A Randomized Trial to Evaluate the Impact of Cytokines on Dendritic Cell, T-Cell and T-Cell Function When Mobilizing Normal Donors for Allogeneic Progenitor Cell Transplant: An Interim Analysis.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2854-2854
Author(s):  
Sagar Lonial ◽  
Claire Torre ◽  
Michelle Hicks ◽  
Stephanie Mcmillan ◽  
Amelia A. Langston ◽  
...  

Abstract Introduction:Optimal cellular immunity following allogeneic HPC transplant represents a balance between the induction of sufficient anti-tumor immunity to eradicate residual cancer cells without the induction of life-threatening GvHD. Dendritic cells are potent APCs with the ability to regulate immune responses. Our group has previously reported that increased numbers of donor DC2 result in inferior EFS following allo BMT (Waller et al, Blood 2001), and that myeloid cytokines used for mobilization modulate the DC content of the auto graft (Lonial et al, BBMT in press). The current trial was designed to evaluate the impact of different cytokine combinations on DC content and T-cell function in normal donors mobilized with either G-CSF or the combination of G-CSF + GM-CSF. Methods: 32 normal donors were randomized to mobilization with G-CSF (7.5 mcg/kg BID) or the combination of GM-CSF (7.5 mcg/kg qAM) + G-CSF (7.5 mcg/kg qPM) until completion of the stem cell collection. Side effects between the 2 regimens were documented using a questionnaire filled out by the donors within 2 weeks of stem cell collection. DC, T-cell, and other cell subsets were measured from the graft using flow cytometry. T-cell function was evaluated by measuring T-cell proliferation in response to PMA, Con A, PHA, and PWM. Cytokines (IL2, IL4, IL10,IL12, TNF, and INF) secreted in response to antigens were measured by ELISA. DC1 (myeloid DC) were defined as Lin-/HLA-DR+/CD11c+/CD123- while DC2 (lymphoid DC) were defined as Lin-/HLA-DR+/CD11c-/CD123+. Results: 28 patients have been successfully collected to date (G-CSF n=15, GM+G-CSF n=13). No donor has failed to mobilize in either group. Among the 15 donors mobilized with G-CSF alone, 5 required multiple days of apheresis as compared with 1 of 13 donors who received GM+G-CSF who required multiple days of apheresis (p=0.06). There was no difference in baseline values of T-cells or DC subsets in the peripheral blood prior to cytokine administration. Grafts collected with GM-CSF+ G-CSF contained significantly fewer DC2 cells and T-cells (median DC2 dose of 2.1 x 10E6/kg and CD3 dose of 197x 10E6/kg) compared with grafts from donors who received G-CSF alone (median DC2 dose of 3.8 x 10E6/kg (p=.01) and CD3 dose of 320 x 10E6/kg (p=0.001)). There was no difference in the content of CD34+ or DC1 in the grafts, nor in the ratio of CD4:CD8 T-cells between grafts collected with the 2 cytokine combinations. T-cell proliferation and cytokine secretion in response to mitogens was not different between grafts collected from the two groups. To date, there is no difference in the frequency of GvHD or relapse between the patients transplanted with the grafts collected from the 2 cytokine cohorts. Conclusions: The addition of GM-CSF to the mobilization regimen results in significantly fewer DC2 cells and T-cells in the blood HPC graft which could impact immune function and GvL following allogeneic HPC transplant. Clinical outcomes and further analysis of TH1/TH2 polarization of T-cells in grafts collected with either G-CSF or G-CSF+GM-CSF are in progress..

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 986-986 ◽  
Author(s):  
Christina Krupka ◽  
Franziska Brauneck ◽  
Felix S Lichtenegger ◽  
Peter Kufer ◽  
Roman Kischel ◽  
...  

Abstract Bispecific T-cell engager (BiTE®) antibodies represent a promising tool for anti-leukemic immunotherapy. The CD19/CD3-bispecific antibody blinatumomab was shown to be active in refractory and relapse patients with B-precursor acute lymphoblastic leukemia (Topp et al, ASCO 2014). Transient, blinatumomab-mediated cytokine release syndrome has been linked to target cell numbers as this phenomenon is predominantly observed within the first treatment cycle. In our previous work, we demonstrated that the bispecific CD33/CD3 BiTE® antibody AMG 330 is able to induce activation and proliferation of residual autologous T-cells and effectively mediates lysis of primary acute myeloid leukemia (AML) cells (Krupka et al, Blood 2014; 123(3):356-65). We hypothesize that in AML patients with high initial leukocyte counts (WBC > 30.000/μl) a cytoreductive phase prior to AMG 330 therapy might be beneficial to reduce the incidence and severity of cytokine mediated toxicity. Ideally, the cytoreductive drug does not impair T-cell function or reduce target antigen expression level. In the current study, we evaluated the effect of cytarabine (20 µM), decitabine (5 µM), azacitidine (1 µM and 5 µM) and hydroxyurea (10 µM and 100 µM) on T-cell proliferation and function in close analogy to potential treatment algorithms for AML. Healthy donor (HD) T-cells were pre-incubated with the cytoreductive drugs for 72 hours. T-cells were CFSE-labeled and co-cultured with either HL60 or MV4-11 cells (effector cell:target (E:T) ratio 1:1) in the presence or absence of AMG 330 (5 ng/ml). After 3 days of co-culture, lysis of HL60 cells and T-cell proliferation was assessed by flow cytometry. Pretreatment of T-cells with cytarabine completely abrogated T-cell function (lysis of HL60 cells: untreated (UT): 96.9% vs 20 µM: 4.2%) and significantly impaired T-cell proliferation (UT: 31.2% vs 20 µM: 4.6%). These findings correlated to data using primary AML samples collected 3 and 6 days after discontinuation of cytarabine treatment. After a 3-day chemotherapy-free interval, we observed no relevant T-cell proliferation and lysis of AML cells upon the addition of AMG 330 to the ex-vivo long-term culture system (lysis of AML cells on day 12: 30%; fold change T-cell expansion 0.9). After a 6-day treatment-free interval, high T-cell proliferation and cytotoxicity against primary AML cells were observed (lysis of AML cells on day 12: 61%; fold change T-cell expansion: 3.1). In contrast to cytarabine, decitabine treatment only marginally impaired T-cell function. Similarly, pre-incubation with azacitidine did not convey a negative effect on T-cell function (lysis of HL60 cells: UT: 100% vs 1 µM: 94.9% vs 5µM: 86.8%; proliferation: UT: 90.9% vs 1 µM: 80% vs 5 µM: 66.8%). Pretreatment with hydroxyurea had the least impact on T-cell performance. It did not impair T-cell function (lysis of HL60 cells: UT: 100% vs 10 µM: 100% vs 100 µM: 100%) and proliferation compared to untreated controls (UT: 92.9% vs 100 µM 90.8% vs 10 µM 92.9%). As we have previously shown that the level of CD33 expression correlates to kinetics of AMG 330-mediated lysis (Krupka et.al, EHA 2014), we analyzed the effect of the cytoreductive agents on CD33 expression level in AML cell lines and primary AML cells. Five AML cell lines (HL60, MV4-11, PL21, OCI-AML3, KG1a) and a primary AML patient sample were cultured in the presence or absence of decitabine (5 µM and 50 µM), azacitidine (1 µM and 5 µM) or hydroxyurea (10 µM and 100 µM) for 72 hours. The change of CD33 expression level was evaluated by flow cytometry (median fluorescence intensity, MFI). No significant changes in CD33 expression level were observed after culture of AML cell lines and primary AML cells with decitabine or azacitidine. In contrast, hydroxyurea upregulated surface expression of CD33 on 2/5 cell lines (HL60 and PL21) in a dose dependent manner (HL 60 MFI Ratio: UT 134.9 vs 10 µM 171.3 vs 100 µM 210; PL21 MFI Ratio: UT 166.9 vs 10 µM 177.9 vs 100 µM 191.8). In summary, we could show that pretreatment with hydroxyurea did not impair T-cell function and proliferation. In addition, we observed an upregulation of CD33 expression on AML cell lines. As the BiTE® technology relies on T-cell function and target antigen expression level, sequential and combinatorial immuno-chemotherapeutic approaches need to address both issues. Our data support the use of hydroxyurea in AML patients that require cytoreduction prior to AMG 330 treatment. Disclosures Krupka: AMGEN Inc.: Research Funding. Kufer:AMGEN Research (Munich): Employment; AMGEN Inc.: Equity Ownership. Kischel:AMGEN Research (Munich): Employment; AMGEN Inc.: Equity Ownership. Zugmaier:AMGEN Inc.: Equity Ownership; AMGEN Research (Munich): Employment. Sinclair:AMGEN Inc.: Employment, Equity Ownership. Newhall:AMGEN Inc.: Employment, Equity Ownership. Frankel:AMGEN Inc.: Employment, Equity Ownership. Baeuerle:AMGEN Research (Munich): Employment; AMGEN Inc.: Equity Ownership. Riethmüller:AMGEN Inc.: Equity Ownership. Subklewe:AMGEN Inc.: Research Funding.


2001 ◽  
Vol 193 (3) ◽  
pp. 317-328 ◽  
Author(s):  
Kanaga Sabapathy ◽  
Tuula Kallunki ◽  
Jean-Pierre David ◽  
Isabella Graef ◽  
Michael Karin ◽  
...  

Apoptotic and mitogenic stimuli activate c-Jun NH2-terminal kinases (JNKs) in T cells. Although T cells express both JNK1 and JNK2 isozymes, the absence of JNK2 alone can result in resistance to anti-CD3–induced thymocyte apoptosis and defective mature T cell proliferation. Similar defects in thymocyte apoptosis and mature T cell proliferation, the latter due to reduced interleukin 2 production, are also caused by JNK1 deficiency. Importantly, T cell function was compromised in Jnk1+/−Jnk2+/− double heterozygous mice, indicating that JNK1 and JNK2 play similar roles in regulating T cell function. The reduced JNK dose results in defective c-Jun NH2-terminal phosphorylation in thymocytes but not in peripheral T cells, in which nuclear factors of activated T cells (NK-ATs)–DNA binding activity is affected. Thus, JNK1 and JNK2 control similar functions during T cell maturation through differential targeting of distinct substrates.


Cells ◽  
2020 ◽  
Vol 9 (2) ◽  
pp. 300 ◽  
Author(s):  
Konstantina Antoniou ◽  
Fanny Ender ◽  
Tillman Vollbrandt ◽  
Yves Laumonnier ◽  
Franziska Rathmann ◽  
...  

Activation of the C5/C5a/C5a receptor 1 (C5aR1) axis during allergen sensitization protects from maladaptive T cell activation. To explore the underlying regulatory mechanisms, we analyzed the impact of C5aR1 activation on pulmonary CD11b+ conventional dendritic cells (cDCs) in the context of house-dust-mite (HDM) exposure. BALB/c mice were intratracheally immunized with an HDM/ovalbumin (OVA) mixture. After 24 h, we detected two CD11b+ cDC populations that could be distinguished on the basis of C5aR1 expression. C5aR1− but not C5aR1+ cDCs strongly induced T cell proliferation of OVA-reactive transgenic CD4+ T cells after re-exposure to antigen in vitro. C5aR1− cDCs expressed higher levels of MHC-II and CD40 than their C5aR1+ counterparts, which correlated directly with a higher frequency of interactions with cognate CD4+ T cells. Priming of OVA-specific T cells by C5aR1+ cDCs could be markedly increased by in vitro blockade of C5aR1 and this was associated with increased CD40 expression. Simultaneous blockade of C5aR1 and CD40L on C5aR1+ cDCs decreased T cell proliferation. Finally, pulsing with OVA-induced C5 production and its cleavage into C5a by both populations of CD11b+ cDCs. Thus, we propose a model in which allergen-induced autocrine C5a generation and subsequent C5aR1 activation in pulmonary CD11b+ cDCs promotes tolerance towards aeroallergens through downregulation of CD40.


2005 ◽  
Vol 289 (1) ◽  
pp. R37-R44 ◽  
Author(s):  
Xiaoling Li ◽  
Shadab N. Rana ◽  
Elizabeth J. Kovacs ◽  
Richard L. Gamelli ◽  
Irshad H. Chaudry ◽  
...  

Previous studies showed that alcohol (EtOH) intoxication before burn injury suppresses mesenteric lymph node (MLN) T cell functions and increases gut bacterial translocation. In this study, we examined whether corticosterone (Cort) plays any role in suppressing MLN T cell function and bacterial accumulation after EtOH intoxication and burn injury. Rats were gavaged with EtOH to achieve a blood EtOH level of ∼100 mg/dl before receiving 25% total body surface area burn or sham injury. A group of rats was treated with the Cort synthesis inhibitor metyrapone (25 mg/kg) at the time of injury and on day 1 after injury. Two days after injury, a significant increase in blood Cort levels and suppression of MLN T cell proliferation and IL-2 production was observed in rats receiving combined insult of EtOH intoxication and burn injury compared with rats receiving EtOH intoxication or burn injury alone. There was no change in T cell apoptosis after combined insult of EtOH and burn injury. Furthermore, T cell suppression was accompanied by a significant decrease in p38 and ERK1/2 activation (phosphorylation). There was no difference in JNK activation after EtOH and burn injury. Treatment of rats with metyrapone prevented the suppression of MLN T cell proliferation, IL-2 production, and p38 and ERK1/2 phosphorylation. Restoration of T cell function in metyrapone-treated animals was also associated with the decrease in bacterial accumulation in MLN. These findings suggest that EtOH intoxication before burn injury augments Cort release, which suppresses MLN T cell function by inhibiting p38 and ERK1/2 activation and promotes bacterial accumulation in MLN after EtOH and burn injury.


2017 ◽  
Vol 1 (4) ◽  
pp. 1-13 ◽  
Author(s):  
Flor C. Navarro ◽  
Stephanie K. Watkins

Sex-specific differences exist in innate and adaptive immune responses and are mediated by hormone signaling. Estrogen is able to differentially modulate the development and differentiation of immune cells, including T cells. However, the effect of estrogen on T cell function, especially at concentrations other than physiological, remains controversial and incompletely understood. Immunotherapy is one of the most promising cancer treatments to date with a high probability of future enhancements. The adoptive transfer of genetically modified T cells can mediate tumor regression but there are still many hurdles to enhancing the proficiency of this treatment. This study demonstrates for the first time that one major aspect to consider for designing potent immunotherapies for cancer is the impact of the patient's sex. Herein, using two different Ag-specific T cell groups, we investigated the effect of sex and estrogen in antitumor effector responses, T helper cytokine secretion, and, importantly, on T cell whole polyfunctionality important for memory T cell development and survival. Major differences were observed in T cell function and polyfunctionality between sexes and on E2 treatment. The findings of this study may be critical to understand the results of immunotherapy on different patients and for the enhancement of immunotherapy for cancer.


2018 ◽  
Vol 14 (9) ◽  
pp. e1007289 ◽  
Author(s):  
Asma Ahmed ◽  
Vasista Adiga ◽  
Soumya Nayak ◽  
J. Anto Jesuraj Uday Kumar ◽  
Chirag Dhar ◽  
...  

2020 ◽  
Vol 33 (Supplement_1) ◽  
Author(s):  
N Donlon ◽  
A Sheppard ◽  
M Davern ◽  
C Donohoe ◽  
N Ravi ◽  
...  

Abstract   There is extensive literature demonstrating CD8+ T cells are essential for initial tumour control following radiation, however, effects are reduced after time due to T cell exhaustion and a lack of release Damage Associated Molecular Patterns (DAMPS) which are essential for anti-tumour immune responses. In vivo, activated T-cells migrate to the tumour site within the field of irradiation, however translational studies on the effects of radiotherapy on T-cell activation, function and activity are lacking. Methods EAC patient (n = 6) PBMCs were isolated by density centrifugation in Ficoll Paque. T cells were activated and were irradiated at 1.8Gy, 3.6Gy bolus dosing and fractionation for 72 hrs. A panel of immune checkpoints, DAMPS, activation markers, and cytokines were assessed by flow cytometry. To determine the effect of the TME on T cells, PBMCs were cultured under conditions of nutrient deprivation (No Glucose & No Glutamine) under conditions of normoxia and hypoxia. We then ran the aforementioned panel by flow cytometry. We also activated PBMCs with immune checkpoint blockers to determine its effects on T cell expansion and survival. Results 3.6Gy induced a significantly higher expression of DAPMS (Fig 1 p < 0.001); Calreticulin and HMGB1, most notably under conditions of nutrient deprivation (p < 0.001). Ionising radiation also resulted in an increase in the expression of cytokines and importantly in the context of targeted therapy, IR at both the conventional 1.8Gy and 3.6Gy induced a higher expression of checkpoints PD-1, PD-L1, TIGIT, and TIM-3 (p < 0.001). Interestingly, when T cells are activated in the presence of ICB (Atezolizumab, Pembrolizumab, Nivolumab), it increases the rate of T cell expansion, and enhances their survival compared to T cell activated only. (p < 0.001). Conclusion This work demonstrates the impact of clinically utilised fractions of radiation, and conditions of the TME on T cell function and activity, with improved T cell expansion and survival in the presence of ICB’s suggesting it may be a feasible combination therapy as an adjunct to radiotherapy.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1410-1410 ◽  
Author(s):  
Florian Perner ◽  
Felix C Saalfeld ◽  
Tina M Schnoeder ◽  
Denise Wolleschak ◽  
Corinna Fahldieck ◽  
...  

Abstract Inhibitors of JAK2-kinase (Ruxolitinib, Momelotinib) are already approved or currently investigated in advanced clinical trials for treatment of myeloproliferative neoplasia (MPN). Besides their effect on mutated JAK2-kinase these compounds inhibit wildtype JAK and thereby impair JAK-STAT-signaling, which is an important pathway for proliferation and activation of other cell types such as human T-cells. Accumulating evidence suggests that they may also exert substantial immunosuppressive activity. Very recent reports highlighting hepatitis B reactivation complemented the series of severe infections in ruxolitinib-treated patients among which cryptococcus neoformans pneumonia, toxoplasmosis retinitis, disseminated tuberculosis, and progressive multifocal leukencephalopathy are the most alarming. We hypothesized that JAK-kinase inhibitors may act as immunosuppressant drugs by impairment of T-cell responses through inhibition of T-cell signaling (JAK-STAT pathway) and that specificity of JAK-kinase inhibition may be of major importance for the degree of T-cell inhibition. Therefore we investigated the effects of pharmacological JAK-kinase inhibition on healthy donor (HD-) and MPN patient T-cells. Selective inhibitors of JAK2-kinase (BSK805) and JAK3-kinase (BQM245) as well as clinically relevant inhibitors of JAK1/2-kinases (Ruxolitinib and Momelotinib) were used for pharmacologic inhibition. The SRC-kinase inhibitor Dasatinib served as a positive control for T-cell inhibition. Knockdown of specific JAK-kinases by RNAi was used to control for target specificity. In regard to T-cell receptor (TCR)-mediated signaling we investigated bona fide signaling molecules downstream of the TCR by Western Blotting. Besides SRC-kinases like LCK also ZAP70, PLCG1 and the MAPK/ERK pathway have been described to play a pivotal role in T-cell activation. In our data set, selectivity of JAK-kinase inhibition (JAK2, JAK3 or JAK1/2) influenced TCR-signaling in regard to overall tyrosine phosphorylation but also in regard to downstream effectors such as ERK. As activation and proliferation of primary T-cells is a critical step in immune responses against viral and tumor antigens we aimed to investigate the influence of JAK-kinase inhibition on activation and proliferation of human T-cells. T-cells from healthy donors were stimulated using either PHA 0.5% or CD3/CD28 beads to ensure a more T-cell receptor specific stimulation. CD69 expression was used as a marker for T-cell activation and CFSE staining was applied to assess for T-cell proliferation. Using CD3/CD28 stimulation, CD69 expression was almost abrogated following Dasatinib treatment and proliferation was significantly reduced. Applying relevant doses of specific JAK2 and JAK3 inhibitors to isolated T-cells did neither influence CD69 expression nor T-cell proliferation. These findings are confirmed by RNAi. In contrast, clinically relevant doses of JAK1/2 inhibitors Ruxolitinib and Momelotinib, respectively reduced CD69 expression and T-cell proliferation. Likewise, T-cells derived from MPN patients treated with Ruxolitinib revealed decreased CD69 expression and decreased proliferative capacity upon stimulation, compared to untreated patients or HD-controls. In order to investigate T-cell function, we assessed for allo-reactivity in a mixed lymphocyte culture. Human pan-T-cells were co-cultured with allogeneic antigen presenting cells. T-cell reactivity – as measured by 3H-thymidine incorporation – was significantly impaired by Ruxolitinib and Momelotinib. Specific inhibition of JAK2 or JAK3 kinase, however, did not affect T-cell reactivity. These effects could be confirmed using T-cells derived from Ruxolitinib-treated MPN patients. Investigation of leukemia- and virus-antigen-specific T-cell responses are currently under way to gain deeper insight regarding this clinically relevant scenario. Taken together, specificity of JAK-kinase inhibition influences the inhibitory potential on T-cell function. JAK1 kinase seems to play an important role in T-cell activation, as unspecific inhibitors of JAK1 & JAK2 Kinase inhibit T-cell function while selective inactivation of JAK2 kinase leaves T-cell function almost unaffected. Heterogeneity in T-cell function of Ruxolitinib-treated patients is an important finding that deserves detailed investigation. Disclosures Heidel: Novartis: Consultancy.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3341-3341
Author(s):  
Victor Tkachev ◽  
Scott N. Furlan ◽  
Ben Watkins ◽  
Betty Zheng ◽  
Daniel Hunt ◽  
...  

Abstract While calcineurin inhibition (CNI)-based strategies remain the mainstay for GVHD prevention, CNI are notoriously antagonistic to immune tolerance induction. Rapamycin (Rapa) has been shown to be more pro-tolerogenic; however, the best agents to combine with Rapa are still undetermined, and it remains a second-line GVHD prevention strategy without clear superiority over CNI. Finding tolerogenic partners for Rapa, therefore, represents a critical unmet need in the field. Of the possible partners for Rapa, the OX40/OX40L pathway represents an important target. OX40 is a costimulatory receptor expressed on activated human T cells, which, upon interaction with OX40L delivers activation signals to conventional T cells (Tconv) promoting their proliferation, survival and clonal expansion. Notably, these same OX40/OX40L signals may either inhibit or promote Treg functions, depending on context, suggesting that blockade of this pathway may simultaneously control Tconv activation while permitting Treg homeostasis. During GVHD in non-human primates (NHP), we found OX40L upregulation on myeloid dendritic cells and OX40 upregulation on activated T cells in recipients treated with multiple immunosuppressive agents, including Rapa (Fig 1). These data provided strong rationale for testing KY1005, a novel human monoclonal antibody that binds to OX40L and blocks its interaction with OX40, as a potential partner with Rapa. We tested the outcomes of prophylactic blockade of this pathway on NHP GVHD, using KY1005 alone and in combination with Rapa. These experiments utilized our previously published NHP GVHD model, in which GVHD is studied after T cell-replete haplo-identical HCT. KY1005 was dosed at 10mg/kg weekly from days -2ˆ+54 and Rapa was continued through Day +100. Prophylaxis with KY1005 alone provided initial evidence for its in vivo activity, with control of CD4>CD8 T cell proliferation and mitigation of the expansion of CD4>CD8 T effector/memory cells. Consistent with the partial control of T cell activation, these recipients demonstrated improved GVHD-free survival versus unprophylaxed controls, but disease ultimately broke through (Median Survival Time (MST) = 19.5 days with KY1005 (n=4) compared to 8 days in unprophylaxed recipients (n= 10, Fig 2)). We next investigated the impact of OX40L blockade + Rapa. We have published that Rapa as a monotherapy minimally controlled both immunologic and clinical disease, with an MST = 14 days (n=6). Combined prophylaxis was striking: recipients given KY1005+Rapa (n=5) maintained robust health throughout the entire experiment (MST >100d), and demonstrated high levels of donor T cell chimerism (86 +/- 3% at Day 100), rapid hematopoietic reconstitution, and had a terminal GVHD Grade of 0, compared to a Grade of III-IV in both KY1005- and Rapa-monotherapy cohorts. Immunologic analysis demonstrated synergistic control of both CD4 and CD8 T cell proliferation, restoring it to the level observed during autologous immune reconstitution, and resulting in a concomitant abrogation of CD4 and CD8 memory/effector expansion while preserving T cells with a na•ve phenotype. In striking contrast to the inhibition of Tconv activation by KY1005+Rapa, recipients of dual therapy demonstrated intact Treg reconstitution post-HCT, which resulted in a favorable Treg:Tconv ratio of 5.4 vs 1.4:100 in KY1005+Rapa treated compared to untreated recipients (p < 0.05). Transcriptomic analysis confirmed the unique immunologic state conferred by KY1005+Rapa on purified T cells, with gene arrays from these recipients demonstrating separation from all other transplant cohorts in Principal Component space (Figure 3A) and Class Neighbor Analysis identifying unique expression modules that tracked with KY1005 + Rapa prophylaxis (Figure 3B red and blue boxes). These results underscore the critical role of OX40/OX40L signaling in the development of GVHD and demonstrate the striking control of GVHD in KY1005+Rapa recipients. They represent the first demonstration of uniform, long-term GVHD-free survival in the primate model of high-risk haplo-identical HCT, and the first therapeutic strategy that simultaneously controls Tconv activation while supporting Treg homeostasis in this model. They suggest that OX40L blockade + Rapa is a novel, evidence-based combinatorial strategy to control GVHD that is an exceptional candidate regimen for clinical translation. Disclosures Tkachev: Kymab Ltd: Patents & Royalties: US Patent 9,382,325, Research Funding. Casson:Kymab Ltd: Employment. Kirby:Kymab Ltd: Employment, Patents & Royalties: US Patent 9,382,325. Bland-Ward:Kymab Ltd: Employment, Patents & Royalties: US Patent 9,382,325. Kean:Juno Therapeutics, Inc: Research Funding.


2004 ◽  
Vol 36 (11) ◽  
pp. 741-748 ◽  
Author(s):  
Tie-Cheng Yu ◽  
Yi Liu ◽  
Yan Tan ◽  
Yanfang Jiang ◽  
Xueqing Zheng ◽  
...  

Abstract Shock waves were elicited by transient pressure disturbances, which could be used to treat musculoskeletal disorders. In present studies, we investigated whether the low-density shock waves (LDSWs), which are able to damage plasma membrane without impairing the vimentin or other organelles, might augment T-cell proliferation as well as IL-2 expression, and if mitogen activated protein kinase p38 (p38 MAPK) might be an underlying mechanism through which the LDSWs enhanced T-cell function. We found that the LDSWs increased activation of p38 MAPK in Jurkat T cells. The LDSWs alone didn't result in the T-cell proliferation and IL-2 expression. However, in combination with other stimuli, LDSWs could augment the T-cell proliferation and IL-2 expression. Inhibition of p38 MAPK using SB203580 reduced the stimulatory effects of the LDSWs, which indicated that the LDSWs enhanced IL-2 expression through a mechanism that involved p38 MAPK activation. We concluded that the p38 MAPK activation played a key role in the regulation of T cell function by the LDSWs.


Sign in / Sign up

Export Citation Format

Share Document