scholarly journals Lentinan Inhibits Tumor Progression by Immunomodulation in a Mouse Model of Bladder Cancer

2020 ◽  
Vol 19 ◽  
pp. 153473542094682
Author(s):  
Ming Sun ◽  
Renge Bu ◽  
Bin Zhang ◽  
Yaming Cao ◽  
Chengyang Liu ◽  
...  

Background: Lentinan (LNT), an isolated traditional Chinese herbal component, has antitumor potential. In the current study, the intrinsic mechanism of LNT-induced immunity against bladder cancer was explored in a mouse model. Methods: In the mouse model of bladder cancer, we used flow cytometry to detect the LNT caused population changes of T cells, macrophages, MDSC cells, and Treg cells. ELISA was used to evaluate cytokines expression in the supernatant of splenocytes. Results: We found that the administration of LNT increased the proportions of CD3+CD4+ and CD3+CD8+ T cell subsets as well as CD11b+F480+ macrophages, whereas it diminished the subpopulations of CD4+CD25+Foxp3+ regulatory T cells (Tregs) and Gr-1+CD11b+ myeloid-derived suppressor cells (MDSCs). LNT also upregulated the expression of interferon (IFN)-γ and interleukin (IL)-12, accompanied by a significant reduction in IL-10 and tumor growth factor (TGF)-β ( P < .05). Our research further confirmed the synergy between LNT and gemcitabine (GEM) to activate immunity and inhibit the growth of bladder tumors in mouse model. Conclusions: LNT induced macrophage activation, followed by the enhanced proliferation of CD4+ and CD8+ T cells, and the upregulated expression of IFN-γ and IL-2. Meanwhile, the proportions of MDSCs and Tregs were downregulated, leading to a reduced expression of the anti-inflammatory cytokines IL-10 and TGF-β. The synergy between LNT and GEM provides additional evidence supporting the application of this traditional Chinese herbal component for bladder cancer therapy.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3511-3511
Author(s):  
Sylvia Feyler ◽  
Marie von Lilienfeld-Toal ◽  
Lee Marles ◽  
Andy Rawstron ◽  
Roger G. Owen ◽  
...  

Abstract The immunologically hostile microenvironment of MM contributes to the limited success of immunotherapy strategies. In addition to direct tumour-induced immunosuppression, tumour cells may generate suppressor cells such as Treg cells which can profoundly suppress immune responses and induce tolerance. This study aimed to determine if Treg subsets are increased in the peripheral blood (PB) and bone marrow (BM) of patients with MGUS/MM and how this correlates with increasing disease burden. PB samples from 166 patients with MGUS/MM (Newly diagnosed (ND), n=34; Plateau/low disease burden (LD), n=63; Relapsed/refractory (R/R), n=27 and MGUS, n=42) with a median age of 69 years (range 39–89 yrs) were analysed by FACS and compared to PB from 32 age/sex matched controls. Using a sequential gating strategy, naturally occurring Treg cells (nTregs) were identified as CD4+/CD25+/FoxP3+ T-cells and expressed as a percentage of the CD4+ T-cells. Double negative T cells (DN Tregs) were identified as CD3+/CD4−/CD8−/alphabeta-TCR+/gammadelta−TCR− and expressed as a percentage of the CD3+ cells. Sera were analysed by ELISA for IL10 and TGF-beta. nTReg cells were significantly increased in patients with MGUS/MM compared with controls (Controls 1.6% ± 0.2, MGUS 2.3% ± 0.3, ND 2.4% ± 0.2, LD 4.2% ± 0.7 & R/R 3.8% ± 0.5; p=0.003). There was a positive correlation of nTRegs number with paraprotein level (R=0.3, p=0.005). Patients on Thalidomide at the time of sample collection had significantly higher numbers of nTRegs of 6% in comparison to patients who never received Thalidomide (3%) and patients who previously received the drug (3.5%, p=0.005). Analysis of BM (n=12) demonstrated a significantly reduced number of nTRegs in comparison to PB (1.6% vs 3.0%, p=0.025), which is higher than the number of nTRegs in the BM of controls (0.7%). Functionally, nTRegs from patients demonstrated suppressive activity of both autologous and allogeneic T-cells similar to nTRegs cells from control PB (p=NS). In contrast, DN TRegs were significantly reduced in patients with myeloma (ND 1.0% ± 0.2, LD 1.1% ± 0.1, R/R 1.3% ± 0.2) compared with MGUS and controls (2.1% ± 0.9 & 3.3% ± 0.7, respectively; p=0.02). Serum IL10 levels were significantly lower in ND (17 ± 38 pg/ml) and LD (29 ± 36 pg/ml) than in Controls (55 ± 89 pg/ml), MGUS (72 ± 151 pg/ml) and R/R (90 ±182 pg/ml, p=0.02). TGF-b levels differed significantly between groups (Controls 3765 ± 1593 pg/ml, MGUS 3506 ± 1504 pg/ml, ND 4774 ± 9879 pg/ml, LD 1915 ± 1324 pg/ml, R/R 2802 ± 2011 pg/ml, p=0.025). These results provide further evidence of immune dysregulation in MM. The association with advanced disease stage suggests a causal association.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 11577-11577 ◽  
Author(s):  
Jooeun Bae ◽  
Brandon Nguyen ◽  
Yu-Tzu Tai ◽  
Teru Hideshima ◽  
Dharminder Chauhan ◽  
...  

11577 Background: Characterization of expression and function of immune regulatory molecules in tumor microenvironment will provide the framework for developing novel therapeutic strategies. Methods: We evaluated the expression and functional impact of various immuno-regulatory molecules, PD-1, PDL-1, PDL-2, LAG3, TIM3, OX40 and GITR, on the CD138+ tumor cells, myeloid derived suppressor cells (MDSC), and T cell subsets from patients with MGUS, SMM and active MM (newly diagnosed, relapsed, relapsed/refractory), and the myeloma-specific cytotoxic T lymphocytes (CTL) induced with XBP1/CD138/CS1 peptides. Results: PDL-1/PDL-2 was more highly expressed on CD138+ myeloma cells in active MM than SMM or MGUS. G-type MDSC (CD11b+CD33+HLA-DRlowCD15+). Treg cells (CD3+CD4+/CD25+FOXP3+) numbers were increased and expressed higher levels of PD1/PD-L1 in active MM than in MGUS, SMM or healthy donors. Among the checkpoint molecules (PD-1, PDL-1, PDL-2, LAG3, OX40, GITR) evaluated, PD-1 showed the highest expression on CD3+CD4+ and CD3+CD8+T cells in BMMC and PBMC from patients with active MM. Functionally, T cells from MM patients showed increased proliferation upon treatment with an individual immune agonist ( > 150%) or checkpoint inhibitor ( > 100%). Interestingly, each individual anti-checkpoint molecule induced proliferation of T cells expressing other checkpoint molecules. In addition, the blockade of PD1, LAG3 or TIM3 enhanced MM antigen-specific cytotoxicity, assessed by parameters including CD107a, granzyme B and IFN-g production, which was most prominent within the memory CTL subset of MM antigen-specific T cells. Conclusions: These results demonstrate an increased frequency of immune regulatory cells, which highly express checkpoint inhibitors in active MM. Direct stimulation with an immune agonist or blockade of a checkpoint inhibitor increased MM patients’ T cell proliferation and myeloma-specific CTL function, supporting development of combination immune regulatory therapies to improve patient outcome in MM.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4396-4396
Author(s):  
Zenghua Lin ◽  
Hong Liu ◽  
Han Wang

Abstract Abstract 4396 Aplastic anemia (AA) is defined as pancytopenia with a hypocellular marrow, and interferon-gamma (IFN-γ) plays a key role in the injury to stem and progenitor compartments in AA. In recent years, animal models of AA have helped us to strengthen understanding of the mechanisms causing AA. However, few well-characterized mouse models have been developed to study the pathogenesis of AA. We have first developed a novel mouse model of severe aplastic anemia (SAA) induced by administration with IFN-γ and busulphan. In this study, 60 clean-class 8-week-old BALB/c female mice were randomly selected to set up the SAA model using intraperitoneal injection with IFN-γ and intragastric administration with busulphan for 10 days (SAA group), the IFN-γ group (n=60), the busulphan group (n=60) and the un-treated group (n=60) were as control. A mortality of 20% was found in the SAA group at day 10 after treatment and increased to 100% at day 30 after treatment. All mice in SAA group developed the typical clinical and pathological patterns of SAA from day 10 post dosing. They presented bleedings in association with anemia and infections. The peripheral blood smear shows paucity of leukocytes, platelets and absolute reticulocytes with decreased hemoglobin concentration, lower compared with other groups (P<0.05, respectively). The same result also been found in spleen index and nucleated cells per tibia. The bone marrow smears and the patho-morphological examinations showed hypocellularity in marrow cell proliferation, while an increase in the number of fat cells, residual lymphocytosis, plasma cells, stromal elements. The depression was severe and irreversible with time. Furthermore, in order to confirm the immunological features of the novel mouse SAA model, the mononuclear cells of the peripheral blood and spleen were subjected to assess the percentage of Th1□ATh2□ATh17 and regulatory T cells in CD4+ T cell subsets by flow cytometry (FCM). In the SAA group, the FCM analysis showed increased frequencies of Th1 cells, Th17 cells, and ratio of Th1/Th2 with decreased frequencies of CD4+CD25+ regulatory T cells (Tregs) and CD4+CD25+FoxP3+ Tregs in peripheral blood and spleen (P<0.05, respectively). To further explore the mechanism of decreased frequencies of Tregs in SAA, after being pured by immunomagnetic beads, the splenic Tregs was subjected to assess the expression of Akt and transforming growth factor-β (TGF-β) using Western blot and the apoptosis of splenic Tregs was detected by FCM. Results showed that a greater proportion of apoptotic cells in splenic Tregs with down-expression of Akt and TGF-β was found in SAA group (P<0.05, respectively). Obviously, this improved mouse model of SAA induced by IFN-γ and busulphan closely duplicates human SAA in a short time and our findings may contribute to understanding the decreased number of Tregs characteristic of AA. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4105-4105
Author(s):  
Xiaochuan Chen ◽  
Chien-Hsing Chang ◽  
Rhona Stein ◽  
Thomas M Cardillo ◽  
David V. Gold ◽  
...  

Abstract Abstract 4105 Introduction: Graft-versus-host disease (GVHD) is a life-threatening complication of allogeneic hematopoietic stem cell transplantation. Prevention and treatment of GVHD remains a major challenge, because current T-cell depletion and mainstay immunosuppressive therapies compromise preexisting T-cell immunity, leading to severe infections and disease relapse. Thus, novel anti-GVHD agents that can spare protective T-cell memory are critically needed. Milatuzumab (hLL1) is a humanized anti-CD74 antagonist IgG1κ monoclonal antibody (mAb) currently under clinical investigation as a therapeutic mAb for relapsed or refractory B-cell malignancies. Since CD74 is widely expressed in both hematopoietic and non-hematopoietic antigen-presenting cells (APCs), and because host APCs, especially non-hematopoietic APCs, play an important role in initiating GVHD, whereas donor APCs contribute and are required to maximize GVHD, we reasoned milatuzumab could affect recipient or donor APCs, thereby modulating GVHD. We report herein the in vitro effect of milatuzumab on the survival and function of human blood APCs and T cells, including CMV-specific T cells, and the in vivo therapeutic efficacy on preventing acute GVHD in a humanized SCID mouse model. Methods: The effects of milatuzumab on APCs and T cells in human peripheral blood mononuclear cells (PBMCs) were assessed by multi-color flow cytometry. The effect of milatuzumab on the proliferation of T cells and T cell subsets in allogeneic mixed lymphocyte reactions (allo-MLRs) was measured by CFSE labeling of allogeneic PBMCs that were mixed to each other and cultured for 11 days before flow cytometric analysis of the proliferating cells that lost fluorescence. The impact of milatuzumab on preexisting anti-viral T-cell immunity was evaluated by intracellular staining of CD3+CD8+IFN-γ+T cells in allo-MLRs upon stimulation with HLA-A2-restricted cytomegalovirus (CMV) pp65 peptide (NLVPMVATV). The therapeutic effect of milatuzumab on acute GVHD was evaluated in a human PBMC-transplanted SCID mouse model, in which GVHD is developed and mediated by engrafted human T cells and DCs. Results: Milatuzumab moderately reduces the number of myeloid DC type 1 (mDC1), myeloid DC type 2 (mDC2), and B cells in PBMCs, but has little effect on plasmacytoid DC (pDC), monocytes, or T cells, which correlates with the level of CD74 expression on these cells. As a consequence, milatuzumab inhibits the proliferation of total T cells, CD4 and CD8 T cell subsets in allo-MLRs. In a human/mouse xenogeneic SCID mouse model, milatuzumab effectively prevents the onset and manifestations of acute GVHD, suppresses the serum levels of human interferon-g and IL-5, eliminates the infiltration of human lymphocytes in GVHD target organs (lung, liver and spleen), and significantly promotes the survival of animals (90% versus 20% for controls, P=0.0012). Furthermore, exposure to milatuzumab does not affect the number of CMV-specific, IFN-γ-producing, human CD8+ T cells in allo-MLRs. Conclusion: CD74 is a potential target for antibody-mediated mitigation of GVHD, as demonstrated by the encouraging results obtained with milatuzumab. Further exploration of milatuzumab as a new therapeutic agent for GVHD is warranted. Disclosures: Cardillo: Immunomedics, Inc: Employment. Goldenberg:Immunomedics: Employment, Equity Ownership.


Cancers ◽  
2021 ◽  
Vol 13 (10) ◽  
pp. 2428
Author(s):  
Frank Liang ◽  
Azar Rezapour ◽  
Peter Falk ◽  
Eva Angenete ◽  
Ulf Yrlid

TILs comprise functionally distinct conventional and unconventional T cell subsets and their role in responses to CRC treatments is poorly understood. We explored recovery of viable TILs from cryopreserved tumor biopsies of (chemo)-radiated patients with rectal cancer to establish a platform for retrospective TIL analyses of frozen tumors from pre-selected study cohorts. Frequencies of TIL subsets and their capacity to mount IFN-γ responses in cell suspensions of fresh vs. cryopreserved portions of the same tumor biopsies were determined for platform validation. The percentages and proportions of CD4+ TILs and CD8+ cytotoxic T lymphocytes (CTLs) among total TILs were not affected by cryopreservation. While recovery of unconventional γδ T cells and mucosal-associated invariant T cells (MAIT cells) was stable after cryopreservation, the regulatory T cells (Tregs) were reduced, but in sufficient yields for quantification. IFN-γ production by in vitro-stimulated CD4+ TILs, CTLs, γδ T cells, and MAIT cells were proportionally similar in fresh and cryopreserved tumor portions, albeit the latter displayed lower levels. Thus, the proposed platform intended for TIL analyses on cryopreserved tumor biobank biopsies holds promises for studies linking the quantity and quality of TIL subsets with specific clinical outcome after CRC treatment.


2021 ◽  
Vol 9 (4) ◽  
pp. e002051
Author(s):  
Ryan Michael Reyes ◽  
Yilun Deng ◽  
Deyi Zhang ◽  
Niannian Ji ◽  
Neelam Mukherjee ◽  
...  

BackgroundAnti-programmed death-ligand 1 (αPD-L1) immunotherapy is approved to treat bladder cancer (BC) but is effective in <30% of patients. Interleukin (IL)-2/αIL-2 complexes (IL-2c) that preferentially target IL-2 receptor β (CD122) augment CD8+ antitumor T cells known to improve αPD-L1 efficacy. We hypothesized that the tumor microenvironment, including local immune cells in primary versus metastatic BC, differentially affects immunotherapy responses and that IL-2c effects could differ from, and thus complement αPD-L1.MethodsWe studied mechanisms of IL-2c and αPD-L1 efficacy using PD-L1+ mouse BC cell lines MB49 and MBT-2 in orthotopic (bladder) and metastatic (lung) sites.ResultsIL-2c reduced orthotopic tumor burden and extended survival in MB49 and MBT-2 BC models, similar to αPD-L1. Using antibody-mediated cell depletions and genetically T cell-deficient mice, we unexpectedly found that CD8+ T cells were not necessary for IL-2c efficacy against tumors in bladder, whereas γδ T cells, not reported to contribute to αPD-L1 efficacy, were indispensable for IL-2c efficacy there. αPD-L1 responsiveness in bladder required conventional T cells as expected, but not γδ T cells, altogether defining distinct mechanisms for IL-2c and αPD-L1 efficacy. γδ T cells did not improve IL-2c treatment of subcutaneously challenged BC or orthotopic (peritoneal) ovarian cancer, consistent with tissue-specific and/or tumor-specific γδ T cell contributions to IL-2c efficacy. IL-2c significantly altered bladder intratumoral γδ T cell content, activation status, and specific γδ T cell subsets with antitumor or protumor effector functions. Neither IL-2c nor αPD-L1 alone treated lung metastatic MB49 or MBT-2 BC, but their combination improved survival in both models. Combination treatment efficacy in lungs required CD8+ T cells but not γδ T cells.ConclusionsMechanistic insights into differential IL-2c and αPD-L1 treatment and tissue-dependent effects could help develop rational combination treatment strategies to improve treatment efficacy in distinct cancers. These studies also provide insights into γδ T cell contributions to immunotherapy in bladder and engagement of adaptive immunity by IL-2c plus αPD-L1 to treat refractory lung metastases.


2014 ◽  
Vol 307 (2) ◽  
pp. G177-G186 ◽  
Author(s):  
Yuying Liu ◽  
Dat Q. Tran ◽  
Nicole Y. Fatheree ◽  
J. Marc Rhoads

Necrotizing enterocolitis (NEC) is an inflammatory disease with evidence of increased production of proinflammatory cytokines in the intestinal mucosa. Lactobacillus reuteri DSM 17938 (LR17938) has been shown to have anti-inflammatory activities in an experimental model of NEC. Activated effector lymphocyte recruitment to sites of inflammation requires the sequential engagement of adhesion molecules such as CD44. The phenotype of CD44+CD45RBlo separates T effector/memory (Tem) cells from naive (CD44−CD45RBhi) cells. It is unknown whether these Tem cells participate in the inflammation associated with NEC and can be altered by LR17938. NEC was induced in 8- to 10-day-old C57BL/6J mice by gavage feeding with formula and exposure to hypoxia and cold stress for 4 days. Survival curves and histological scores were analyzed. Lymphocytes isolated from mesenteric lymph nodes and ileum were labeled for CD4, CD44, CD45RB, intracellular Foxp3, and Helios and subsequently analyzed by flow cytometry. LR17938 decreased mortality and the incidence and severity of NEC. The percentage of Tem cells in the ileum and mesenteric lymph nodes was increased in NEC but decreased by LR17938. Conversely, the percentage of CD4+Foxp3+ regulatory T (Treg) cells in the intestine decreased during NEC and was restored to normal by LR17938. The majority of the Treg cells preserved by LR17938 were Helios+ subsets, possibly of thymic origin. In conclusion, LR17938 may represent a useful treatment to prevent NEC. The mechanism of protection by LR17938 involves modulation of the balance between Tem and Treg cells. These T cell subsets might be potential biomarkers and therapeutic targets during intestinal inflammation.


2015 ◽  
Vol 213 (1) ◽  
pp. 123-138 ◽  
Author(s):  
Arata Takeuchi ◽  
Mohamed El Sherif Gadelhaq Badr ◽  
Kosuke Miyauchi ◽  
Chitose Ishihara ◽  
Reiko Onishi ◽  
...  

Naive T cells differentiate into various effector T cells, including CD4+ helper T cell subsets and CD8+ cytotoxic T cells (CTL). Although cytotoxic CD4+ T cells (CD4+CTL) also develop from naive T cells, the mechanism of development is elusive. We found that a small fraction of CD4+ T cells that express class I–restricted T cell–associated molecule (CRTAM) upon activation possesses the characteristics of both CD4+ and CD8+ T cells. CRTAM+ CD4+ T cells secrete IFN-γ, express CTL-related genes, such as eomesodermin (Eomes), Granzyme B, and perforin, after cultivation, and exhibit cytotoxic function, suggesting that CRTAM+ T cells are the precursor of CD4+CTL. Indeed, ectopic expression of CRTAM in T cells induced the production of IFN-γ, expression of CTL-related genes, and cytotoxic activity. The induction of CD4+CTL and IFN-γ production requires CRTAM-mediated intracellular signaling. CRTAM+ T cells traffic to mucosal tissues and inflammatory sites and developed into CD4+CTL, which are involved in mediating protection against infection as well as inducing inflammatory response, depending on the circumstances, through IFN-γ secretion and cytotoxic activity. These results reveal that CRTAM is critical to instruct the differentiation of CD4+CTL through the induction of Eomes and CTL-related gene.


2021 ◽  
Vol 22 (21) ◽  
pp. 11977
Author(s):  
Jocelyn C. Pérez-Lara ◽  
Enrique Espinosa ◽  
Leopoldo Santos-Argumedo ◽  
Héctor Romero-Ramírez ◽  
Gabriela López-Herrera ◽  
...  

CD38 is a transmembrane glycoprotein expressed by T-cells. It has been reported that patients with systemic lupus erythematosus (SLE) showed increased CD38+CD25+ T-cells correlating with immune activation and clinical signs. Contrariwise, CD38 deficiency in murine models has shown enhanced autoimmunity development. Recent studies have suggested that CD38+ regulatory T-cells are more suppressive than CD38− regulatory T-cells. Thus, we have suggested that CD38 overexpression in SLE patients could play a role in regulating immune activation cells instead of enhancing it. This study found a correlation between CD38 with FoxP3 expression and immunosuppressive molecules (CD69, IL-10, CTLA-4, and PD-1) in T-cells from lupus-prone mice (B6.MRL-Faslpr/J). Additionally, B6.MRL-Faslpr/J mice showed a decreased proportion of CD38+ Treg cells regarding wild-type mice (WT). Furthermore, Regulatory T-Cells (Treg cells) from CD38-/- mice showed impairment in expressing immunosuppressive molecules and proliferation after stimulation through the T-cell receptor (TCR). Finally, we demonstrated an increased ratio of IFN-γ/IL-10 secretion in CD38-/- splenocytes stimulated with anti-CD3 compared with the WT. Altogether, our data suggest that CD38 represents an element in maintaining activated and proliferative Treg cells. Consequently, CD38 could have a crucial role in immune tolerance, preventing SLE development through Treg cells.


2020 ◽  
Author(s):  
Xiaoxia Guo ◽  
Fang Du ◽  
Qin Liu ◽  
Yan Guo ◽  
Qingbing Wang ◽  
...  

Abstract Background This study intends to investigate the immunological effects of tumor ablation with irreversible electroporation (IRE). Methods We evaluated the systemic immune response in patients with hepatocellular carcinoma (HCC) after IRE treatment. Furthermore, we analyzed the tumor infiltrating T lymphocytes and the level of serum cytokines in IRE and control groups of tumor-bearing mice. Results We observed that IRE induced an increase in WBC, neutrophil and monocyte counts and a decrease in lymphocyte count 1 day post-IRE and returned to baseline values within 7 days in the patients. Meanwhile, circulating CD4+ T cell subsets, but not CD8+, decreased 1 day post-IRE. The activated T cells and natural killer (NK) cells increased, and regulatory T (Treg) cells decreased. Furthermore, a significant increase in cytotoxic CD8+ T cells infiltration was observed on ablative tumors in mice. The level of serum IFN-γ also significantly increased in the IRE group. Conclusions Our study demonstrated that IRE not only induced immediate innate immune response dominated by the increase of neutrophils, monocytes and NK cells, but also upregulated activated T cells and downregulated Treg. Meanwhile, the results from the animal model indicated that IRE could induce antitumor adaptive immunity dominated by cytotoxic CD8+ T cells.


Sign in / Sign up

Export Citation Format

Share Document