scholarly journals TP53 missense mutations in PDAC are associated with enhanced fibrosis and an immunosuppressive microenvironment

2021 ◽  
Vol 118 (23) ◽  
pp. e2025631118
Author(s):  
Martino Maddalena ◽  
Giuseppe Mallel ◽  
Nishanth Belugali Nataraj ◽  
Michal Shreberk-Shaked ◽  
Ori Hassin ◽  
...  

Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal cancer, which is refractory to all currently available treatments and bears dismal prognosis. About 70% of all PDAC cases harbor mutations in the TP53 tumor suppressor gene. Many of those are missense mutations, resulting in abundant production of mutant p53 (mutp53) protein in the cancer cells. Analysis of human PDAC patient data from The Cancer Genome Atlas (TCGA) revealed a negative association between the presence of missense mutp53 and infiltration of CD8+ T cells into the tumor. Moreover, CD8+ T cell infiltration was negatively correlated with the expression of fibrosis-associated genes. Importantly, silencing of endogenous mutp53 in KPC cells, derived from mouse PDAC tumors driven by mutant Kras and mutp53, down-regulated fibrosis and elevated CD8+ T cell infiltration in the tumors arising upon orthotopic injection of these cells into the pancreas of syngeneic mice. Moreover, the tumors generated by mutp53-silenced KPC cells were markedly smaller than those elicited by mutp53-proficient control KPC cells. Altogether, our findings suggest that missense p53 mutations may contribute to worse PDAC prognosis by promoting a more vigorous fibrotic tumor microenvironment and impeding the ability of the immune system to eliminate the cancer cells.

2021 ◽  
Vol 16 (1) ◽  
Author(s):  
Steven F. Gameiro ◽  
Farhad Ghasemi ◽  
Peter Y. F. Zeng ◽  
Neil Mundi ◽  
Christopher J. Howlett ◽  
...  

Abstract Background Frequent mutations in the nuclear receptor binding SET domain protein 1 (NSD1) gene have been observed in head and neck squamous cell carcinomas (HNSCC). NSD1 encodes a histone 3 lysine-36 methyltransferase. NSD1 mutations are correlated with improved clinical outcomes and increased sensitivity to platinum-based chemotherapy agents in human papillomavirus-negative (HPV-) tumors, despite weak T-cell infiltration. However, the role of NSD1 and related family members NSD2 and NSD3 in human papillomavirus-positive (HPV+) HNSCC is unclear. Methods Using data from over 500 HNSCC patients from The Cancer Genome Atlas (TCGA), we compared the relative level of mRNA expression of NSD1, NSD2, and NSD3 in HPV+ and HPV- HNSCC. Correlation analyses were performed between T-cell infiltration and the relative level of expression of NSD1, NSD2, and NSD3 mRNA in HPV+ and HPV- HNSCC. In addition, overall survival outcomes were compared for both the HPV+ and HPV- subsets of patients based on stratification by NSD1, NSD2, and NSD3 expression levels. Results Expression levels of NSD1, NSD2 or NSD3 were not correlated with altered lymphocyte infiltration in HPV+ HNSCC. More importantly, low expression of NSD1, NSD2, or NSD3 correlated with significantly reduced overall patient survival in HPV+, but not HPV- HNSCC. Conclusion These results starkly illustrate the contrast in molecular features between HPV+ and HPV- HNSCC tumors and suggest that NSD1, NSD2, and NSD3 expression levels should be further investigated as novel clinical metrics for improved prognostication and patient stratification in HPV+ HNSCC.


2021 ◽  
Vol 49 (1) ◽  
pp. 030006052098153
Author(s):  
Qing Bi ◽  
Yang Liu ◽  
Tao Yuan ◽  
Huizhen Wang ◽  
Bin Li ◽  
...  

Objective The role of tumor-infiltrating lymphocytes (TILs) has not yet been characterized in sarcomas. The aim of this bioinformatics study was to explore the effect of TILs on sarcoma survival and genome alterations. Methods Whole-exome sequencing, transcriptome sequencing, and survival data of sarcoma were obtained from The Cancer Genome Atlas. Immune infiltration scores were calculated using the Tumor Immune Estimation Resource. Potential associations between abundance of infiltrating TILs and survival or genome alterations were examined. Results Levels of CD4+ T cell infiltration were associated with overall survival of patients with pan-sarcomas, and higher CD4+ T cell infiltration levels were associated with better survival. Somatic copy number alterations, rather than mutations, were found to correlate with CD4+ T cell infiltration levels. Conclusions This data mining study indicated that CD4+ T cell infiltration levels predicted from RNA sequencing could predict sarcoma prognosis, and higher levels of CD4+ T cells infiltration indicated a better chance of survival.


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.


2021 ◽  
Vol 3 (Supplement_2) ◽  
pp. ii17-ii18
Author(s):  
Apeng Chen ◽  
Yinan Jiang ◽  
Zhengwei Li ◽  
Xiangwei Xiao ◽  
Dean Yimlamai ◽  
...  

Abstract Glioblastoma (GBM) is the most common and highly malignant brain tumor in adults. Despite advances in multimodal treatment, GBM remains largely incurable. While immunotherapies have been highly effective in some types of cancer, the disappointing results from clinical trials for GBM immunotherapy represent continued challenges. GBM is highly immunosuppressive and resistant to immunotherapy because of glioma cells escaping from immune surveillance by reprograming the tumor microenvironment (TME). However, understanding the mechanisms of immune evasion by GBM remains elusive. Based on unbiased approaches, we found that Chitinase-3-like-1 (CHI3L1), also known as human homolog YKL-40, is highly expressed in GBM, which is regulated by the CHI3L1-PI3K/AKT/mTOR signaling in a positive feedback loop. Gain- and loss-function studies reveal that CHI3L1 plays a predominant role in regulating an immunosuppressive microenvironment by reprogramming tumor-associated macrophages (TAMs). Using the liquid chromatography-mass spectrometry and orthogonal structure-based screening, we found that Galectin-3 binding protein (Gal3BP) and its binding partner, Galectin-3 (Gal3), can interact competitively with the same binding motif on CHI3L1, leading to selective migration of M2-like versus M1-like bone marrow-derived macrophages (BMDMs) and resident microglia (MG). Mechanistically, the CHI3L1-Gal3 protein complex governs a transcriptional program of NFκB/CEBPβ to control the protumor phenotype of BMDMs, leading to inhibition of T cell infiltration and activation in the GBM TME. However, Gal3BP can reverse CHI3L1-Gal3 induced signaling pathway activation and subsequent protumor phenotype in TAMs. Based on protein binding motifs, a newly developed Gal3BP mimetic peptide can attenuate immune suppression and tumor progression in the syngeneic GBM mouse models, including decreasing M2-like TAMs and increasing M1-like TAMs and T cell infiltration. Together, these results shed light on the role of CHI3L1 protein complexes in immune evasion by glioblastoma and as a potential immunotherapeutic target for this devastating disease.


2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Guohao Wang ◽  
Junji Xu ◽  
Jiangsha Zhao ◽  
Weiqin Yin ◽  
Dayong Liu ◽  
...  

AbstractCancer stem cells (CSCs) may be responsible for treatment resistance, tumor metastasis, and disease recurrence. Here we demonstrate that the Arf1-mediated lipid metabolism sustains cells enriched with CSCs and its ablation induces anti-tumor immune responses in mice. Notably, Arf1 ablation in cancer cells induces mitochondrial defects, endoplasmic-reticulum stress, and the release of damage-associated molecular patterns (DAMPs), which recruit and activate dendritic cells (DCs) at tumor sites. The activated immune system finally elicits antitumor immune surveillance by stimulating T-cell infiltration and activation. Furthermore, TCGA data analysis shows an inverse correlation between Arf1 expression and T-cell infiltration and activation along with patient survival in various human cancers. Our results reveal that Arf1-pathway knockdown not only kills CSCs but also elicits a tumor-specific immune response that converts dying CSCs into a therapeutic vaccine, leading to durable benefits.


2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Alexander F. Haddad ◽  
Jia-Shu Chen ◽  
Taemin Oh ◽  
Matheus P. Pereira ◽  
Rushikesh S. Joshi ◽  
...  

Abstract Cytolytic score (CYT), calculated from mRNA expression levels of granzyme and perforin, positively correlates with CD8+ T cell infiltration/activity in a variety of cancers. Unlike other cancers, higher CYT has been associated with worse prognosis in glioblastoma (GBM). To address this discrepancy, we sought to investigate the relationship between CYT and immune checkpoint gene score (ICGscore), as well as their correlation with patient survival and tumor immune cell infiltration. Clinical and RNA-sequencing data for patients with newly diagnosed GBM were obtained from The Cancer Genome Atlas. Maximally-selected rank statistics was used to dichotomize subgroups. CIBERSORT was used to estimate abudence of immune cell-types. Spearman correlation was used to characterize the relationship between CYT and ICGscore. Kaplan–Meier curves were generated for survival analysis. Overall, 28/151 patients had high CYT. High CYT was associated with a mesenchymal subtype (p < 0.001) and worse survival (7.45 vs. 12.2 months, p < 0.001). There were no differences in patient demographics, IDH/MGMT mutation status, or treatment. On subgroup analysis, patients with high CYT/ICGscore had significantly increased CD8+ infiltration (p < 0.001), as expected, and worse survival (HR 0.445, p < 0.01). Furthermore, CYT strongly correlated with ICGscore (RS = 0.675, p < 0.001). The high CYT/ICGscore subgroup was associated with greater infiltration of M2 macrophages (p = 0.011) and neutrophils (p = 0.055). Our study highlights a multidimensional immunosuppressive GBM microenvironment in patients with higher CYT and potentially identifies patients with high CYT/ICGscore as a subgroup that may particularly benefit from multi-faceted immunotherapies, given their already elevated tumor CD8+ T cell levels.


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.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Maria-Fernanda Senosain ◽  
Yong Zou ◽  
Tatiana Novitskaya ◽  
Georgii Vasiukov ◽  
Aneri B. Balar ◽  
...  

AbstractLung adenocarcinoma (ADC) is a heterogeneous group of tumors associated with different survival rates, even when detected at an early stage. Here, we aim to investigate whether CyTOF identifies cellular and molecular predictors of tumor behavior. We developed and validated a CyTOF panel of 34 antibodies in four ADC cell lines and PBMC. We tested our panel in a set of 10 ADCs, classified into long- (LPS) (n = 4) and short-predicted survival (SPS) (n = 6) based on radiomics features. We identified cellular subpopulations of epithelial cancer cells (ECC) and their microenvironment and validated our results by multiplex immunofluorescence (mIF) applied to a tissue microarray (TMA) of LPS and SPS ADCs. The antibody panel captured the phenotypical differences in ADC cell lines and PBMC. LPS ADCs had a higher proportion of immune cells. ECC clusters (ECCc) were identified and uncovered two ADC groups. ECCc with high HLA-DR expression were correlated with CD4+ and CD8+ T cells, with LPS samples being enriched for those clusters. We confirmed a positive correlation between HLA-DR expression on ECC and T cell number by mIF staining on TMA slides. Spatial analysis demonstrated shorter distances from T cells to the nearest ECC in LPS. Our results demonstrate a distinctive cellular profile of ECC and their microenvironment in ADC. We showed that HLA-DR expression in ECC is correlated with T cell infiltration, and that a set of ADCs with high abundance of HLA-DR+ ECCc and T cells is enriched in LPS samples. This suggests new insights into the role of antigen presenting tumor cells in tumorigenesis.


2020 ◽  
Vol 18 (10) ◽  
pp. 1589-1602 ◽  
Author(s):  
Jing Han ◽  
Zhen Jiang ◽  
Chennan Wang ◽  
Xin Chen ◽  
Rongqing Li ◽  
...  

2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A771-A771
Author(s):  
Audrey Darmon ◽  
Ping Zhang ◽  
Jordan Da silva ◽  
Sebastien Paris

BackgroundWhen exposed to radiotherapy (RT), NBTXR3 nanoparticles increase radiation dose deposition from within the cancer cells. NBTXR3 is intended for a single intratumor injection. Results from a phase II/III clinical trial in patients with locally advanced Soft Tissue Sarcoma demonstrated significant superiority and clinical benefits of NBTXR3 activated by RT compared to RT alone, and was well tolerated. NBTXR3 is currently being evaluated in several other tumors including head and neck, liver, and pancreatic cancer as a single agent or in combination with anti-PD1. Moreover, preclinical studies have demonstrated that NBTXR3 can produce a significant abscopal effect, whereas RT alone cannot. Here, we explored the impact of NBTXR3 activated by RT on CD8+ infiltrates and TcR repertoire diversity change, and the effect on the immunopeptidome of cancer cells.MethodsCT26 (murine colorectal cancer cells) were subcutaneously injected in BALB/c mice in one flank. Then, tumors were intratumorally injected with NBTXR3 (or vehicle) and irradiated 24 hours later with 4Gy per fraction for 3 consecutive days. Tumors were collected 3 days after the last RT fraction and immune cell infiltrates were measured using immunohistochemistry (IHC) and digital pathology. For TcR repertoire sequencing, the same workflow was followed, except cells were injected in both flanks. Only right tumors received treatment, while left tumors remained untreated. For immunopeptidome analysis, in vitro cells were irradiated by 4Gy. After one day, cells were collected for isolation and sequencing of MHC I-loaded peptides.ResultsIHC analyses showed a significant increase of CD8+ T cell infiltrates in tumors of mice treated with NBTXR3+RT, while RT alone had no significant effect. In addition, NBTXR3+RT treatment was able to increase TcR repertoire diversity, both in treated and untreated tumors, compared to RT alone. Finally, analysis of immunopeptidome showed that NBTXR3+RT changed the profile of MHC-I-loaded peptides.ConclusionsOur in vivo data indicate that NBTXR3+RT can modulate the microenvironment of treated tumors, leading to enhanced CD8+ T cell infiltration as well as modification of the TcR repertoire, both in treated and distant untreated tumors. These NBTXR3+RT-induced responses may be related to changes in the immunopeptidome of cancer cells triggered by this treatment.


Sign in / Sign up

Export Citation Format

Share Document