Siglec-F Promotes IL-33–Induced Cytokine Release from Bone Marrow–Derived Eosinophils Independently of the ITIM and ITIM-like Motif Phosphorylation

2022 ◽  
pp. ji2100184
Author(s):  
Stefanie Westermann ◽  
Axel Dietschmann ◽  
Daniela Doehler ◽  
Kirstin Castiglione ◽  
Bruce S. Bochner ◽  
...  
Keyword(s):  
2013 ◽  
Vol 31 (15_suppl) ◽  
pp. 10007-10007 ◽  
Author(s):  
Lia Gore ◽  
Gerhard Zugmaier ◽  
Rupert Handgretinger ◽  
Franco Locatelli ◽  
Tanya M. Trippett ◽  
...  

10007 Background: Pediatric B-precursor acute lymphoblastic leukemia (ALL) in second or later relapse is an aggressive malignancy that needs therapeutic approaches with new mechanisms of action. Blinatumomab, a bispecific T-cell engaging (BiTE) antibody, has shown a hematological remission rate of 69% in adult patients with relapsed/refractory ALL. In order to establish a recommended dose in pediatric patients, a phase I multicenter trial was initiated. Methods: The primary endpoint is to determine the maximum tolerable dose defined by ≤1 of 6 patients experiencing dose limiting toxicity (DLT) within the 1st course of treatment. Up to 6 different dose levels of blinatumomab are being evaluated. Eligible patients must be <18 years old and have B-precursor ALL that is refractory or in second or later bone marrow relapse, or in any marrow relapse after allogeneic hematopoetic stem cell transplantation (HSCT). Blinatumomab is administered by continuous IV infusion over 28 days followed by a 14-day treatment-free interval (up to 5 cycles). To date, 3 dose levels have been explored (Table). Results: Seventeen patients have been treated thus far with a total of 32 cycles. One DLT (gastrointestinal hemorrhage) at dose level 2 (15 µg/m²/d) and two DLTs at dose level 3 (30 µg/m²/d; both cytokine release syndrome) with 1 death have been observed. One patient had generalized seizures on the 3rd day of the 2nd treatment cycle at the first dose level of 5 µg/m2/d, which was completely reversible. The patient successfully underwent an allogeneic HSCT after blinatumomab. Eight (47%) of the 17 patients reached a cytological complete remission in bone marrow and a molecular remission defined as MRD by PCR <10-4. Conclusions: A phase I trial of blinatumomab in patients with relapsed/refractory pediatric ALL has shown hematological and molecular remissions. Dose-limiting cytokine release syndrome has been noted. Alternative dosing regimens are being explored in current cohorts to refine the recommended dose of blinatumomab in this patient population. Clinical trial information: NCT01471782. [Table: see text]


2021 ◽  
Vol 220 (3) ◽  
Author(s):  
Xingchen Dong ◽  
Xiangming Hu ◽  
Yan Bao ◽  
Guo Li ◽  
Xiao-dong Yang ◽  
...  

NLRC4 inflammasome activation and the subsequent maturation of IL-1β and IL-18 are critical for protection against infection by bacterial pathogens. The epigenetic regulator Brd4 has emerged as a key player in inflammation by regulating the expression of inflammatory cytokines. However, whether Brd4 has any role in inflammasome activation remains undetermined. Here, we demonstrated that Brd4 is an important regulator of NLRC4 inflammasome activation in response to Salmonella typhimurium infection. Brd4-deficient bone marrow–derived macrophages (BMDMs) displayed impaired caspase-1 activation, ASC oligomerization, IL-1β maturation, gasdermin-D cleavage, and pyroptosis in response to S. typhimurium infection. RNA sequencing and RT-PCR results revealed that the transcription of Naips was decreased in Brd4-deficient BMDMs. Brd4 formed a complex with IRF8/PU.1 and bound to the IRF8 and PU.1 binding motifs on the promoters of Naips to maintain the expression of Naips. Furthermore, myeloid lineage–specific Brd4 conditional knockout mice were more susceptible to S. typhimurium infection with increased mortality, bacterial loads, and tissue damage; impaired inflammasome-dependent cytokine production; and pyroptosis. Our studies identify a novel function of Brd4 in innate immunity by controlling inflammasome-mediated cytokine release and pyroptosis to effectively battle S. typhimurium infection.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1335-1335
Author(s):  
Christian P. Pallasch ◽  
Leskov Ilya ◽  
Yadira M Soto ◽  
Christian J. Braun ◽  
Nadine Kutsch ◽  
...  

Abstract Abstract 1335 Therapy-resistant microenvironments represent a major barrier to the effective elimination of disseminated malignancies. However, microenvironment dependent resistance mechanisms as potential synergistic drug interactions particularly for biologicals and monoclonal therapeutic antibodies are not completely understood. Here, we used the hMB humanized lymphoma mouse model as primary human B-ALL xenograft mice to address mechanisms of resistance and potential synergy of the clinical grade antibodies alemtuzumab and rituximab. Response to antibody therapy was shown to be mediated by macrophages as effector cells by direct phagocytosis - indicated by an abrogated response in macrophage-depleted mice. Antibody mono-therapy however still showed limited response in the bone marrow as a site of a primarily resistant microenvironment. In order to overcome therapeutic resistance and generate a functional state of the tumor microenvironment allowing effective antibody-mediated phagocytosis of lymphoma cells we applied a series of combinatorial regimens. Supplementing treatment with GM-CSF in order to improve the effector to target ratio slightly enhanced the therapeutic response as only modest additive affects were seen with total body irradiation. By combining alemtuzumab and cyclophosphamide in the hMB model as rituximab and cyclophosphamide respectively in CD20+ B-ALL xenografts we identified a striking synergy leading to profound depletion of malignant cells from bone marrow and spleen. Mice treated in the combinatorial arm survived significantly longer (Median survival 7 weeks vs. 28 weeks, p<0.001). Notably, synergy was exclusively demonstrated in cyclophosphamide while other alkylating agents, topoisomerase inhibitors, spindle poisons or steroids did not elicit a strong therapeutic synergy. Cyclophosphamide treatment induced an increased frequency of F4/80+ macrophages in the primarily resistant bone marrow in and multiphoton confocal microscopy of leukemia infiltrated and treated tumor tissue revealed enhanced phagocytic activity. Analyzing the underlying mechanisms of cyclophosphamide – antibody synergy using conditioned media from cyclophosphamide pretreated leukemia cells we identified an acute secretory response significantly enhancing macrophage mediated leukemia cell depletion upon antibody treatment in vivo. Analyzing a panel of human cytokines we could identify VEGF, CLL4, TNFα, and IL8 to be specifically induced by cyclophosphamide while not being induced by total body irradiation. Using recombinant cytokines or blocking antibodies in conditioned media we could recapitulate the significant influence of these cytokines on enhancing macrophage-dependent leukemia cell removal in vitro. Since the cyclophosphamide-induced acute secretory activating phenotype (ASAP) shows a rapid transient cytokine release synergistic chemo-immunotherapy of antibody and cyclophosphamide is limited to a 24h time-frame of simultaneous co-dosing to provide synergy. Here we could identify secretory phenotypes of malignant cells determining therapeutic outcome in antibody-based therapies. These findings underline the importance of tumor-microenvironment interactions for therapeutic outcome. Thus, the acute induction of stress-related cytokines represents a highly effective strategy to target cancer cells for targeted removal by the innate immune system. Here we identified so far unraveled mechanisms of synergy in chemo-immunotherapy and will thereby improve future design of clinical treatment regimens. Disclosures: No relevant conflicts of interest to declare.


2005 ◽  
Vol 158 (1-2) ◽  
pp. 50-57 ◽  
Author(s):  
Kuzhali Muthu ◽  
Jiangping Deng ◽  
Richard Gamelli ◽  
Ravi Shankar ◽  
Stephen B. Jones

Blood ◽  
1995 ◽  
Vol 86 (3) ◽  
pp. 890-899 ◽  
Author(s):  
E Holler ◽  
HJ Kolb ◽  
J Mittermuller ◽  
M Kaul ◽  
G Ledderose ◽  
...  

Abstract Contribution of host-related cytokine release in the course of pretransplant conditioning to early tissue damage and induction of acute graft-versus-host disease (GVHD) after allogeneic bone marrow transplantation (BMT) has been shown in experimental models. We performed a clinical phase I/II trial applying a monoclonal antibody neutralizing human tumor necrosis alpha (TNF alpha) during pretransplant conditioning as additional prophylaxis in high-risk patients admitted to allogeneic BMT; TNF alpha serum levels and clinical courses in 21 patients receiving anti-TNF alpha prophylaxis were compared with data from 22 historical controls. Absence of significant release of TNF alpha in the period of busulphan (BUS) treatment, but significant induction of TNF alpha by total body irradiation (TBI) and cyclophosphamide (CY) conditioning were correlated with significantly earlier onset of acute GVHD in patients receiving TBI/CY regimens as compared with BUS/CY-treated patients. Prophylactic application of monoclonal anti-TNF alpha seemed to postpone onset of acute GVHD from day 15 to day 25 (P < .05) after TBI/CY and from day 33 to day 53 after BUS/CY (P < .10) conditioning. Application of monoclonal anti-TNF alpha in low and intermediate doses was safe and not associated with an increased incidence of infectious or hematologic complications. Thus, our data provide indirect and direct evidence for involvement of conditioning-related cytokine release in induction of early acute GVHD in the clinical setting and support further investigation of this novel approach in randomized trials.


1994 ◽  
Vol 424 (4) ◽  
Author(s):  
D. Rohde ◽  
C. Wickenhauser ◽  
S. Denecke ◽  
A. Stach ◽  
J. Lorenzen ◽  
...  

2001 ◽  
Vol 19 (1) ◽  
pp. 61-71 ◽  
Author(s):  
Gordana Jovči ◽  
Diana Bugarski ◽  
Marijana Petakov ◽  
Jelena Stanković ◽  
Nevenka Stojanović ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document