The ability of disseminated cancer cells to evade the immune response is a critical step for efficient metastatic progression. Protection against an immune attack is often provided by the tumor microenvironment that suppresses and excludes cytotoxic CD8 T cells. Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive metastatic disease with unmet needs, yet the immunoprotective role of the metastatic tumor microenvironment in pancreatic cancer is not completely understood. In this study, we find that macrophage-derived granulin contributes to cytotoxic CD8 T-cell exclusion in metastatic livers. Granulin expression by macrophages was induced in response to colony-stimulating factor 1. Genetic depletion of granulin reduced the formation of a fibrotic stroma, thereby allowing T-cell entry at the metastatic site. Although metastatic PDAC tumors are largely resistant to anti-PD-1 therapy, blockade of PD-1 in granulin-depleted tumors restored the antitumor immune defense and dramatically decreased metastatic tumor burden. These findings suggest that targeting granulin may serve as a potential therapeutic strategy to restore CD8 T-cell infiltration in metastatic PDAC, thereby converting PDAC metastatic tumors, which are refractory to immune checkpoint inhibitors, into tumors that respond to immune checkpoint inhibition therapies. These findings uncover a mechanism by which metastatic PDAC tumors evade the immune response and provide the rationale for targeting granulin in combination with immune checkpoint inhibitors for the treatment of metastatic PDAC. http://cancerres.aacrjournals.org/content/canres/78/15/4253/F1.large.jpg .

Download full-text PDF

Source
http://www.ncbi.nlm.nih.gov/pmc/articles/PMC6076440PMC
http://dx.doi.org/10.1158/0008-5472.CAN-17-3876DOI Listing

Publication Analysis

Top Keywords

immune checkpoint
16
metastatic pdac
16
metastatic
12
macrophage-derived granulin
8
immune
8
checkpoint inhibition
8
pancreatic cancer
8
evade immune
8
immune response
8
tumor microenvironment
8

Similar Publications

Optimizing T cell inflamed signature through a combination biomarker approach for predicting immunotherapy response in NSCLC.

Sci Rep

December 2024

Interventional Oncology, Johnson & Johnson Enterprise Innovation, Inc, 10th Floor 255 Main St, 02142, Cambridge, Boston, MA, USA.

The introduction of anti-PD-1/PD-L1 therapies revolutionized treatment for advanced non-small cell lung cancer (NSCLC), yet response rates remain modest, underscoring the need for predictive biomarkers. While a T cell inflamed gene expression profile (GEP) has predicted anti-PD-1 response in various cancers, it failed in a large NSCLC cohort from the Stand Up To Cancer-Mark (SU2C-MARK) Foundation. Re-analysis revealed that while the T cell inflamed GEP alone was not predictive, its performance improved significantly when combined with gene signatures of myeloid cell markers.

View Article and Find Full Text PDF

LAG3 plays a regulatory role in immunity and emerged as an inhibitory immune checkpoint molecule comparable to PD-L1 and CTLA-4 and a potential target for enhancing anti-cancer immune responses. We generated 3D cancer cultures as a model to identify novel molecular biomarkers for the selection of patients suitable for α-LAG3 treatment and simultaneously the possibility to perform an early diagnosis due to its higher presence in breast cancer, also to achieve a theragnostic approach. Our data confirm the extreme dysregulation of LAG3 in breast cancer with significantly higher expression in tumor tissue specimens, compared to non-cancerous tissue controls.

View Article and Find Full Text PDF

In recent years, immune checkpoint inhibitors (ICIs) has emerged as a fundamental component of the standard treatment regimen for patients with head and neck squamous cell carcinoma (HNSCC). However, accurately predicting the treatment effectiveness of ICIs for patients at the same TNM stage remains a challenge. In this study, we first combined multi-omics data (mRNA, lncRNA, miRNA, DNA methylation, and somatic mutations) and 10 clustering algorithms, successfully identifying two distinct cancer subtypes (CSs) (CS1 and CS2).

View Article and Find Full Text PDF

Expression and relationship of PD-L1, CD24, and CD47 in hepatitis B virus associated hepatocellular carcinoma.

Sci Rep

December 2024

Department of Gastroenterology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, Fujian, China.

Immune checkpoint inhibitor (ICI) therapy is the new standard treatment for advanced or metastatic hepatocellular carcinoma (HCC); however, many patients still fail to respond. This study explored the expression and prognosis of programmed death ligand 1 (PD-L1), cluster of differentiation 24 (CD24), and cluster of differentiation 47 (CD47) in patients with hepatitis B virus-associated HCC (HBV-associated HCC). We analyzed sequencing data from the Cancer Genome Atlas (TCGA) and investigated the expression of PD-L1, CD24, and CD47 in HBV-associated HCC patients by immunohistochemistry and their relationship with prognosis and clinicopathological factors.

View Article and Find Full Text PDF

This study aims to assess the predictive value of certain markers of inflammation in patients with locally advanced or recurrent/metastatic cervical cancer who are undergoing treatment with anti-programmed death 1 (PD-1) therapy. A total of 105 patients with cervical cancer, who received treatment involving immunocheckpoint inhibitors (ICIs), were included in this retrospective study. We collected information on various peripheral blood indices, including the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), lymphocyte-to-monocyte ratio (LMR), systemic immune-inflammation index (SII), and prognostic nutritional index (PNI).

View Article and Find Full Text PDF

Want AI Summaries of new PubMed Abstracts delivered to your In-box?

Enter search terms and have AI summaries delivered each week - change queries or unsubscribe any time!