scholarly journals GPR15 in colon cancer development and anti-tumor immune responses.

2021 ◽  
Author(s):  
Hong NamKoong ◽  
Bomi Lee ◽  
Gayathri Swaminathan ◽  
Seong-Joon Koh ◽  
Stephan Rogalla ◽  
...  

G protein-coupled receptor 15 (GPR15) is a chemoattractant receptor which in response to its ligand, C10orf99/GPR15L, promotes colon homing of T cells in health and colitis. The functional role of GPR15 in colon cancer is largely unexplored, motivating our current studies using murine colon cancer models and human colorectal cancer (CRC) tissues. Our initial analysis of human CRC specimen revealed significant reduction in GPR15 expression and frequency of GPR15+ immune cells in tumors compared to ′tumor-free′ surgical margins. In the AOM/DSS murine model of colitis associated colon cancer (CAC), we observed increased colonic polyps/tumor burden and lower survival rate in Gpr15-deficient (KO) compared to Gpr15-sufficient (Het) mice. Analysis of immune cell infiltrates in the colonic polyps showed significantly decreased CD8+ T cells and increased IL-17+ CD4+ and IL-17+ CD8+ T cells in Gpr15-KO than in Het mice. GPR15 deficiency thus alters the immune environment in colonic polyps to mitigate T cell-mediated anti-tumor responses resulting in severe disease. Consistent with a protective role of GPR15, administration of GPR15L to established tumors in the MC38 CRC mouse model increased CD45+ cell infiltration, enhanced TNFα expression on CD4+ and CD8+ T cells at the tumor site and dramatically reduced tumor burden. Our findings highlight an important, unidentified role of the GPR15-GPR15L axis in promoting a tumor-suppressive immune microenvironment and unveils a novel, colon-specific therapeutic target for CRC.

2021 ◽  
Vol 39 (3_suppl) ◽  
pp. 116-116
Author(s):  
Priya Jayachandran ◽  
Joanne Xiu ◽  
Shivani Soni ◽  
Richard M. Goldberg ◽  
Benjamin Adam Weinberg ◽  
...  

116 Background: Cachexia affects many cancer patients. Growth differentiation factor-15 (GDF15) is a protein that regulates weight and the stress response of cells. The GDF15 gene encodes a ligand of TGF-beta that triggers cachexia and modulates the progression from tumorigenesis to metastasis. Inhibition of GDF15 with an antibody restored muscle mass and fat in animal models. Serum levels rise in proportion to the progression of colon cancer, predict outcome, and have been correlated with CEA. Methods: We retrospectively reviewed 7607 CRC tumors profiled by Caris Life Sciences (Phoenix, AZ) from 2019 to 2020. Profiling included whole transcriptome sequencing (RNA-Seq by NovoSeq). Tumor mutational burden, mismatch repair status, and pathway genomic alterations were evaluated. QuantiSEQ was used to assess immune cell infiltration in the tumor microenvironment. Results: GDF15 expression ranged from 0 to 593 transcripts per million (TPM) with median of 30 (IQR = 15.02). There was no association with age, sex, or primary tumor sidedness. MSI-H/dMMR tumors had higher GDF15 expression (median 37 vs 30, p = 0.0004); TMB > = 17 tumors was seen in 5.9% of bottom quartile (Q1) GDF15 expressors and 8.3% of top quartile (Q4). PDL1 IHC positivity was inversely correlated with GDF15 expression (7.1% in Q1 vs. 2.6% in Q4, p < 0.0001). Genomic alterations associated with higher GDF15 expression (Q4 vs Q1) included genes on TGF-B (SMAD2/4), PI3K (PIK3CA, MTOR), chromatin remodeling (ARID1A, KMT2C), DDR (ATM) and Wnt pathway (APC); those inversely associated included MYC CNA and TP53. Q1 tumors had higher CNA of ERBB2 and FGFR1. Relative neutrophils and NK cells in the TME increased from Q1 to Q4 (p < 0.001). There was a decrease in CD8+ T-cells and Treg cells from Q1 to Q4. Conclusions: GDF15 expression correlates with increased dMMR/MSI-H and TMB, but not with PDL1 expression. Mutations and activated pathways associated with GDF15 expression may explain increased cachexia with more aggressive disease. The association with chromatin remodeling may warrant therapies targeting histone modification and epigenetics. The increase in NK cells but decrease in CD8+ T cells in the TME with increasing GDF15 suggests approaches to treatment. Higher CD8+ lymphocyte counts correlate with PFS with immunotherapy. Anti-PD-L1 therapy reinvigorates the killing function of CD8+ T cells. The decrease in CD8+ T cells and PDL1 positivity with rising GDF15 suggests worse outcome and a lack of response to anti-PDL1 therapy. NK cell checkpoint inhibitors, CARs, and an anti-GFRAL antibody are now in clinical trials and might be utilized in high GDF15 cancers. GDF15 is emerging as a target in the treatment of obesity and cachexia and as a prognostic marker in oncology. Understanding its expression in metastatic colon cancer may reveal which patients could benefit from developing anti-GDF15 targeted therapies against cancer progression.


Cancers ◽  
2020 ◽  
Vol 12 (3) ◽  
pp. 714
Author(s):  
Jean Philippe Nesseler ◽  
Mi-Heon Lee ◽  
Christine Nguyen ◽  
Anusha Kalbasi ◽  
James W. Sayre ◽  
...  

The purpose of this study was to determine the dynamic contributions of different immune cell subsets to primary and abscopal tumor regression after hypofractionated radiation therapy (hRT) and the impact of anti-PD-1 therapy. A bilateral syngeneic FSA1 fibrosarcoma model was used in immunocompetent C3H mice, with delayed inoculation to mimic primary and microscopic disease. The effect of tumor burden on intratumoral and splenic immune cell content was delineated as a prelude to hRT on macroscopic T1 tumors with 3 fractions of 8 Gy while microscopic T2 tumors were left untreated. This was performed with and without systemic anti-PD-1. Immune profiles within T1 and T2 tumors and in spleen changed drastically with tumor burden in untreated mice with infiltrating CD4+ content declining, while the proportion of CD4+ Tregs rose. Myeloid cell representation escalated in larger tumors, resulting in major decreases in the lymphoid:myeloid ratios. In general, activation of Tregs and myeloid-derived suppressor cells allow immunogenic tumors to grow, although their relative contributions change with time. The evidence suggests that primary T1 tumors self-regulate their immune content depending on their size and this can influence the lymphoid compartment of T2 tumors, especially with respect to Tregs. Tumor burden is a major confounding factor in immune analysis that has to be taken into consideration in experimental models and in the clinic. hRT caused complete local regression of primary tumors, which was accompanied by heavy infiltration of CD8+ T cells activated to express IFN-γ and PD-1; while certain myeloid populations diminished. In spite of this active infiltrate, primary hRT failed to generate the systemic conditions required to cause abscopal regression of unirradiated microscopic tumors unless PD-1 blockade, which on its own was ineffective, was added to the RT regimen. The combination further increased local and systemically activated CD8+ T cells, but few other changes. This study emphasizes the subtle interplay between the immune system and tumors as they grow and how difficult it is for local RT, which can generate a local immune response that may help with primary tumor regression, to overcome the systemic barriers that are generated so as to effect immune regression of even small abscopal lesions.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 576-576
Author(s):  
Li Ma ◽  
Elisa K. Simpson ◽  
June Li ◽  
Min Xuan ◽  
Miao Xu ◽  
...  

Abstract Background:Immune thrombocytopenia (ITP) is a common bleeding disorder. Autoantibodies against platelet GPIIbIIIa (integrin αIIbβ3, 70-80%) and GPIb-complex (20-40%) are considered to be the major mechanism leading to autologous platelet destruction. Recent studies demonstrated that in addition to autoantibodies, CD8+ cytotoxic T cells (CTLs) also contribute to thrombocytopenia, either through direct cytotoxicity against platelets or megakaryocytes. However, the roles of CD8+ regulatory T cells (Tregs) in ITP have not been adequately explored. Methods and Results: We developed the first animal models of steroid treatment in ITP, encompassing both the passive and active forms. In the passive model, we injected anti-β3 antibodies to induce transient antibody mediated thrombocytopenia. We found that a single intraperitoneal (IP) injection of steroids post-antibody injection was effective at rescuing platelet counts. We also adapted an active model of ITP whereby wild-type (WT) BALB/c mice were transfused with splenocytes from WT platelet immunized β3-/-mice. This model encompasses both antibody and cell-mediated ITP resulting in sustained thrombocytopenia. In this model, we found steroid treatment (prednisone and dexamethasone) administered daily either orally or through IP-injection were equally efficacious at ameliorating thrombocytopenia. Furthermore, immunophenotyping and cytokine analysis reveal a similar profile as reported of human ITP patients responsive to steroid treatments. Thus, successful steroid treatments in these animal models are representative of the therapeutic effects of steroid treatments seen in human ITP patients. To study the role of CD8+ T cells in the pathogenesis and response to steroid treatments in ITP, we depleted CD8+ T cells from splenocytes prior to its transfusion into WT mice. Unexpectedly,we found CD8+ T cell depleted splenocyte (lacking in CTL cells) engrafted mice had lower, but not higher, platelet counts and were less responsive to dexamethasone (DEX) treatment compared to non-depleted engrafted mice. Furthermore, in the passive ITP model, depletion of CD8+ T cells from mice prior to injection of anti-β3 antibodies resulted in more severe thrombocytopenia, compared with non-depleted mice. Conversely, transfusion of either antigen-primed CD8+ (isolated from immunized β3-/- splenocytes) or WT/β3-/- naïve CD8+ T cells alone was sufficient to rescue platelet counts and improve response to DEX in the passive ITP model. These results indicate for the first time that CD8+ T cells from both antigen-primed and naïve populations play a protective role in attenuating platelet clearance. In further support of these observations, we detected significant increased populations of both CTLs and CD8+ Tregs including, CD8+CD25+Foxp3+, CD8+CD103+, CD8+CD122+ and CD8+CD28- in the blood, and spleen of immunized β3-/- mice. Interestingly, the CD8+ Tregs populations were further increased while CTL population decreased following DEX treatment in the active ITP model. In vitro splenocyte cultures were used to explore putative regulatory mechanisms of CD8+ Tregs. It was found that antigen-primed CD8+ Tregs exerted significantly stronger inhibition CD4+ T- and CD19+ B cell proliferation, platelet apoptosis, and platelet associated IgG production in the presence of platelet antigens, while both antigen-primed and naïve CD8+ Tregs could effectively inhibit macrophage mediated phagocytosis of anti-β3 opsonized platelets. Conclusion: To the best of our knowledge, these are the first reported animal models of effective steroid treatment of ITP. Utilizing these models we uncovered a previously unidentified regulatory role of CD8+ T cells in both ITP and steroid treatment. The increased populations of various CD8+ Tregs following β3-/- immunization exerted a significant inhibitory function against other immune-cell mediated anti-platelet responses. In addition, therapeutic administration of both antigen-primed and naïve CD8+ T cells were able to rescue platelet counts in the passive ITP model. This suggests that CD8+ Treg may play a predominantly protective role in ITP. These data provides significant insights into the understanding of immunopathogenesis of ITP, which may be important in designing effective therapy including the potential usage of CD8+ Tregs as a cellular target in the treatment of ITP. Disclosures No relevant conflicts of interest to declare.


1993 ◽  
Vol 178 (5) ◽  
pp. 1837-1842 ◽  
Author(s):  
J M Penninger ◽  
N Neu ◽  
E Timms ◽  
V A Wallace ◽  
D R Koh ◽  
...  

Experimental induction of most autoimmune diseases appears to depend on the activation of CD4+ T helper cells, while CD8+ lymphocytes may have a role in disease progression. To study the role of CD4+ and CD8+ T cell subsets in T cell-dependent autoimmunity, mice lacking CD4 or CD8 molecules after gene targeting were injected with cardiac myosin to induce organ specific autoimmune myocarditis. Mice homozygous for the CD8 mutation (CD8-/-) developed significantly more severe disease as compared to CD4+/-CD8+/- controls. Surprisingly, CD4-/- mice developed autoimmune myocarditis with infiltration of TCR alpha beta +CD4-CD8- T cells in the heart tissue and appearance of autoantibodies. These data demonstrate that the lack of CD4+ or CD8+ T cells has no significant influence on the initiation of autoimmune myocarditis. CD4+ and CD8+ cells regulate disease severity and these results may explain the occurrence of autoimmunity in CD4 immunodeficiencies.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Yinwen Cheng ◽  
Nicholas Borcherding ◽  
Ayomide Ogunsakin ◽  
Caitlin D. Lemke-Miltner ◽  
Katherine N. Gibson-Corley ◽  
...  

AbstractThe Toll-like receptor 8 (TLR8) agonist VTX-2337 (motolimod) is an anti-cancer immunotherapeutic agent that is believed to augment natural killer (NK) and dendritic cell (DC) activity. The goal of this work is to examine the role of TLR8 expression/activity in head and neck squamous cell carcinoma (HNSCC) to facilitate the prediction of responders to VTX-2337-based therapy. The prognostic role of TLR8 expression in HNSCC patients was assessed by TCGA and tissue microarray analyses. The anti-tumor effect of VTX-2337 was determined in SCCVII/C3H, mEERL/C57Bl/6 and TUBO-human EGFR/BALB/c syngeneic mouse models. The effect of combined VTX-2337 and cetuximab treatment on tumor growth, survival and immune cell recruitment was assessed. TLR8 expression was associated with CD8+ T cell infiltration and favorable survival outcomes. VTX-2337 delayed tumor growth in all 3 syngeneic mouse models and significantly increased the survival of cetuximab-treated mice. The anti-tumor effects of VTX-2337+ cetuximab were accompanied by increased splenic lymphoid DCs and IFNγ+ CD4+ and tumor-specific CD8+ T cells. Depletion of CD4+ T cells, CD8+ T cells and NK cells were all able to abolish the anti-tumor effect of VTX-2337+ cetuximab. Altogether, VTX-2337 remains promising as an adjuvant for cetuximab-based therapy however patients with high TLR8 expression may be more likely to derive benefit from this drug combination compared to patients with low TLR8 expression.


Cancers ◽  
2021 ◽  
Vol 13 (11) ◽  
pp. 2630
Author(s):  
Annabel Meireson ◽  
Simon J. Tavernier ◽  
Sofie Van Gassen ◽  
Nora Sundahl ◽  
Annelies Demeyer ◽  
...  

(1) Background: Blockade of the PD-1/PD-L1 pathway has revolutionized the oncology field in the last decade. However, the proportion of patients experiencing a durable response is still limited. In the current study, we performed an extensive immune monitoring in patients with stage III/IV melanoma and stage IV UC who received anti-PD-1 immunotherapy with SBRT. (2) Methods: In total 145 blood samples from 38 patients, collected at fixed time points before and during treatment, were phenotyped via high-parameter flow cytometry, luminex assay and UPLC-MS/MS. (3) Results: Baseline systemic immunity in melanoma and UC patients was different with a more prominent myeloid compartment and a higher neutrophil to lymphocyte ratio in UC. Proliferation (Ki67+) of CD8+ T-cells and of the PD-1+/PD-L1+ CD8+ subset at baseline correlated with progression free survival in melanoma. In contrast a higher frequency of PD-1/PD-L1 expressing non-proliferating (Ki67−) CD8+ and CD4+ T-cells before treatment was associated with worse outcome in melanoma. In UC, the expansion of Ki67+ CD8+ T-cells and of the PD-L1+ subset relative to tumor burden correlated with clinical outcome. (4) Conclusion: This study reveals a clearly different immune landscape in melanoma and UC at baseline, which may impact immunotherapy response. Signatures of proliferation in the CD8+ T-cell compartment prior to and early after anti-PD-1 initiation were positively correlated with clinical outcome in both cohorts. PD-1/PD-L1 expression on circulating immune cell subsets seems of clinical relevance in the melanoma cohort.


2020 ◽  
Author(s):  
Zhengshui Xu ◽  
Chao Qu ◽  
Jing Guo ◽  
Xiaopeng Li ◽  
Yunhua Wu ◽  
...  

Abstract Backgroud:Tumor mutation burden has become a powerful bio-marker to predict prognosis and immunotherapy responsiveness to patients in various cancers, but the role of TMB in colon cancer is still unclear.Methods:The transcriptome profiling data of colon patients and the simple nucleotide variation data of colon cases were downloaded from the Cancer Genome Atlas (TCGA) database. The groups were divided into high TMB and low TMB group according to the median of TMB. Then we explored the relationship between immune checkpoints, immune cells and TMB, respectively. Results: Mutation profiles of 399 colon cancer samples were analyzed in TCGA database. The senior (age>65) had a strong relationship with higher-TMB level(p=0.001). Low-TMB group correlated with advanced N stage (P<0.001), M stage (P<0.001), and pathologic stage(P<0.001). High-TMB group had significantly higher mRNA level of PD-L1, TIGIT, HAVCR2, and LAG3 than low-TMB group, which indicated high-TMB referred to better immunotherapy responsiveness in colon cancer. And high-TMB level correlated with higher fractions of CD8T cells (p=0.021), higher CD4 memory T cells(p=0.039), follicular helper T cells (p=0.002)and M1 macrophages (p<0.001), while the low-TMB groups correlated with higher regulator T cells (p=0.002). So high-TMB correlated with stronger immune cell infiltrationConclusions:The high TMB referred to better clinical pathologic features, better immunotherapy responsiveness and stronger immune cells infiltration in colon cancer. Hence TMB may be a very promising bio-marker to predict prognosis and immunotherapy responsiveness to patients in colon cancer.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 5-5
Author(s):  
Spyridoula Vasileiou ◽  
Manik Kuvalekar ◽  
Aster Workineh ◽  
Ayumi Watanabe ◽  
Yovana Velazquez ◽  
...  

Background. On 11 March, 2020 the World Health Organization declared COVID-19, caused by SARS-CoV-2, a global pandemic with almost 17,000,000 confirmed cases worldwide by the end of July, of which 4,500,000 were in the US. Approximately 20% of patients develop severe disease that can evolve into acute respiratory distress syndrome leading to respiratory or multiorgan failure, with an overall mortality of up to 4%. Older age, comorbidities such as hypertension and diabetes, and immune compromise have been identified as major risk factors associated with poor prognosis. For example, in immunocompromised HSCT patients mortality rates as high as 20% have been reported (www.cibmtr.org/COVID19). Furthermore, there is accumulating evidence regarding the protective role of T cells, with reduced counts and dysregulation seen more prominently in individuals with severe rather than mild COVID-19. Our group has previously demonstrated the feasibility, safety and clinical efficacy of administering allogeneic ex vivo expanded multivirus-specific T cells (multi-VSTs) as a banked, off-the-shelf product for the treatment of EBV, CMV, BKV, HHV6 and AdV infections/disease in immunocompromised individuals. Given the lack of preventative or therapeutic agents and the emerging evidence of the pivotal protective role of SARS-CoV-2-specific CD4+ and CD8+ T cells, we sought to explore the feasibility of developing a banked, SARS-CoV-2-specific VST product to treat those at highest risk of severe COVID-19 disease (i.e. HSCT recipients, elderly individuals, patients with comorbidities). Methods. To first identify immunogenic and protective SARS-CoV-2 antigens we screened PBMCs from convalescent individuals with mild COVID-19 (not requiring hospitalization) for T cell activity against overlapping peptide libraries (pepmixes) spanning 18 structural and non-structural SARS-CoV-2 proteins of which 8 [structural proteins: Spike (S), Membrane (M) and Nucleoprotein (N); non-structural proteins (Nsps): 3, 4, 6, 12; and the accessory protein (AP) 7a] were identified as immunodominant and advanced for VST manufacturing. We subsequently utilized our optimized VST manufacturing process and culture in a G-Rex device in medium supplemented with activating cytokines to generate SARS-CoV-2-specific T cells with activity against this combination of immunodominant targets. Results. We achieved a mean 29±7 fold expansion (mean±SEM; n=5) of cells that were comprised almost exclusively of CD3+ T cells (97.1±0.7%; mean±SEM), with a mixture of cytotoxic (CD8+; 10.2±1.2%) and helper (CD4+; 85.5±1.8%) T cells. These cells had a phenotype consistent with effector function and memory potential, as evidenced by upregulation of the activation markers CD25, CD69, and CD28 and expression of central (CD45RO+/CD62L+) and effector memory markers (CD45RO+/CD62L−), with minimal PD1 or Tim3 expression. To confirm the anti-viral activity of our expanded cells we performed an IFNγ ELIspot using each of the individual stimulating antigens as an immunogen and all lines proved to be reactive against the target antigens [S: 2,118±479 SFC/2x105; M: 1,084±182; N: 1,124±335; Nsp3: 71±48.6; Nsp4: 68±30; Nsp6: 23±6.7; AP7a: 65±43; and Nsp12: 29±9]. As demonstrated by intracellular cytokine staining (ICS), the immune response was mediated by both CD4+ and CD8+ T cell subsets, and the majority of IFNg-producing cells also produced TNFa. Reactive cells exhibited a primarily Th1-polarized profile as measured by Granzyme B production, luminex array and single-cell protein analysis. In addition, the expanded cells were able to kill viral pepmix-loaded autologous PHA blasts with minimal/no activity against non-antigen-expressing autologous and allogeneic targets (Figure 1). Conclusion. SARS-CoV-2 VSTs generated from convalescent individuals are Th1-polarized, polyfunctional and selectively able to kill viral antigen-expressing targets with no auto- or alloreactivity, indicative of both their selectivity and safety for clinical use. We are rapidly advancing this product to the clinic for administration in a randomized clinical trial (VSTs+SOC vs SOC) to prevent the development of severe disease in high risk hospitalized patients such as those post-transplant or therapy for hematologic malignancy. Figure 1: SARS-CoV-2 Specific T cells Demonstrate Selective Cytolytic Activity against viral antigen-expressing targets. Figure 1 Disclosures Vasileiou: AlloVir: Consultancy. Kuvalekar:AlloVir: Consultancy. Workineh:AlloVir: Current Employment. Watanabe:AlloVir: Consultancy. Heslop:Novartis: Consultancy; Gilead Biosciences: Consultancy; PACT Pharma: Consultancy; Kiadis: Consultancy; Tessa Therapeutics: Consultancy, Research Funding; AlloVir: Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees; Marker Therapeutics: Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees. Hill:Incyte: Membership on an entity's Board of Directors or advisory committees. Leen:AlloVir: Consultancy, Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees; Marker Therapeutics: Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees.


2021 ◽  
Vol 218 (7) ◽  
Author(s):  
Naveen Sharma ◽  
Oluwatomisin T. Atolagbe ◽  
Zhongqi Ge ◽  
James P. Allison

Immune receptors expressed on TAMs are intriguing targets for tumor immunotherapy. In this study, we found inhibitory receptor LILRB4 on a variety of intratumoral immune cell types in murine tumor models and human cancers, most prominently on TAMs. LILRB4, known as gp49B in mice, is a LILRB family receptor. Human and murine LILRB4 have two extracellular domains but differ in the number of intracellular ITIMs (three versus two). We observed a high correlation in LILRB4 expression with other immune inhibitory receptors. After tumor challenge, LILRB4−/− mice and mice treated with anti-LILRB4 antibody showed reduced tumor burden and increased survival. LILRB4−/− genotype or LILRB4 blockade increased tumor immune infiltrates and the effector (Teff) to regulatory (Treg) T cell ratio and modulated phenotypes of TAMs toward less suppressive, CD4+ T cells to Th1 effector, and CD8+ T cells to less exhausted. These findings reveal that LILRB4 strongly suppresses tumor immunity in TME and that alleviating that suppression provides antitumor efficacy.


Sign in / Sign up

Export Citation Format

Share Document