolation of T cells from a skin biopsy of one patient in complete remission revealed that <6% of T cells resident in skin expressed the malignant Vb subunit. At this reduced dose, no infectious complications were encountered. We conclude from that low-dose Aletuzumab is a well tolerated, safe, and highly effective therapy in a carefully selected population of patients with refractory L-CTCL. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A130-A130
Author(s):  
Hannah Knochelmann ◽  
Connor Dwyer ◽  
Aubrey Smith ◽  
Megan Wyatt ◽  
Guillermo Rangel RIvera ◽  
...  

BackgroundTranslation of novel T cell therapies is limited by cost and time-consuming protocols involving long-term T cell expansion. We found that shortening ex vivo expansion of either TCR-specific murine Th17 cells or human CAR Th17 cells licenses the cell product to eradicate large tumors in low doses and generate long-lived memory against tumor.1 Therapeutic Th17 cells induce the systemic release of IL-6, IL-17, GM-CSF, and MCP-1 among other cytokines in tumor-bearing hosts, reminiscent of clinical cytokine release syndrome. As the toxicity of cytokine release is managed in patients through IL-6 blockade, we addressed the impact of IL-6 on efficacy and durability of Th17 cell therapy. We hypothesized that IL-6, induced by Th17 cells, was fueling the durable memory properties of this cell product.MethodsTh17 cells were expanded ex vivo using the TRP-1 transgenic mouse model in which CD4+ T cells express a TCR that recognizes tyrosinase-related protein 1 on melanoma. Naïve CD4+ T cells were polarized to the Th17 phenotype and infused into mice with B16F10 melanoma after a nonmyeloablative total body irradiation (5 Gy) preparative regimen.ResultsIL-6 blockade, targeting either IL-6R or neutralization of the cytokine, did not significantly impact the primary immune response of adoptively transferred Th17 cells against tumor. However, administering IL-6 blockade acutely after Th17 transfer resulted in a higher incidence of tumor relapse upon secondary tumor challenge, thereby compromising long-lived antitumor immunity.1 Mounting a secondary response to tumor was dependent on CD4+ T cells, but not CD8+ T cells, persisting in the host. Mechanistically, IL-6 blockade reduced pSTAT3 and Bcl2 in transferred T cells but did not greatly impact the concentration of other systemic cytokines. As a small fraction of Tregs remain in the Th17 cell product ex vivo, we examined the engraftment of those Tregs after transfer. IL-6 was critical to suppress engraftment of FoxP3+ donor T cells from the CD4+ T cell product. Thus, IL-6 promoted robust tumor infiltration by donor effector over regulatory cells for early Th17 cells relative to cell products expanded longer durations ex vivo.1ConclusionsOverall, short-term expanded Th17 cells uniquely induced IL-6 unlike Th17 cells expanded longer ex vivo. IL-6 promoted Th17 survival, reduced engraftment of tumor-specific Tregs, and was critical to durable memory. This work may suggest that the universal strategy to inhibit IL-6 during cytokine release syndrome may come at the expense of long-term efficacy for specific cell therapy approaches.ReferenceKnochelmann HM, Dwyer CJ, Smith AS, Bowers JS, Wyatt MM, Nelson MH, Rangel Rivera GO, Horton JD, Krieg C, Armeson K, Lesinski GB, Rubinstein MP, Li Z, Paulos CM. IL-6 fuels durable memory for Th17 cell-mediated responses to tumors. Cancer Res. 2020. Epub ahead of print. PMID: 32561531.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3747-3747
Author(s):  
Anandharaman Veerapathran ◽  
Joseph Pidala ◽  
Francisca Beato ◽  
Xue-Zhong Yu ◽  
Claudio Anasetti

Abstract Abstract 3747 Evidence from animal studies has demonstrated that allogeneic regulatory T (Treg) cells can be used as effective therapeutic tools in the prevention of allograft rejection and graft-vs-host disease (GVHD). However, translating Treg-based therapies from animal models to human clinical trials requires methods for the isolation and expansion of functional Treg. Loss of function has been a concern for Treg expanded broadly with anti-CD3 and CD28 antibody-coated beads. Based on preclinical data in rodents, allospecific Tregs are anticipated to be more potent and exert more selective immunoregulation than polyclonal Treg in the prevention or treatment of GVHD. Currently, no effective approach has been established for selective expansion of human allospecific Treg ex vivo. In this study, we show the selective direct and indirect recognition of alloantigen by specific human CD4+CD25+CD127− Tregs. CD4+CD25+CD127− Treg were freshly isolated from normal human blood, labeled with CFSE and stimulated by MHC (HLA)-mismatched APC's, in the presence of IL-2 and IL-15 to amplify the proliferative responses, and sirolimus to suppress other T cells. The precursor frequency of the antigen specific Tregs was calculated by Limiting Dilution Analysis and found to be 1/49 -1/138 among all Tregs. The expanded antigen specific Tregs with low CFSE content were in general greater than 90% by days 12–15 in culture. CFSE-low Tregs were sorted by flow cytometry and exhibited complete suppression against CD25-negative CD4 T cell responses up to ratios of 1:100, in contrast CFSE-high Tregs were not suppressive. Ex vivo expanded antigen specific Tregs maintained high Foxp3, retained lymphoid homing receptor CD62L and chemokine receptor CCR7 expression, suggesting that they are functional and able to migrate to lymphoid tissue in vivo. Among the APC's tested, DC was found to be better stimulus with potent suppressive potential when compared to T-cell stimulated PBMC's. Specific indirect allorecognition was elicited when Tregs were stimulated with autologous APC's pulsed with allogeneic cell lysate in the presence of sirolimus, IL-15 and IL-2. Precursor frequency of antigen specific Tregs with indirect allorecognition was 100 fold lower than the precursor frequency of Tregs with direct allospecificity. Highly suppressive antigen specific Tregs show a duplication time of approximately 24 hours in the presence alloantigen, cytokines and sirolimus, and can be expanded in vitro by 400 fold in a 12 day period. This magnitude of expansion predicts the feasibility of conducting translational clinical trials. Our data may provide a platform for the selective expansion of Tregs against minor histocompatibility antigens to prevent GVHD while sparing graft-vs.-leukemia effects. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 28-28
Author(s):  
Sinéad Kinsella ◽  
Cindy Evandy ◽  
Kayla S Hopwo ◽  
Kirsten Cooper ◽  
Lorenzo Iovino ◽  
...  

T cell reconstitution after transplant is critically dependent on the thymus; an inverse relationship between a transplant recipient's age and their capacity to generate T lymphocytes (in particular CD4+T cells) has been found in several studies, and thymic function pre-transplant can have a significant impact on clinical outcomes. Although the thymus has a remarkable ability to repair following damage, the mechanisms underlying this endogenous regeneration remain poorly understood. Despite this regenerative capacity, delayed T cell reconstitution is associated with an increased risk of infections, relapse of malignancy and the development of secondary malignancies. Therefore, there is a clinical demand for therapeutics that restore immune function after damage. Our recent studies have identified two key pathways driving thymic regeneration; centered on the secretion of BMP4 by endothelial cells (ECs) and IL-22 by innate lymphoid cells (Dudakov 2012 Science 336:91; Dudakov 2017 Blood130:933; Wertheimer 2018 Sci Immunol3:19). However, the specific regulatory mechanisms that trigger these regeneration-associated factors after damage remain unclear. Our previous work identified that the presence of homeostatic apoptotic CD4+CD8+ (DP) thymocytes, as apoptotic thymocytes form the bulk of developing T cells, suppress the production of IL-23 in dendritic cells (DCs), a key downstream mediator for IL-22, and BMP4 in ECs (Fig. 1A), and that the depletion of apoptotic thymocytes after damage precedes the production of these regenerative factors. Therefore, together with our findings that the metabolic needs of key thymus populations alter drastically following injury due to damage-induced metabolic remodeling, we hypothesized that further to the loss of DP-specific suppression, metabolic dysfunction in DPs after damage triggers mitochondrial-induced pyroptotic cell death, which can directly promote regeneration of the thymus. Consistent with this hypothesis, our preliminary data shows increased levels of cl-caspase 1 (pyroptotic caspase) and a decrease in cl-caspase 3 (apoptotic caspase) in DPs after SL-TBI (550 cGy), demonstrating a preferential induction of pyroptotic cell death in DPs after damage (Fig. 1B). Furthermore, we demonstrated an increase in extracellular lactate dehydrogenase (LDH) levels, HMGB-1 and TNF⍺[canonical damage-associated molecular patterns (DAMPs) released during ICD] acutely after damage caused by SL-TBI (Fig. 1C).Given our previous findings that stromal cells are more radio-resistant than DP thymocytes (Wertheimer 2018 Sci Immunol3:19), and evidence for mitochondrial-induced pyroptosis, we identified hyperpolarization of the mitochondrial membrane potential accompanied by increased levels of ROS in DPs, an effect not observed in TECs, suggesting metabolic stability confers protection against acute damage (Fig. 1D). Furthermore, co-culture of pyroptotic thymocytes results in increased IL12p40+ DCs and increased Foxn1 expression in TECs (Fig. 1E), strengthening our hypothesis that cell-cell communication drives thymic regeneration after damage by inducing regenerative factors as well as directly promoting TEC function via secreted factors from pyroptotic DPs. One way in which DAMPs, such as ATP, can initiate cell signaling is by the activation of cell surface purinergic receptors, including P2Y2 which is widely expressed on TECs, and here we demonstrate that in vitro treatment with ATP or P2Y2 agonist increases Foxn1 in cTECs, and P2Y2 antagonism reverses this effect (Fig 1F). As P2Y2 activation promotes Ca2+efflux from the ER, we have further demonstrated that stimulating the intracellular release of Ca2+, using tunicamycin, induced Foxn1 expression in cTECs, which was reversed upon inhibition of Ca2+release (Fig. 1G). Importantly, we demonstrate here that this pathway can be therapeutically targeted by activating P2Y2 signaling in vivo with MRS2568 or ATP enhances thymus cellularity and expands cTECs in models of acute injury (Fig. 1H&I). These findings not only reveal a novel metabolic-mediated molecular mechanism governing tissue regeneration; but also by targeting FOXN1 directly offers a potentially superior therapeutic strategy for boosting thymic regeneration and T cell reconstitution after damage such as that caused by HCT, infection or cytoreductive therapy. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2439-2439
Author(s):  
Paul A. Castillo ◽  
Jan J Melenhorst ◽  
Patrick J Hanley ◽  
Michael Keller ◽  
Robert A. Krance ◽  
...  

Abstract Refractory acute lymphoblastic leukemia (ALL) remains a disease with poor prognosis, and a small percentage of patients that undergo allogeneic stem cell transplantation will respond. Following allogeneic transplant, donor T-cells may cause graft-versus-host (GVH) alloreactions but also confer immunity to viruses and exert a graft-versus-leukemia (GVL) effect contributing to maintenance of minimal residual disease and cure of leukemia. Control of viral disease can be enhanced by infusion of ex-vivo expanded virus-specific T-cells. Since some malignancies such as Epstein-Barr virus (EBV)-driven lymphoproliferative diseases can express viral antigens they can be successfully eradicated by EBV specific CTL. However cross-reactivity between virus-specific T cells and non-viral hematological malignancies has not been described. Here we report for the first time an apparent GVL effect against ALL from ex-vivo expanded multivirus specific CTL. A 12-year-old boy with refractory ALL received a haploidentical transplant from his mother. As prophylaxis for EBV, CMV and adenovirus, he received ex-vivo expanded virus-specific donor T-cells 3.5 months after HSCT. Four weeks later leukemic blasts bearing the E2A deletion, appeared transiently in the blood followed by a FISH negative hematological remission, which was sustained until a testicular relapse 3.5 months later. The infused T cell line displayed predominant reactivity against CMV antigens and epitope mapping indicated broad reactivity with a number of pp65-derived epitopes including 1.88% of CD8+ T-cells that recognized the HLA-A24-restricted pp65 epitope QYDPVAALF. Within 2weeks post-CTL, markedly increased frequencies of CMV and adenovirus-specific T-cells were detectable in the patient suggesting that the virus specific T-cells expanded in-vivo. The occurrence of leukemic relapse followed by remission at the peak of virus-specific immune reconstitution suggested that virus-specific T cells were reactive against host leukemia. The observation conformed more with a GVL rather than a GVH effect since the patient did not develop GVHD following the T-cell infusion and no anti-recipient CTL activity was demonstrated. To explore leukemia-specific targeting by the virus-specific CTL, the T cell line was incubated with patient PHA blasts, patient ALL blasts and with donor EBV-LCL loaded with the CMV pp65 peptide library. CD4+ T cells, and, to a lesser extent CD8+ T cells, displayed a strong, polyfunctional reactivity against patient leukemia blasts, indicating that the virus-specific T cells were cross-reactive with the leukemia cells. (Figure 1a and 1b) Additionally, using IFN-γ ELISpot, the virus-specific T cells not only recognized viral-derived overlapping peptides, but also peptides derived from the tumor- associated antigens (TAA), Wilm's tumor-1 (WT-1), preferentially expressed antigen in melanoma (PRAME), and melanoma-associated antigen-3 (MAGEA3) known to be expressed at high frequencies on ALL blasts. (Figure 2) The possibility that the effect was mediated by anti-leukemic NK cells was eliminated by gating CD4+ and CD8+ T-cell subpopulations from the CTL line, separately. Despite a temporary remission, our patient eventually relapsed in the testis, recognized as an immune privileged leukemia sanctuary site and at a time when CMV, EBV or AdV specific T cells were no longer detectable in the peripheral blood. Evidence for a role of virus reactivation in reducing leukemia relapses post allogeneic HSCT has accumulated. This is, to our knowledge, the first report of virus-specific T cells with heterologous reactivity against ALL cells eliciting a GVL response without GVHD suggesting the need for further studies to identify how frequently virus-specific CTL lines can recognize and kill leukemia. Figure 1 Figure 1. Figure 2 Figure 2. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 4288-4288
Author(s):  
Laura Llaó Cid ◽  
Philipp M Rößner ◽  
Ekaterina Lupar ◽  
Bola S Hanna ◽  
Jérôme Paggetti ◽  
...  

Introduction: Genome-wide association studies showed that a single-nucleotide polymorphism (SNP) affecting the transcription factor Eomesodermin (Eomes) is associated with a significantly increased risk to develop chronic lymphocytic leukemia (CLL). Eomes and its paralogue T-bet are known master regulators of CD8+ effector T cells and CD4+ T helper cells and critical for T cell-mediated immune responses against pathogens and cancer. While Eomes has been shown to be essential for effector function of CD8 T cells, its role in CD4 T cells is less well understood. Recent work suggested that Eomes drives the development of IL-10 and IFNγ co-producing, FoxP3-negative, regulatory T cells, named Tr1 cells, a population that was found enriched in inflamed tissues in patients with chronic inflammatory disorders. In CLL, the T cell compartment is altered with an enrichment of effector and memory T cells that were shown to control leukemia development in a mouse model, but also to acquire a dysfunctional or exhausted state. Methods: We investigated Eomes-expressing CD4 and CD8 T cells in blood and lymph node (LN) samples of patients with CLL, as well as in the Eµ-TCL1 mouse model of CLL by flow cytometry, CyTOF, mRNA sequencing, and ex vivo functional assays. The role and function of these cells was explored in bone marrow-chimeric mice harbouring an Eomes-deficient hematopoietic microenvironment that were used for adoptive transfer of TCL1 leukemia, as well as by co-transfer experiments of Eomes- or IL-10R-deficent CD4 or CD8 T cells with TCL1 leukemia cells in Rag2-/- mice lacking B and T cells. Results: We detected an accumulation of Eomes-expressing CD4 and CD8 T cells in CLL patients, which was more severe in LN compared to blood samples, and significantly stronger in CLL LN compared to reactive LN samples as non-cancer control (Fig. 1A). This was in line with an observed expansion of Eomes-positive T cells in the spleen of leukemic Eµ-TCL1 mice and upon adoptive transfer of TCL1 leukemia. Eomes expression in CD8 T cells correlated with the expression of CD69, IFNγ and PD-1, suggesting a link between Eomes and CD8 T cell activation and function in CLL. The importance of Eomes in CD8 T cell-mediated control of CLL was demonstrated in mice that were transplanted with TCL1 leukemia, where Eomes-deficient CD8 T cells failed to control leukemia development. As we detected significantly less Eomes-deficient CD8 T cells in these mice compared to respective wildtype controls, and a lower percentage of them was positive for the proliferation marker Ki-67, we conclude that Eomes drives the differentiation and expansion of CD8 T cells in mice with CLL-like disease. We further explored Eomes-expressing CD4 T cells in CLL by transcriptome analysis, flow cytometry and ex vivo functional assays, and observed increased expression of IFNγ and IL-10, as well as inhibitory receptors, like PD-1, BLIMP-1 and LAG3, features that are described for Tr1 cells. Transfer of Eomes-deficient or wildtype CD4 T cells in Rag2-/- mice that were injected with TCL1 leukemia cells, lead to a comparable expansion of CD4 T cells independent of Eomes. But even though wildtype CD4 T cells were able to control leukemia development in this setting, Eomes-deficient CD4 T cells failed to do so (Fig. 1B). As Eomes is a known driver of IL-10 expression, we tested whether IL-10R signalling in CD4 T cells is involved in the anti-tumor activity by performing respective CD4 T cell transfer experiments comparing this time wildtype with IL-10R-deficent CD4 T cells. Interestingly, lack of IL-10R in CD4 T cells lead to a reduction in anti-tumor control (Fig. 1C) and therefore suggests that IL-10 is involved in Eomes-driven regulation of CD4 T cell-mediated immune control. Conclusions: In summary, we conclude that Eomes is required for CD8 T cell-mediated control of CLL, and Eomes+ PD-1+ IL-10-producing CD4 T cells contribute to adaptive immunity in CLL. The increased risk of developing CLL in individuals harbouring a SNP in the Eomes gene might be therefore explained by a negative impact of this alteration on CD4 and/or CD8 T cell-mediated immune control of CLL. Disclosures Stilgenbauer: Novartis: Consultancy, Honoraria, Research Funding, Speakers Bureau; Amgen: Consultancy, Honoraria, Research Funding, Speakers Bureau; Celgene: Consultancy, Honoraria, Research Funding, Speakers Bureau; Gilead: Consultancy, Honoraria, Research Funding, Speakers Bureau; GSK: Consultancy, Honoraria, Research Funding, Speakers Bureau; Hoffmann La-Roche: Consultancy, Honoraria, Research Funding, Speakers Bureau; Pharmacyclics: Other: Travel support; AbbVie: Consultancy, Honoraria, Research Funding, Speakers Bureau; Janssen: Consultancy, Honoraria, Research Funding, Speakers Bureau; AstraZeneca: Consultancy, Honoraria, Research Funding, Speakers Bureau.


Sign in / Sign up

Export Citation Format

Share Document