The Influence of Anesthetic Agents on the Immune Response: A Crucial Factor in Immune Suppression?

Author(s):  
P. Schmucker ◽  
C. Hammer
1983 ◽  
Vol 43 (1) ◽  
pp. 43-54 ◽  
Author(s):  
A. A. Broek ◽  
F. J. Keuning ◽  
R. Soeharto ◽  
N. Prop

1990 ◽  
Vol 72 (3) ◽  
pp. 542-552 ◽  
Author(s):  
G. W. Stevenson ◽  
Steven C. Hall ◽  
Steven Rudnick ◽  
Frank L. Seleny ◽  
Henry C. Stevenson

Author(s):  
Mark W Hall ◽  
Ila Joshi ◽  
Luis Leal ◽  
Eng Eong Ooi

Abstract We are learning that the host response to severe acute respiratory syndrome coronavirus 2 ( SARS-CoV-2) infection is complex and highly dynamic. Effective initial host defense in the lung is associated with mild symptoms and disease resolution. Viral evasion of the immune response can lead to refractory alveolar damage, ineffective lung repair mechanisms, and systemic inflammation with associated organ dysfunction. The immune response in these patients is highly variable and can include moderate to severe systemic inflammation and/or marked systemic immune suppression. There is unlikely to be a “one size fits all” approach to immunomodulation in patients with coronavirus disease 2019 (COVID-19). We believe that a personalized, immunophenotype-driven approach to immunomodulation that may include anticytokine therapy in carefully selected patients and immunostimulatory therapies in others is the shortest path to success in the study and treatment of patients with critical illness due to COVID-19.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 128-128
Author(s):  
Gullu Gorgun ◽  
Teru Hideshima ◽  
Noopur S. Raje ◽  
Naoya Mimura ◽  
James E. Bradner ◽  
...  

Abstract Abstract 128 The interaction of myeloma (MM) cells with bone marrow accessory cells and/or the extracellular matrix induces genomic, epigenomic and functional changes which promote tumor development, progression, cell adhesion mediated-drug resistance (CAM-DR), and immune suppression. To develop the most efficient anti-MM treatment strategy and prevent tumor escape from immune recognition, both enhancing anti-MM effector immune response and overcoming MM-induced immune suppression is essential. Suppressive immune cells including myeloid derived suppressor cells (MDSC), regulatory T cells (Treg) and IL-17 secreting Th (Th17) cells act as tumor promoters and suppressors of effector immune response, and therefore represent a significant barrier to current anti-tumor therapeutic strategies. Since, we and others have reported increased numbers of Treg and Th17 cells in MM, we here assessed MDSCs in both peripheral blood (PBMC) and bone marrow (BMMC) of patients with MM compared to healthy donors. Phenotypic analysis by flow cytometry showed a significant increase in CD14−CD11b+HLA-DRlowCD15+ MDSCs in both PBMC and BMMC from MM patients compared to healthy donors (p<0.01). Furthermore, coculture of MM cell lines with healthy PBMCs for 6 days demonstrated that MM cells significantly induce MDSC differentiation in healthy PBMCs (p<0.03). Recent studies have demonstrated that histone deacytlase 6 (HDAC6) is an important regulator of monocyte/macrophage-mediated immune response. We therefore next analysed the immunomodulatory effects of WT-161, a novel small molecule inhibitor of HDAC6, alone or in combination with lenalidomide (len) and bortezomib (bort), on suppressive immune cells in the MMBM microenvironment. To keep cell-cell interaction intact reflective of the MMBM microenvironment, PBMCs or BMMCs from MM patients were cultured in the absence or presence of WT-161 (0.5–5uM), len (1–10uM), and/or bort (2–5nM), and individual cell populations were analysed by flow cytometry. Phenotypic characterization of suppressive immune cells showed a significant decrease in both CD4+CD25+Foxp3+ Treg cells and MDSCs in MM-PBMCs and MM-BMMCs cultured with WT-161, alone or in combination with len or bort (p<0.01); however, there was no change in the expression of Th17 cells. To determine the functional mechanism of immune suppression, MDSC and Treg cells were isolated by magnetic-Ab sorting and cultured for 6 days with autologous T cells (TCR/IL-2 stimulated), with or without WT-161, len and bort, alone or in combination. T cell proliferation (by 3H-thymidine assay) was significantly inhibited in the presence of MDSCs, whereas WT-161 notably reversed MDSC-mediated T cell suppression. In contrast, len and bort did not show any significant effect. Intracellular reactive oxygen species (ROS, an MDSC-derived metabolic immune inhibitory molecule) expression was significantly decreased in MDSCs from MM cultured with WT-161, alone or together with len and bort (p<0.05). Additionally, WT-161 also reversed Treg-mediated T cell suppression as well as len. Cytokine profiling by intracellular flow cytometric analysis demonstrated that WT-161 significantly decreased IL-6 and GM-CSFR expression in MDSCs, whereas it induced IFNγ and IL-12 production in effector CD4T, CD8T and NKT cells. Finally, unstimulated or IL-2 prestimulated (36h) PBMCs or NK cells were cultured with MM cell lines (MM1.S, RPMI8226), in the absence or presence of WT-161 alone or with len and bort (4h), and anti-MM cytotoxic activity was determined by Cr51-release cytotoxicity assay. While len (48% killing) and WT-161 (39% killing) induced CTL-mediated cytotoxicity, WT-161 (53% killing) and len (56% killing) induced more potent NK cell-mediated anti-MM cytotoxicity. These data suggest that HDAC6 may have an immune regulatory function, and that inhibition of HDAC6 induces changes in suppressor immune cells leading to enhanced anti-MM immune response in MM microenvironment. Ongoing analysis of the effects of HDAC6 inhibition on immune cells in the tumor microenvironment will further define the role of HDAC6 in disease pathogenesis and suggest novel immune-based epigenetic-targeted therapies. Disclosures: Hideshima: Acetylon: Consultancy. Raje:Celgene: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees; Millenium: Membership on an entity's Board of Directors or advisory committees; Astra Zeneca: Research Funding; Amgen: Membership on an entity's Board of Directors or advisory committees; Acetylon: Research Funding. Bradner:Acetylon: Scientific Founder. Richardson:Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees; Johnson & Johnson: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Millennium: Membership on an entity's Board of Directors or advisory committees. Munshi:Celgene: Membership on an entity's Board of Directors or advisory committees; Millennium: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees; Onyx: Consultancy, Membership on an entity's Board of Directors or advisory committees. Anderson:Novartis: Membership on an entity's Board of Directors or advisory committees; Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees; Merck: Membership on an entity's Board of Directors or advisory committees; Onyx: Membership on an entity's Board of Directors or advisory committees; Millennium: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Acetylon: Membership on an entity's Board of Directors or advisory committees.


1970 ◽  
Vol 131 (2) ◽  
pp. 247-274 ◽  
Author(s):  
Marc Feldmann ◽  
Erwin Diener

Antibody-mediated suppression of the in vitro immune response to polymerized flagellin of Salmonella adelaide and to sheep erythrocytes was studied at the cellular level. Normal mouse spleen cells, preincubated in vitro with mixtures of antigen and antibody for short periods of time before being washed, did not respond to an optimal antigenic challenge in vitro, whereas similar cells treated with antibody alone gave a normal response. The degree of immune suppression was found to depend on the time of preincubation. Significant immune suppression could be induced in as short a time as 15 min, whereas profound suppression (90%) required the incubation of cells with mixtures of antigen and antibody for 4–6 hr. Mouse spleen cells treated similarly were also unable to respond subsequently to the antigen upon transfer to lethally irradiated hosts, as measured at both the level of the antigen-reactive cell and that of serum antibody production. These results were taken as evidence that in vitro an effect of antibody-mediated suppression occurred at the level of the immunocompetent cell. Similarities between immune tolerance and antibody-mediated suppression in vitro were described, and the significance of the findings discussed in the light of current concepts of the mechanism of antibody-mediated suppression.


Author(s):  
Srdjan Gavrilovic ◽  
Ana Andrijevic ◽  
Aida Mujakovic ◽  
Yewande Odeyemi ◽  
Belma Paralija ◽  
...  

Pneumonia is the leading infectious cause of death worldwide. While inflammation is critically important in host response to microbial invasion, exaggerated inflammation can damage the lungs, contributing to respiratory failure and mortality. Corticosteroids are effective in reducing inflammation and can also cause immune suppression. Presently, clinicians are unable to reliably distinguish between exaggerated and appropriate immune response and thus cannot rapidly identify patients most likely to benefit from adjunctive corticosteroids. In this review, we propose a biomarker-guided, precision medicine approach to corticosteroid treatment, aimed to give these medications at appropriate dose and time and only to patients who have exaggerated inflammation.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 708-708
Author(s):  
Cheryl Maier ◽  
Amanda Mener ◽  
Seema R Patel ◽  
Ashley L Bennett ◽  
Sean R Stowell

Abstract Background: Exposure to red blood cell (RBC) alloantigens during pregnancy or transfusion can lead to the development of alloantibodies and result in transfusion-related complications, like hemolytic transfusion reactions, or hemolytic disease of the fetus and newborn. Currently, antibody-mediated immune suppression (AMIS) is the only known methodology to actively inhibit RBC alloantibody formation. The best-known example of AMIS is Rh immune globulin, which is given to Rh(D) negative women to prevent the development of anti-D alloantibodies. Mouse models to elucidate the mechanism by which AMIS occurs have used mice genetically engineered to specifically express the human KEL antigen (KEL RBCs) or the Hel-Ova-Duffy antigen (HOD RBCs) on murine RBCs, and found that AMIS to these two antigens is dependent on antigen loss (also called antigen modulation). As RBCs express many antigens simultaneously, we sought to determine whether AMIS to one antigen can impact the immune response to a completely different antigen on the same RBC by generating mice that express both the KEL and HOD antigens (HOD x KEL RBCs). Methods: WT recipients received polyclonal anti-KEL antibody, a cocktail of two anti-HEL monoclonal antibodies, or PBS, followed by transfusion with WT and HOD x KEL donor RBCs. HOD x KEL RBCs were labeled with the lipophilic dye DiI prior to transfusion, while WT RBCs were labeled with the lipophilic dye DiO and served as a RBC tracer population. RBC survival and levels of detectable IgG, Kel antigen, Hel antigen, Duffy antigen, and complement (C3) on transfused cells were measured by flow cytometry at 10min, 1hr, 2hr, and 4hr, and on days 1, 2, 3, and 5 post-transfusion. Serum was collected on day 5 and analyzed for IgM antibody development by flow-crossmatch using KEL or HOD RBCs. Results: Anti-KEL and anti-HEL treated WT recipients demonstrated significant IgG deposition on transfused HOD x KEL RBCs at 10min post-transfusion. Levels of IgG on transfused RBCs decreased over time but did not correlate with complete RBC clearance. RBC survival was nearly 100% over 5 days in recipients passively immunized with anti-HEL antibodies, while a fraction of RBCs (~20%) were cleared within 4hr post-transfusion but not thereafter (survival plateaued at 80% thru day 5) in anti-KEL immunized recipients. Conversely, decreased bound IgG on transfused RBCs was found to correlate with a decrease in target antigen levels. KEL antigen levels decreased on transfused RBCs in the anti-KEL immunized recipients beginning at 1hr post transfusion, as compared to anti-HEL and PBS recipients. Similarly, HEL antigen expression decreased on transfused RBCs in the anti-HEL immunized recipients, but not in the anti-KEL or PBS recipients, demonstrating that passive immunization causes antigen modulation for the corresponding antigen only. C3 deposition was found to occur on transfused RBCs at 10min post-transfusion in the anti-KEL recipients that remained significantly increased at other time-points, but was not detected on RBCs in the anti-HEL or PBS recipients until day 3, when recipients develop IgM antibodies. Flow crossmatch of serum samples showed that anti-KEL treated recipients did not make anti-KEL IgM but did make anti-HOD IgM, while anti-HEL treated recipients did not make anti-HOD IgM but did make anti-KEL IgM. Conclusions: Although AMIS serves as the basis of a widely successful clinical intervention, the mechanism by which it occurs, including any impact on additional antigens, remains incompletely understood. Here we demonstrate that AMIS is antigen specific - that is, passively acquired antibody can inhibit the immune response only to the corresponding antigen. Specifically, murine RBCs expressing both the human and model RBC antigen KEL and HOD, respectively, demonstrate similar clearance kinetics, antigen modulation, complement deposition, and antibody development after transfusion into WT mice as is observed when KEL-only or HOD-only RBCs are used. Although it is unclear whether these findings apply to all RBC antigens and what relevance this holds for human patients, our study provides insight into the specificity of AMIS that may enlighten development of additional therapeutics based on this mode of immune suppression. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Author(s):  
Weiling He ◽  
Hui Zhang ◽  
Shuhua Li ◽  
Yongmei Cui ◽  
Ying Zhu ◽  
...  

AbstractLung adenocarcinoma (LUAD) remains one of the leading causes of death in patients with cancer. The association of CD155 with CD96 transmits an inhibitory signal and suppresses antitumor immune response. This study investigates the effect of CD155/CD96 on immune suppression in LUAD. We demonstrate that LUAD patients with high CD155 expression suffer from immune suppression and experience a poor prognosis, which coincides with an inhibited AKT-mTOR signaling pathway in CD8 T cells and subsequently up-regulated CD96 expression. Moreover, the inhibition effect can be reversed by CD96 blocking antibody. High CD155 expression inhibited the release of IFNγ from CD8 cells. Moreover, Blocking CD96 restored IFNγ production in CD8 T cells and neutralized the inhibition of IFNγ production in CD8 T cells mediated by CD155. Animal experiments showed that CD155-mediated LUAD growth might depend on its suppression antitumor immune response in the tumor microenvironment in PDX mice. In conclusion, our results suggest that LUAD cells suppress antitumor immune response in the tumor microenvironment through CD155/CD96. CD155/CD96 could be a potential therapeutic target for LUAD patients.AbbreviationsLUAD: lung adenocarcinoma; IFNγ: interferon gamma; PDX: patient-derived xenograft; NSCLC: non-small cell lung cancer; PRR: poliovirus receptor–related; MDSCs: myeloid-derived suppressor cells; PRR: poliovirus receptor–related; STR: short tandem repeat; IRS: immunoreactive score; SI: staining intensity; PP: percentage of positive cells; RT-PCR: reverse transcription-polymerase chain reaction; PBS: phosphate-buffered saline; PBMCs: peripheral blood mononuclear cells; SDS–PAGE: sodium dodecyl sulfate-polyacrylamide gel electrophoresis; rCD155: recombinant human CD155; LUAD cells: lung adenocarcinoma cells; TILs: tumor-infiltrating lymphocytes; GzmB: granzyme B; IL-2 (Interleukin-2); TNF-α : tumor necrosis factor-alpha; PI: propidium Iodide; PDX: patient-derived xenograft; TIGIT: T cell immunoreceptor with Igand ITIM domains; WBC: white blood cells; MFI: mean fluorescence intensity; HPF: high power field


Sign in / Sign up

Export Citation Format

Share Document