Cancer associated fibroblasts: the major player in pancreatic cancer

2021 ◽  
Author(s):  
Débora B. Vendramini Costa

Tumor cells are not alone in the tumor mass; in fact they are surrounded by a complex and active microenvironment, composed by fibroblasts and their extracellular matrix (ECM), immune cells, nerves, blood vessels, and secreted factors. It is now well recognized that the microenvironment plays an important role in tumor development and thus it is imperative for the comprehensive understanding of the interplay between cancer and its microenviroment for the development of better preventative and therapeutic strategies. Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with a 5 year survival rate of only 10% after diagnosis[1]. One of the main reasons for this outcome is the poor understanding of the unique microenvironment of PDAC, where up to 90% of the tumor mass can be composed by the stroma, with most of it being the expansion of activated fibroblasts and their ECM[2]. Even though CAFs represent an important component of PDAC (and other types of cancers), they are still incompletely understood. This can be partially explained by the fact that there are no specific markers to discriminate CAFs, and researchers need to rely on negative selections, absence of mutations that characterize the transformed epithelial cells plus the presence of mesenchymal markers, all together with the assessment of fibroblastic function2. There are still controversies about the pro- and anti-tumor effects of CAFs and their origin. Recently, a consensus was published2, where authors suggest that CAFs are mostly originated by the local activation and proliferation of resident fibroblasts, stimulated by tissue injury, reactive oxidative species, growth factors and more. These CAFs are characterized by their plasticity, as they can interchange between functions according to the signals of the environment. Many are the functions attributed to these cells; CAFs produce a very dense ECM, which can lead to the collapse of blood vessels, thus affecting nutrient supply and the delivery of therapies to this environment[3]. Moreover, CAFs can be immunusuppressive, producing a millieu of cytokines and chemokines that can turn off anti-tumor immune cells and recruit pro-tumor ones[4]. Interestingly, CAFs can also contribute to the metabolic signature of the environment, as they produce and modify a large range of metabolites, often supporting cancer cell survival[5]. Recently, our research group uncovered the pro-tumor roles of the synaptic protein Netrin G1 in CAFs[6]. The expression of Netrin G1 in CAFs from PDAC patients inversely correlated with overall survival. Moreover, the loss of Netrin G1 in CAFs led to decreased production of immunosuppressive factors, allowing NK cells to kill PDAC cells in vitro. Netrin G1 expression was also important for the production of metabolites (mainly glutamine and glutamate) by CAFs and for the support of nutrient deprived PDAC cells. It is important to mention that the Netrin G1 related studies were perfomed using 3D co-cultures, and that its expression can only be detected in CAFs growing in their 3D environment, further reinforcing the need for better strategies to study and understand the tumor microenvironment in PDAC. Therefore, CAFs can be seen as the major intermediaries of PDAC microenvironment and to target these cells in an attempt to normalize them rather than eliminate them, might be an effective strategy for PDAC therapy. References: [1] Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin, 2020; 70: 7-30. [2] Sahai E et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer, 2020; 20:174-186 [3] Provenzano et al. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell, 2012; 21:418-29. [4] Ziani L, Chouaib S, Thiery J. Alteration of the Antitumor Immune Response by Cancer-Associated Fibroblasts. Front Immunol, 2018; 9:414. [5] Lyssiotis CA and Kimmelman AC. Metabolic interactions in the tumor microenvironment. Trends Cell Biol, 2017; 27: 863-875. [6] Francescone et al. Netrin G1 promotes pancreatic tumorigenesis through cancer associated fibroblast driven nutritional support and immunosuppression. Cancer Discov. 2020 Oct 30:CD-20-0775. Epub ahead of print.

2021 ◽  
Vol 22 (24) ◽  
pp. 13408
Author(s):  
Utpreksha Vaish ◽  
Tejeshwar Jain ◽  
Abhi C. Are ◽  
Vikas Dudeja

Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related morbidity and mortality in the western world, with limited therapeutic strategies and dismal long-term survival. Cancer-associated fibroblasts (CAFs) are key components of the pancreatic tumor microenvironment, maintaining the extracellular matrix, while also being involved in intricate crosstalk with cancer cells and infiltrating immunocytes. Therefore, they are potential targets for developing therapeutic strategies against PDAC. However, recent studies have demonstrated significant heterogeneity in CAFs with respect to their origins, spatial distribution, and functional phenotypes within the PDAC tumor microenvironment. Therefore, it is imperative to understand and delineate this heterogeneity prior to targeting CAFs for PDAC therapy.


Cells ◽  
2021 ◽  
Vol 10 (7) ◽  
pp. 1653
Author(s):  
Ester Pfeifer ◽  
Joy M. Burchell ◽  
Francesco Dazzi ◽  
Debashis Sarker ◽  
Richard Beatson

Pancreatic ductal adenocarcinoma (PDAC) is associated with poor prognosis. This is attributed to the disease already being advanced at presentation and having a particularly aggressive tumor biology. The PDAC tumor microenvironment (TME) is characterized by a dense desmoplastic stroma, dominated by cancer-associated fibroblasts (CAF), extracellular matrix (ECM) and immune cells displaying immunosuppressive phenotypes. Due to the advanced stage at diagnosis, the depletion of immune effector cells and lack of actionable genomic targets, the standard treatment is still apoptosis-inducing regimens such as chemotherapy. Paradoxically, it has emerged that the direct induction of apoptosis of cancer cells may fuel oncogenic processes in the TME, including education of CAF and immune cells towards pro-tumorigenic phenotypes. The direct effect of cytotoxic therapies on CAF may also enhance tumorigenesis. With the awareness that CAF are the predominant cell type in PDAC driving tumorigenesis with various tumor supportive functions, efforts have been made to try to target them. However, efforts to target CAF have, to date, shown disappointing results in clinical trials. With the help of sophisticated single cell analyses it is now appreciated that CAF in PDAC are a heterogenous population with both tumor supportive and tumor suppressive functions. Hence, there remains a debate whether targeting CAF in PDAC is a valid therapeutic strategy. In this review we discuss how cytotoxic therapies and the induction of apoptosis in PDAC fuels oncogenesis by the education of surrounding stromal cells, with a particular focus on the potential pro-tumorigenic outcomes arising from targeting CAF. In addition, we explore therapeutic avenues to potentially avoid the oncogenic effects of apoptosis in PDAC CAF.


Cancers ◽  
2021 ◽  
Vol 13 (12) ◽  
pp. 2995
Author(s):  
Laia Gorchs ◽  
Helen Kaipe

Less than 10% of patients diagnosed with pancreatic ductal adenocarcinoma (PDAC) survive 5 years or more, making it one of the most fatal cancers. Accumulation of T cells in pancreatic tumors is associated with better prognosis, but immunotherapies to enhance the anti-tumor activity of infiltrating T cells are failing in this devastating disease. Pancreatic tumors are characterized by a desmoplastic stroma, which mainly consists of activated cancer-associated fibroblasts (CAFs). Pancreatic CAFs have emerged as important regulators of the tumor microenvironment by contributing to immune evasion through the release of chemokines, cytokines, and growth factors, which alters T-cell migration, differentiation and cytotoxic activity. However, recent discoveries have also revealed that subsets of CAFs with diverse functions can either restrain or promote tumor progression. Here, we discuss our current knowledge about the interactions between CAFs and T cells in PDAC and summarize different therapy strategies targeting the CAF–T cell axis with focus on CAF-derived soluble immunosuppressive factors and chemokines. Identifying the functions of different CAF subsets and understanding their roles in T-cell trafficking within the tumor may be fundamental for the development of an effective combinational treatment for PDAC.


2020 ◽  
Vol 19 ◽  
pp. 153303382092096
Author(s):  
Hongzhi Sun ◽  
Bo Zhang ◽  
Haijun Li

Pancreatic ductal adenocarcinoma has extremely high malignancy and patients with pancreatic ductal adenocarcinoma have dismal prognosis. The failure of pancreatic ductal adenocarcinoma treatment is largely due to the tumor microenvironment, which is featured by ample stromal cells and complicated extracellular matrix. Recent genomic analysis revealed that pancreatic ductal adenocarcinoma harbors frequently mutated genes including KRAS, TP53, CDKN2A, and SMAD4, which can widely alter cellular processes and behaviors. As shown by accumulating studies, these mutant genes may also change tumor microenvironment, which in turn affects pancreatic ductal adenocarcinoma progression. In this review, we summarize the role of such genetic mutations in tumor microenvironment regulation and potential mechanisms.


2011 ◽  
Vol 17 (22) ◽  
pp. 7015-7023 ◽  
Author(s):  
David Z. Chang ◽  
Ying Ma ◽  
Baoan Ji ◽  
Huamin Wang ◽  
Defeng Deng ◽  
...  

Author(s):  
Xuefei Liu ◽  
Ziwei Luo ◽  
Xuechen Ren ◽  
Zhihang Chen ◽  
Xiaoqiong Bao ◽  
...  

Background: Pancreatic ductal adenocarcinoma (PDAC) is dominated by an immunosuppressive microenvironment, which makes immune checkpoint blockade (ICB) often non-responsive. Understanding the mechanisms by which PDAC forms an immunosuppressive microenvironment is important for the development of new effective immunotherapy strategies.Methods: This study comprehensively evaluated the cell-cell communications between malignant cells and immune cells by integrative analyses of single-cell RNA sequencing data and bulk RNA sequencing data of PDAC. A Malignant-Immune cell crosstalk (MIT) score was constructed to predict survival and therapy response in PDAC patients. Immunological characteristics, enriched pathways, and mutations were evaluated in high- and low MIT groups.Results: We found that PDAC had high level of immune cell infiltrations, mainly were tumor-promoting immune cells. Frequent communication between malignant cells and tumor-promoting immune cells were observed. 15 ligand-receptor pairs between malignant cells and tumor-promoting immune cells were identified. We selected genes highly expressed on malignant cells to construct a Malignant-Immune Crosstalk (MIT) score. MIT score was positively correlated with tumor-promoting immune infiltrations. PDAC patients with high MIT score usually had a worse response to immune checkpoint blockade (ICB) immunotherapy.Conclusion: The ligand-receptor pairs identified in this study may provide potential targets for the development of new immunotherapy strategy. MIT score was established to measure tumor-promoting immunocyte infiltration. It can serve as a prognostic indicator for long-term survival of PDAC, and a predictor to ICB immunotherapy response.


2020 ◽  
Author(s):  
S. Mahnaz ◽  
L. Das Roy ◽  
M. Bose ◽  
C. De ◽  
S. Nath ◽  
...  

ABSTRACTMyeloid-derived suppressor cells (MDSCs) are immature myeloid cells that are responsible for immunosuppression in tumor microenvironment. Here we report the impact of mucin 1 (MUC1), a transmembrane glycoprotein, on proliferation and functional activity of MDSCs. To determine the role of MUC1 in MDSC phenotype, we analyzed MDSCs derived from wild type (WT) and MUC1-knockout (MUC1KO) mice bearing pancreatic ductal adenocarcinoma KCKO and breast cancer C57MG xenografts. We observed enhanced tumor growth in MUC1KO mice compared to WT mice in both pancreatic KCKO and breast C57MG cancer models due to increased MDSC population and enrichment of Tregs in tumor microenvironment. Our current study shows that knockdown of MUC1 in MDSCs promotes proliferation and immature suppressive phenotype indicated by increased level of iNOS, ARG1 activity and TGF-β secretion under cancer conditions. Increased activity of MDSCs leads to repression of IL-2 and IFN-ɣ production by T-cells. We were able to find that MDSCs from MUC1KO mice have higher levels of c-Myc and activated pSTAT3 as compared to MUC1 WT mice, that are signaling pathways leading to increased survival, proliferation and prevention of maturation. In summary, MUC1 regulates signaling pathways that maintain immunosuppressive properties of MDSCs. Thus, immunotherapy must target only tumor associated MUC1 on epithelial cells and not MUC1 on hematopoietic cells to avoid expansion and suppressive functions of MDSC.


Cancers ◽  
2021 ◽  
Vol 13 (21) ◽  
pp. 5447
Author(s):  
Maria Iorio ◽  
Nikkitha Umesh Ganesh ◽  
Monica De Luise ◽  
Anna Maria Porcelli ◽  
Giuseppe Gasparre ◽  
...  

Metabolic reprogramming is a well-known hallmark of cancer, whereby the development of drugs that target cancer cell metabolism is gaining momentum. However, when establishing preclinical studies and clinical trials, it is often neglected that a tumor mass is a complex system in which cancer cells coexist and interact with several types of microenvironment populations, including endothelial cells, fibroblasts and immune cells. We are just starting to understand how such populations are affected by the metabolic changes occurring in a transformed cell and little is known


Sign in / Sign up

Export Citation Format

Share Document