scholarly journals P08.02 CCR2/CCR5 dual-antagonist ‘licenses’ the radiation-induced effector T-cell infiltration in the anti-PD-1 antibody-treated pancreatic adenocarcinoma

2021 ◽  
Vol 9 (Suppl 1) ◽  
pp. A25.1-A25
Author(s):  
J Wang ◽  
M Tun Saung ◽  
K Fujiwara ◽  
N Niu ◽  
A Narang ◽  
...  

BackgroundThe resistance of pancreatic ductal adenocarcinoma(PDAC) to immune checkpoint inhibitors(ICIs) is mainly attributed to the immune-quiescent nature of its tumor microenvironment(TME). Radiotherapy(RT) activates innate responses including the RAGE and TLR2/4 pathways and subsequently modifies the TME by promoting the release of chemokines that recruit inflammatory cells into the TME. In this preclinical study, we examined the PDAC vaccine or RT as a T-cell priming mechanism together with BMS-687681, a small molecule dual-antagonist of CCR2 and CCR5(CCR2/5i) as an immunosuppressive TME-targeting agent, in combination with the anti-PD-1 antibody(αPD-1) as a new treatment.Materials and MethodsThe hemi-spleen and Orthotopic mice model were used to investigate both GVAX and RT as T-cell priming agents in combination regimens that included αPD-1 and CCR2/5i. Dissected orthotopic pancreatic tumors were collected for analysis of tumor-infiltrating immune cells by flow cytometry. RNA from tumor-infiltrating immune cell pellets and whole-exome RNA sequencing was performed for further mechanism research.ResultsCCR2 and CCR5 are associated with the immunosuppressive TME of PDAC patients and their expression were induced after treatment with GVAX+nivolumab. Using a mouse model of PDAC, we demonstrated that the addition of GVAX to CCR2/5i+αPD-1 combination therapy did not significantly improve antitumor activity. However, RT followed by αPD-1 and prolonged treatment with CCR2/5i conferred significantly better antitumor efficacy compared to the other combination treatments we studied. The combination of RT, αPD-1, and CCR2/5i enhanced intratumoral effector and memory T-cell infiltration. This combination suppressed Treg, M2-like TAM, and M-MDSC infiltration, but not M1-like TAM and PMN-MDSC infiltration. Finally, RNA sequencing showed that CCR2/5i partially inhibited RT-induced TLR2/4&RAGE signaling, which would have otherwise led to the release of immunosuppressive cytokines including CCL2 and CCL5. The inhibition of TLR2/4&RAGE signaling permitted the expression of effector T-cell chemokines such as CCL17 and CCL22.ConclusionsThis study thus supports the clinical development of CCR2/5i in combination with RT and ICIs for PDAC treatment.Disclosure InformationJ. Wang: None. M. Tun Saung: None. K. Fujiwara: None. N. Niu: None. A. Narang: None. J. He: None. L. Zheng: None.

2019 ◽  
Vol 1445 (1) ◽  
pp. 62-73 ◽  
Author(s):  
Neeha Zaidi ◽  
Sergio A. Quezada ◽  
Janelle M.Y. Kuroiwa ◽  
Li Zhang ◽  
Elizabeth M. Jaffee ◽  
...  

2015 ◽  
Vol 38 (1) ◽  
pp. 1-11 ◽  
Author(s):  
Kevin C. Soares ◽  
Agnieszka A. Rucki ◽  
Annie A. Wu ◽  
Kelly Olino ◽  
Qian Xiao ◽  
...  

2021 ◽  
Vol 8 ◽  
Author(s):  
Yiping Zou ◽  
Zhihong Chen ◽  
Hongwei Han ◽  
Shiye Ruan ◽  
Liang Jin ◽  
...  

Background: Hepatocellular carcinoma (HCC) is the most common histological type of liver cancer, with an unsatisfactory long-term survival rate. Despite immune checkpoint inhibitors for HCC have got glories in recent clinical trials, the relatively low response rate is still a thorny problem. Therefore, there is an urgent need to screen biomarkers of HCC to predict the prognosis and efficacy of immunotherapy.Methods: Gene expression profiles of HCC were retrieved from TCGA, GEO, and ICGC databases while the immune-related genes (IRGs) were retrieved from the ImmPort database. CIBERSORT and WGCNA algorithms were combined to identify the gene module most related to CD8+ T cells in the GEO cohort. Subsequently, the genes in hub modules were subjected to univariate, LASSO, and multivariate Cox regression analyses in the TCGA cohort to develop a risk signature. Afterward, the accuracy of the risk signature was validated by the ICGC cohort, and its relationships with CD8+ T cell infiltration and PDL1 expression were explored.Results: Nine IRGs were finally incorporated into a risk signature. Patients in the high-risk group had a poorer prognosis than those in the low-risk group. Confirmed by TCGA and ICGC cohorts, the risk signature possessed a relatively high accuracy. Additionally, the risk signature was demonstrated as an independent prognostic factor and closely related to the CD8+ T cell infiltration and PDL1 expression.Conclusion: A risk signature was constructed to predict the prognosis of HCC patients and detect patients who may have a higher positive response rate to immune checkpoint inhibitors.


2020 ◽  
Vol 8 (Suppl 2) ◽  
pp. A59.2-A60
Author(s):  
A Osipov ◽  
L Zheng

BackgroundPancreatic ductal adenocarcinoma (PDAC) is resistant to immune checkpoint inhibition. One of the major resistance mechanisms is attributed to myeloid cells as an immunosuppressive element within the stroma of PDAC. It has been reported that focal adhesion kinase inhibitor (FAKi) can suppress immunosuppressive myeloid cells such as tumor associated macrophages (TAMs) and myeloid derived suppressor cells (MDSC), consequently sensitizing tumor to anti-PD1 antibody in mouse models of PDAC. Our group has previously shown in a murine model that targeting the stroma via PEGylated recombinant human hyaluronidase (PEGPH20) enhanced the anti-tumor activity of the whole cell vaccine (GVAX) by targeting CXCR4-expressing myeloid cells and led to an increase in infiltration of CCR7- effector memory T cell subsets. Here, we evaluate the hypothesis that FAK expressing myeloid cell subsets modulate T cell infiltration in human PDAC and FAKi can synergize with PEGPH20 by targeting myeloid cells in PDAC.Material and MethodsResected human PDAC tissue specimens treated with GVAX and anti-PD1 therapy was used to assess FAK expression in myeloid cell subsets and its impact on T cell infiltration. A sequential staining and stripping multiplex IHC technique that incorporates 28 myeloid and lymphoid biomarkers, as well as phosphorylated FAK (pFAK) combined with computational image processing was used to assess myeloid cell populations, T cell infiltration and FAK expression.An established murine model of metastatic PDAC treated with and without anti-PD1 therapy was used to assess the synergy and immune-modulating effect of FAKi and stromal degradation of hyaluronan via PEGPH20.ResultsIn human PDAC, FAK is widely expressed in TAMs and neutrophils. Increased FAK expression is associated with increased CXCR4 expression. Lower pFAK density in neutrophils and M2 TAMs, but not lower pFAK density in M1 TAMs, is associated with higher CD8+ T cell infiltration.FAKi and combination of FAKi with anti-PD1 extends survival in the mouse metastasis model of PDAC. Adding PEGPH20 to FAKi and anti-PD1 antibody significantly prolonged survival in this model. Comparing to the combination of FAKi and anti-PD1 antibody, adding PEGPH20 significantly decreased the number of CXCR4-expressing myeloid cells in the tumor microenvironment (TME) of PDAC and consequently led to an increase in the amount of CCR7+ central memory T cells. Additionally, the amount of G-MDSCs, inflammatory resident monocytes and PDL1 expressing myeloid cells in the TME of PDAC, was also decreased in PDAC treated with the triple combination of PEGPH20, FAKi and anti-PD1 antibody compared to FAKi and anti-PD1 antibody.ConclusionFAK is widely expressed in myeloid cell populations, directly correlated with CXCR4 expression and decreased FAK expression in a myeloid (M2 TAMs, neutrophil) inflamed stroma is associated with infiltration of effector CD8 T cells in human PDAC. Stromal degradation of hyaluronan via PEGPH20 combined with FAKi and anti-PD1 antibody further depletes immunosuppressive cells in the TME including G-MDSCs, inflammatory resident monocytes and PDL1 expressing myeloid cells and appears to target the CXCR4 pathway through PEGPH20. These findings support testing the combination of FAKi and anti-PD1 antibody with agents targeting CXCR4 directly or indirectly by PEGPH20 in human PDAC.Disclosure InformationA. Osipov: None. L. Zheng: None.


2011 ◽  
Vol 17 (13) ◽  
pp. 4296-4308 ◽  
Author(s):  
Jennifer Lohr ◽  
Thomas Ratliff ◽  
Andrea Huppertz ◽  
Yingzi Ge ◽  
Christine Dictus ◽  
...  

2019 ◽  
Vol 37 (15_suppl) ◽  
pp. e17542-e17542
Author(s):  
Theodoros Rampias ◽  
Christos K. Kontos ◽  
Alexandros Polyzos ◽  
Aris Giotakis ◽  
Evangelos Giotakis ◽  
...  

e17542 Background: We sought to analyze the transcriptional landscape of HNSCC in an attempt to identify tumor-intrinsic oncogenic pathways that appear to mediate T-cell infiltration of tumor tissue. In this direction, we employ a methodology that integrates histopathology data of the tumor microenvironment with its corresponding transcriptome. Methods: 32 frozen HNSCCs were subjected to RNA-seq and corresponding FFPE were scored for plasma cells, tertiary lymphoid structures and CD8a+ TILs (center, invasive margin). RNA-seq data were analyzed to identify differentially expressed genes (DEGs) between tumors scored by immunohistochemistry (IHC) as CD8a high and CD8a low. Gene ontology analysis (GO) was performed based on DEGs > 1.5 fold expression change between CD8a high and CD8a low groups. Candidate genes were investigated by hierarchical clustering in TCGA RNA-seq data and further validated by IHC and quantitative RT-PCR in our cohort. Results: 32 HNSCCs were either scored as CD8a high or CD8a low based on IHC detection of CD8a+ cells in invasive margin of tumors. Comparative analysis of mRNA expression data between CD8a high and CD8a low groups in our cohort revealed that Muc1/16 overexpression and glycosylation was highly enriched in T cell infiltrated group of tumors. This finding was further validated using antibodies that detect glycosylated epitopes for both mucins. Analysis of TCGA RNA-seq data indicated that Muc1/16 overexpressing tumors share signatures of early keratinocyte differentiation and stem cell identity and co-express high levels of enzymes that promote Muc1/16 glycosylation. Interestingly, loss of CDH1 and acquisition of epithelial mesenchymal transition (EMT) markers in the cluster of Muc1/16 overexpressing tumors is strongly correlated with elevated CD8a, IDO1, CD274 and CXCL10 mRNA levels (P < 0.0001). Conclusions: Muc1/16 overexpressing tumors represent a very immunogenic HNSCC cluster. Previous studies have shown that mucins 1 and 16 in cancer cells expose glycosylated-specific epitopes that are recognized by T cells as cancer antigens. To this end, MUC1/16 expression may serve as predictive biomarkers for response to immunotherapy and MUC-targeted immunotherapy may function as an attractive partner to checkpoint inhibitors in HNSCC.


2017 ◽  
Vol 35 (6_suppl) ◽  
pp. 472-472
Author(s):  
Haris Zahoor ◽  
Paul G Pavicic ◽  
Christopher Przybycin ◽  
Paul Elson ◽  
C. Marcela Diaz-Montero ◽  
...  

472 Background: T cell infiltration in tumors has been investigated as a biomarker of response to checkpoint inhibitors. A neo-adjuvant trial of checkpoint inhibition in locally-advanced RCC is ongoing at Cleveland Clinic, where T cell infiltration in pre-treatment renal mass bx will be compared to post-treatment nx specimens. However, there are no data regarding the association of T cell infiltration in matched bx and nx samples without intervening treatment. Understanding this association will enable further study of this potential biomarker in future neo-adjuvant studies. Methods: Matched bx and nx samples (without intervening systemic therapy) were identified from patients with non-metastatic RCC. Demographic and pathological data were collected from chart review. Selected tissue sections from bx and nx samples of each patient were reviewed, and marked for tumor and intra-tumoral lymphocytes by the pathologist. Immunohistochemistry (IHC) was utilized to stain these selected tissue sections for T cell markers (CD3, CD4 and CD8). Intra-tumoral T cells were then quantified in the pre-marked tissue sections as counts per total tumor area surveyed, using Image-Pro Plus (Media Cybernetics, Inc.). Spearman correlation (ρ) was used to measure the strength of association of T cell infiltration between matched samples. Results: 30 matched pairs were investigated. The median interval between bx and nx was 2.8 (0.2-87.7) months. Clear cell was the most common histology (29/30; 97%). 15/30 (50%) had grade 3-4 tumors, 2/19 (11%) patients had sarcomatoid features, 7/25 (29%) had necrosis, and 8/28 (29%) had lymphovascular invasion. We found a positive correlation between the frequencies of CD8+ T cells between matched bx and nx samples (ρ= 0.39; p=0.03). CD3+ and CD4+T cells did not show significant correlation. (Table) Conclusions: Bx material can be used to accurately assess the degree of CD8+T cell infiltration in RCC. [Table: see text]


Sign in / Sign up

Export Citation Format

Share Document