1
|
Dreyer SB, Beer P, Hingorani SR, Biankin AV. Improving outcomes of patients with pancreatic cancer. Nat Rev Clin Oncol 2025; 22:439-456. [PMID: 40329051 DOI: 10.1038/s41571-025-01019-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2025] [Indexed: 05/08/2025]
Abstract
Research studies aimed at improving the outcomes of patients with pancreatic ductal adenocarcinoma (PDAC) have brought about limited progress, and in clinical practice, the optimized use of surgery, chemotherapy and supportive care have led to modest improvements in survival that have probably reached a plateau. As a result, PDAC is expected to be the second leading cause of cancer-related death in Western societies within a decade. The development of therapeutic advances in PDAC has been challenging owing to a lack of actionable molecular targets, a typically immunosuppressive microenvironment, and a disease course characterized by rapid progression and clinical deterioration. Yet, the progress in our understanding of PDAC and identification of novel therapeutic opportunities over the past few years is leading to a strong sense of optimism in the field. In this Perspective, we address the aforementioned challenges, including biological aspects of PDAC that make this malignancy particularly difficult to treat. We explore specific areas with potential for therapeutic advances, including targeting mutant KRAS, novel strategies to harness the antitumour immune response and approaches to early detection, and propose mechanisms to improve clinical trial design and to overcome various community and institutional barriers to progress.
Collapse
Affiliation(s)
- Stephan B Dreyer
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK
- West of Scotland Hepato-Biliary and Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
- Department of Hepatobiliary Surgery, Royal Liverpool University Hospital, Liverpool, UK
| | - Philip Beer
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK
- Hull York Medical School, University of York, York, UK
| | - Sunil R Hingorani
- Department of Internal Medicine, Division of Hemotology/Oncology, University of Nebraska Medical Center, Omaha, NE, USA
- Pancreatic Cancer Center of Excellence, University of Nebraska Medical Center, Omaha, NE, USA
| | - Andrew V Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK.
- West of Scotland Hepato-Biliary and Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK.
| |
Collapse
|
2
|
Mitra P, Saha U, Stephen KJ, Prasad P, Jena S, Patel AK, Bv H, Mondal SK, Kurkalang S, Roy S, Ghosh A, Roy SS, Das Sarma J, Biswas NK, Acharya M, Sharan R, Arun P, Jolly MK, Maitra A, Singh S. Tie2 activity in cancer associated myofibroblasts serves as novel target against reprogramming of cancer cells to embryonic-like cell state and associated poor prognosis in oral carcinoma patients. J Exp Clin Cancer Res 2025; 44:142. [PMID: 40349056 PMCID: PMC12065280 DOI: 10.1186/s13046-025-03405-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Myofibroblastic cancer-associated fibroblasts (CAF) in tumor stroma serves as an independent poor prognostic indicator, supporting higher stemness in oral cancer; however, the underlying biology is not fully comprehended. Here, we have explored the crucial role of Tunica Interna Endothelial Cell Kinase (Tie2/TEK) signaling in transition and maintenance of myofibroblastic phenotype of CAFs, and as possible link with the poor prognosis of head and neck squamous cell carcinoma (HNSCC) patients. METHODS Bulk and single cell RNA-sequencing (scRNAseq) methods and in-depth bioinformatic analysis were applied for CAF and cancer cells co-culture for studying molecular relationships. In vitro 3D-spheroid-forming ability, expression of stemness markers, in vivo tumor formation ability in zebrafish embryo and syngeneic mouse allografts formation was conducted to test stemness, upon targeting CAF-specific Tie2 activity by gene silencing or with small molecule inhibitor. Immunohistochemistry analysis was performed to locate the distribution of Tie2 and αSMA in primary tumors of oral carcinoma. Prognosis in HNSCC patient cohort from The Cancer Genome Atlas (TCGA) study was analysed based on single sample gene set enrichment score (ssGSEA) and Kaplan-Meier analysis. RESULTS Autocrine or exogenous TGFβ-induction in CAF led to the recruitment of histone deacetylase 2 (HDAC2) on the promoter of Tie2-antagonist, Angiopoietin-2 (ANGPT2), resulting in its downregulation, leading to phosphorylation of Tie2 (Y992) and subsequent activation of SRC (Y418). This led to SRC/ROCK mediated αSMA-positive stress-fiber formation with gain of myofibroblast phenotype. The CAF-specific Tie2-signaling was responsible for producing embryonic-like cell state in co-cultured cancer cells; with enhanced tumor initiating ability. Tie2 activity in CAF exerted the dynamic gene expression reprogramming, with the upregulation of 'cell migration' and downregulation of 'protein biosynthesis' related gene-regulatory-network modules in malignant cells. The AUCell scores calculated for gene signatures derived from these modules showed significant concordance in independently reported scRNAseq studies of HNSCC tumors and significant association with poor prognosis in HNSCC patient cohort. CONCLUSIONS CAF-specific Tie2 activity may serve as direct stromal-target against cancer cell plasticity leading to poor prognosis of oral cancer patients. Overall, our work has provided wider applicability of Tie2-specific functions in tumor biology, along with its known role in endothelial cell-specific function.
Collapse
Affiliation(s)
- Paromita Mitra
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Uday Saha
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Regional Centre for Biotechnology, Faridabad, India
| | | | - Priyanka Prasad
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
| | - Subhashree Jena
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Ankit Kumar Patel
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Umea University, Umea, Sweden
| | | | | | - Sillarine Kurkalang
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Comprehensive Cancer Center, University of Chicago Medicine, Chicago, IL, USA
| | - Sumitava Roy
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Arnab Ghosh
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Shantanu Saha Roy
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
| | | | - Nidhan Kumar Biswas
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Moulinath Acharya
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Regional Centre for Biotechnology, Faridabad, India
| | | | | | | | - Arindam Maitra
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Sandeep Singh
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India.
- Regional Centre for Biotechnology, Faridabad, India.
| |
Collapse
|
3
|
Qian G, Zhang H, Liu Y, Shribak M, Eliceiri KW, Provenzano PP. Computationally Enabled Polychromatic Polarized Imaging Enables Mapping of Matrix Architectures that Promote Pancreatic Ductal Adenocarcinoma Dissemination. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00160-9. [PMID: 40350060 DOI: 10.1016/j.ajpath.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 02/22/2025] [Accepted: 04/08/2025] [Indexed: 05/14/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is an extremely metastatic and lethal disease. In PDA, extracellular matrix (ECM) architectures, known as tumor-associated collagen signatures (TACSs), regulate invasion and metastatic spread in both early dissemination and late-stage disease. As such, TACS has been suggested as a biomarker to aid in pathologic assessment. However, despite its significance, approaches to quantitatively capture these ECM patterns currently require advanced optical systems with signaling processing analysis. Here, we present an expansion of polychromatic polarized microscopy (PPM) with inherent angular information coupled to machine learning and computational pixel-wise analysis of TACS. Using this platform, we are able to accurately capture TACS architectures in hematoxylin and eosin-stained histology sections directly through PPM contrast. Moreover, PPM facilitated identification of transitions to dissemination architectures (ie, transitions from sequestration through expansion to dissemination from both PanINs and throughout PDA). Last, PPM evaluation of architectures in liver metastases, the most common metastatic site for PDA, demonstrates TACS-mediated focal and local invasion as well as identification of unique patterns anchoring aligned fibers into normal-adjacent tumor, suggesting that these patterns may be precursors to metastasis expansion and local spread from micrometastatic lesions. Combined, these findings demonstrate that PPM coupled to computational platforms is a powerful tool for analyzing ECM architecture that can be used to advance cancer microenvironment studies and provide clinically relevant diagnostic information.
Collapse
Affiliation(s)
- Guhan Qian
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota; Center for Multiparametric Imaging of Tumor Immune Microenvironments, University of Minnesota and University of Wisconsin-Madison
| | - Hongrong Zhang
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota; Center for Multiparametric Imaging of Tumor Immune Microenvironments, University of Minnesota and University of Wisconsin-Madison
| | - Yuming Liu
- Center for Multiparametric Imaging of Tumor Immune Microenvironments, University of Minnesota and University of Wisconsin-Madison; Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, Wisconsin
| | - Michael Shribak
- Marine Biological Laboratory, University of Chicago, Woods Hole, Massachusetts
| | - Kevin W Eliceiri
- Center for Multiparametric Imaging of Tumor Immune Microenvironments, University of Minnesota and University of Wisconsin-Madison; Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, Wisconsin; Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Paolo P Provenzano
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota; Center for Multiparametric Imaging of Tumor Immune Microenvironments, University of Minnesota and University of Wisconsin-Madison; Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota; Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Institute for Engineering in Medicine, University of Minnesota, Minneapolis, Minnesota; Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
4
|
Qian G, Provenzano PP. Cellular teamwork in cancer invasion. Biophys J 2025; 124:1323-1324. [PMID: 40017031 DOI: 10.1016/j.bpj.2025.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/01/2025] Open
Affiliation(s)
- Guhan Qian
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota; Center for Multiparametric Imaging of Tumor Immune Microenvironments, University of Minnesota, Minneapolis, Minnesota
| | - Paolo P Provenzano
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota; Center for Multiparametric Imaging of Tumor Immune Microenvironments, University of Minnesota, Minneapolis, Minnesota; Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota; Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
5
|
Jia X, Zhang Q, Wang Z, Cao J, Song A, Lan C, Hu Y. Integrative single-cell RNA sequencing and bulk RNA sequencing reveals the characteristics of glutathione metabolism and protective role of GSTA4 gene in pancreatic cancer. Front Immunol 2025; 16:1571431. [PMID: 40375987 PMCID: PMC12078301 DOI: 10.3389/fimmu.2025.1571431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/07/2025] [Indexed: 05/18/2025] Open
Abstract
Background Recent studies have increasingly reported abnormal glutathione (GSH) metabolism within the tumor microenvironment across various solid tumors. However, the specific mechanisms underlying aberrant GSH metabolism in pancreatic cancer (PC) remain unclear. This study aims to investigate the prognostic significance of GSH metabolism-related genes in PC and to identify key molecular targets, thereby providing novel perspectives for targeted PC therapy. Methods The GSH metabolism gene set was retrieved from the KEGG database. Utilizing single-cell transcriptomic data from the GSE205049 dataset, this study analyzed the variation in GSH metabolic signaling intensity across distinct cell types within the tumor microenvironment of PC. Additionally, transcriptomic data from multiple repositories, including TCGA, ICGC, and GEO, comprising a total of 930 patients with PC, were integrated to construct a prognostic molecular classifier related to GSH metabolism. Furthermore, the role of the key gene GSTA4 in PC was experimentally validated through a series of in vitro assays. Results Significant differences in GSH metabolic signaling intensity were observed across various cell types in both normal pancreatic and PC tissues. A prognostic signature comprising six GSH metabolism-related genes (GSTA5, PGD, IDH2, GSTA4, GPX2, and GPX3) was established, wherein a high-risk score was associated with a poorer patient prognosis. Notably, GSTA4 expression was significantly reduced in PC tissues, and higher GSTA4 levels were linked to a favorable prognosis. In vitro functional analyses demonstrated that GSTA4 overexpression markedly inhibited PC cell proliferation and migration. Conclusion The GSH metabolism-associated prognostic signature developed in this study effectively identifies high-risk patients with PC. As a prognostic protective factor, GSTA4 exhibits downregulated expression in PC tissues and suppresses tumor proliferation and migration, highlighting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Xinya Jia
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiang Zhang
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhe Wang
- Department of Urology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianliang Cao
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Anran Song
- Department of Physical Diagnosis, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chao Lan
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuepeng Hu
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
6
|
Seidel T, Ohri N, Glaß M, Sunami Y, Müller LP, Kleeff J. Stromal Cells in Early Inflammation-Related Pancreatic Carcinogenesis-Biology and Its Potential Role in Therapeutic Targeting. Cancers (Basel) 2025; 17:1541. [PMID: 40361466 DOI: 10.3390/cancers17091541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 04/28/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
The stroma of healthy pancreases contains various non-hematopoietic, non-endothelial mesenchymal cells. It is altered by chronic inflammation which in turn is a major contributor to the development of pancreatic adenocarcinoma (PDAC). In PDAC, the stroma plays a decisive and well-investigated role for tumor progression and therapy response. This review addresses the central role of stromal cells in the early inflammation-driven development of PDAC. It focuses on major subpopulations of pancreatic mesenchymal cells, i.e., fibroblasts, pancreatic stellate cells, and multipotent stroma cells, particularly their activation and functional alterations upon chronic inflammation including the development of different types of carcinoma-associated fibroblasts. In the second part, the current knowledge on the impact of activated stroma cells on acinar-to-ductal metaplasia and the transition to pancreatic intraepithelial neoplasia is summarized. Finally, putative strategies to target stroma cells and their signaling in early pancreatic carcinogenesis are reflected. In summary, the current data show that the activation of pancreatic stroma cells and the resulting fibrotic changes has pro- and anti-carcinogenetic effects but, overall, creates a carcinogenesis-promoting microenvironment. However, this is a dynamic process and the therapeutic targeting of specific pathways and cells requires in-depth knowledge of the molecular interplay of various cell types.
Collapse
Affiliation(s)
- Tina Seidel
- Department of Internal Medicine, University Hospital Halle, 06120 Halle (Saale), Germany
| | - Nupur Ohri
- Department of Visceral, Vascular and Endocrine Surgery, University Hospital Halle, 06120 Halle (Saale), Germany
| | - Markus Glaß
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany
| | - Yoshiaki Sunami
- Department of Visceral, Vascular and Endocrine Surgery, University Hospital Halle, 06120 Halle (Saale), Germany
| | - Lutz P Müller
- Department of Internal Medicine, University Hospital Halle, 06120 Halle (Saale), Germany
| | - Jörg Kleeff
- Department of Visceral, Vascular and Endocrine Surgery, University Hospital Halle, 06120 Halle (Saale), Germany
| |
Collapse
|
7
|
Lin H, Zhu S, Chen Y, Lu J, Xie C, Liao C, Huang X, Li G, Wu Y, Li Z, Hu J, Lin X, Tian Y, Li Q, Wang Z, Chen S. Targeting cTRIP12 counteracts ferroptosis resistance and augments sensitivity to immunotherapy in pancreatic cancer. Drug Resist Updat 2025; 81:101240. [PMID: 40154160 DOI: 10.1016/j.drup.2025.101240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 04/01/2025]
Abstract
AIMS Current therapeutic strategies for pancreatic ductal adenocarcinoma (PDAC) have limited efficacy in increasing patient survival rates, largely due to ferroptosis resistance and immunosuppression. The aim of this study is to identify molecular mechanisms associated with ferroptosis resistance and immunosuppression in PDAC tumour cells. METHODS Circular RNA sequencing (circRNA-seq) was performed on clinical samples to identify potential circRNAs that mediate ferroptosis resistance. C11-BODIPY staining, FerroOrange staining, the glutathione ratio, malondialdehyde quantification, and transmission electron microscopy were employed to assess ferroptosis. RNA pulldown, mass spectrometry, RNA immunoprecipitation, and coimmunoprecipitation assays were conducted to investigate the molecular mechanisms involved. A HuNSG mouse xenograft tumour model was utilized to validate therapeutic agents. RESULTS A circRNA derived from TRIP12 (cTRIP12) was identified in PDAC samples resistant to ferroptosis. cTRIP12 knockdown increased the sensitivity of PDAC cells to ferroptosis and immunotherapy. Subsequent mechanistic studies revealed that cTRIP12 specifically binds to the O-linked N-acetylglucosamine transferase (OGT) protein and increases intracellular O-GlcNAcylation levels, leading to increased protein levels of ferritin heavy chain (FTH) and PD-L1 in tumour cells. Notably, high cTRIP12 expression suppressed ferroptosis sensitivity and increased immune resistance in PDAC cells by functioning as a protein scaffold through its interaction with OGT and protein kinase R-like endoplasmic reticulum kinase (PERK). cTRIP12 inhibition induced ferroptosis in PDAC cells by reducing FTH and PD-L1 expression and synergistically increased the immunotherapy efficacy. In vivo animal experiments confirmed that the triple therapy consisting of GSK2656157, erastin, and anti-CTLA-4 effectively suppressed the progression of PDAC in tumours with high cTRIP12 expression. CONCLUSION We elucidated the molecular mechanisms underlying the simultaneous occurrence of ferroptosis resistance and immune suppression in PDAC patients. Our study provides a novel therapeutic strategy that could promote ferroptosis in tumour cells and increase immunotherapy efficacy.
Collapse
Affiliation(s)
- Hongyi Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Shuncang Zhu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Yinhao Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Jinpeng Lu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Chengke Xie
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Chengyu Liao
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China; Fuzhou University, Fuzhou 350001, China
| | - Xiaoxiao Huang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China; Fuzhou University, Fuzhou 350001, China
| | - Ge Li
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Yongding Wu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Zhiyuan Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Jianfei Hu
- Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | | | - Yifeng Tian
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China; Fuzhou University, Fuzhou 350001, China
| | - Qiaowei Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Fuzhou University, Fuzhou 350001, China; Fujian Provincial Institute of Clinical Geriatrics, Fuzhou 350001, China; Fujian Key Laboratory of Geriatrics, Fuzhou 350001, China; Fujian Provincial Center for Geriatrics, Fuzhou 350001, China.
| | - Zuwei Wang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China; Fuzhou University, Fuzhou 350001, China.
| | - Shi Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China; Fuzhou University, Fuzhou 350001, China; Fujian Provincial Institute of Clinical Geriatrics, Fuzhou 350001, China; Fujian Key Laboratory of Geriatrics, Fuzhou 350001, China; Fujian Provincial Center for Geriatrics, Fuzhou 350001, China.
| |
Collapse
|
8
|
Liu YT, Liu HM, Ren JG, Zhang W, Wang XX, Yu ZL, Fu QY, Xiong XP, Jia J, Liu B, Chen G. Immune-featured stromal niches associate with response to neoadjuvant immunotherapy in oral squamous cell carcinoma. Cell Rep Med 2025; 6:102024. [PMID: 40107247 PMCID: PMC11970382 DOI: 10.1016/j.xcrm.2025.102024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 11/19/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025]
Abstract
Tumor stromal cells (TSCs) play a crucial yet underexplored role in the tumor microenvironment (TME). This study uses single-cell sequencing and spatial transcriptomics on paired tumor specimens from 22 patients with oral squamous cell carcinoma (OSCC) enrolled in a randomized two-arm phase 2 trial, receiving neoadjuvant anti-PD-1 mono-immunotherapy or anti-PD-1 plus docetaxel-cisplatin-5-fluorouracil (TPF) immunochemotherapy. Single-cell analysis reveals increased TSCs within the TME of responders in immunochemotherapy. Notably, significant post-treatment upregulation of SELP+ high endothelial venules (HEVs) and APOD+ myofibroblastic cancer-associated fibroblasts (myCAFs), alongside a decline in STMN1+ capillary endothelial cells (cECs), is specific to the immunochemotherapy cohort. In contrast, MYF5+ muscle satellite cells (MSCs) are upregulated in non-responders to mono-immunotherapy. SELP+ HEVs and APOD+ myCAFs foster favorable immunomodulatory stromal niches for improved outcomes, while STMN1+ cECs and MYF5+ MSCs form immunosuppressive niches in tumor invasion regions, highlighting therapeutic targets. The trial was registered at ClinicalTrials.gov, and the registration number is NCT04649476.
Collapse
Affiliation(s)
- Yu-Tong Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hai-Ming Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jian-Gang Ren
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wei Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xin-Xin Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zi-Li Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Qiu-Yun Fu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xue-Peng Xiong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jun Jia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Gang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China.
| |
Collapse
|
9
|
Cai Z, Li Z, Zhong W, Lin F, Dong X, Ye H, Guo Y, Chen G, Yu X, Yu H, Tang R, Liu X. Targeting Mesothelin Enhances Personalized Neoantigen Vaccine Induced Antitumor Immune Response in Orthotopic Pancreatic Cancer Mouse Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407976. [PMID: 39887656 PMCID: PMC11948035 DOI: 10.1002/advs.202407976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 12/25/2024] [Indexed: 02/01/2025]
Abstract
The immunosuppressive microenvironment in pancreatic cancer, characterized by low tumor-specific T cells and excessive fibrosis, limits the effectiveness of immunotherapy. Here, three datasets and multi-immunofluorescence staining of tissue microarrays in pancreatic cancer indicate that mesothelin (MSLN) expression negatively correlates with cytotoxic T cells in tumor. Anti-MSLN antibody (αMSLN) treatment of pancreatic cancer in vivo can significantly increase T cell infiltration. Meanwhile, the combination of αMSLN and neoantigen peptide vaccine identified from pancreatic cancer cell lines is demonstrated to be more effective in inducing neoantigen-specific T cell generation and infiltration at subcutaneous and orthotopic pancreatic cancer models for enhancing antitumor efficacy. Single-cell transcriptome analysis shows that the combined treatment significantly reduces the proportion of fibroblasts, improves the infiltration of IFN-γ+CD4+ and GZMK+CD8+ T cells, as well as reduces the interaction of antigen presentation-associated ligands and receptors between antigen-presenting Cancer-Associated Fibroblasts (apCAFs) and naive CD4+ T cells. The negative correlations between apCAFs and CD8+ T cells/IFN-γ+CD4+ T cells are further confirmed in human pancreatic cancer tissues. Overall, this study demonstrates that targeting MSLN can improve neoantigen vaccine induced immune efficacy by reducing apCAFs to interrupt the conversion of naive CD4+ T cells to Tregs, and therefore increase the infiltration of tumor-specific T cells.
Collapse
Affiliation(s)
- Zhixiong Cai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou 350025P. R. China
- The Liver Center of Fujian ProvinceFujian Medical UniversityFuzhou 350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou 350116P. R. China
| | - Zhenli Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou 350025P. R. China
- The Liver Center of Fujian ProvinceFujian Medical UniversityFuzhou 350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou 350116P. R. China
| | - Wenting Zhong
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou 350025P. R. China
- The Liver Center of Fujian ProvinceFujian Medical UniversityFuzhou 350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou 350116P. R. China
| | - Fangzhou Lin
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou 350025P. R. China
- The Liver Center of Fujian ProvinceFujian Medical UniversityFuzhou 350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou 350116P. R. China
| | - Xiuqing Dong
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou 350025P. R. China
- The Liver Center of Fujian ProvinceFujian Medical UniversityFuzhou 350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou 350116P. R. China
| | - Honghao Ye
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou 350025P. R. China
- The Liver Center of Fujian ProvinceFujian Medical UniversityFuzhou 350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou 350116P. R. China
| | - Yutong Guo
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou 350025P. R. China
- The Liver Center of Fujian ProvinceFujian Medical UniversityFuzhou 350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou 350116P. R. China
| | - Geng Chen
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou 350025P. R. China
- The Liver Center of Fujian ProvinceFujian Medical UniversityFuzhou 350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou 350116P. R. China
| | - Xiaoling Yu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou 350025P. R. China
- The Liver Center of Fujian ProvinceFujian Medical UniversityFuzhou 350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou 350116P. R. China
| | - Haijun Yu
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai 201203P. R. China
| | - Ruijing Tang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou 350025P. R. China
- The Liver Center of Fujian ProvinceFujian Medical UniversityFuzhou 350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou 350116P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou 350025P. R. China
- The Liver Center of Fujian ProvinceFujian Medical UniversityFuzhou 350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou 350116P. R. China
| |
Collapse
|
10
|
Loveless IM, Kemp SB, Hartway KM, Mitchell JT, Wu Y, Zwernik SD, Salas-Escabillas DJ, Brender S, George M, Makinwa Y, Stockdale T, Gartrelle K, Reddy RG, Long DW, Wombwell A, Clark JM, Levin AM, Kwon D, Huang L, Francescone R, Vendramini-Costa DB, Stanger BZ, Alessio A, Waters AM, Cui Y, Fertig EJ, Kagohara LT, Theisen B, Crawford HC, Steele NG. Human Pancreatic Cancer Single-Cell Atlas Reveals Association of CXCL10+ Fibroblasts and Basal Subtype Tumor Cells. Clin Cancer Res 2025; 31:756-772. [PMID: 39636224 PMCID: PMC11831110 DOI: 10.1158/1078-0432.ccr-24-2183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/07/2024] [Accepted: 12/03/2024] [Indexed: 12/07/2024]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) patients with tumors enriched for the basal-like molecular subtype exhibit enhanced resistance to standard-of-care treatments and have significantly worse overall survival compared with patients with classic subtype-enriched tumors. It is important to develop genomic resources, enabling identification of novel putative targets in a statistically rigorous manner. EXPERIMENTAL DESIGN We compiled a single-cell RNA sequencing (scRNA-seq) atlas of the human pancreas with 229 patient samples aggregated from publicly available raw data. We mapped cell type-specific scRNA-seq gene signatures in bulk RNA-seq (n = 744) and spatial transcriptomics (ST; n = 22) and performed validation using multiplex immunostaining. RESULTS Analysis of tumor cells from our scRNA-seq atlas revealed nine distinct populations, two of which aligned with the basal subtype, correlating with worse overall survival in bulk RNA-seq. Deconvolution identified one of the basal populations to be the predominant tumor subtype in nondissociated ST tissues and in vitro tumor cell and patient-derived organoid lines. We discovered a novel enrichment and spatial association of CXCL10+ cancer-associated fibroblasts with basal tumor cells. We identified that besides immune cells, ductal cells also express CXCR3, the receptor for CXCL10, suggesting a relationship between these cell types in the PDAC tumor microenvironment. CONCLUSIONS We show that our scRNA-seq atlas (700,000 cells), integrated with ST data, has increased statistical power and is a powerful resource, allowing for expansion of current subtyping paradigms in PDAC. We uncovered a novel signaling niche marked by CXCL10+ cancer-associated fibroblasts and basal tumor cells that could be explored for future targeted therapies.
Collapse
Affiliation(s)
- Ian M. Loveless
- Department of Public Health Sciences, Center for Bioinformatics, Henry Ford Health, Detroit, Michigan
- Medical Imaging and Data Integration Lab, Department of Computational Mathematics, Science, and Engineering, Michigan State University, East Lansing, Michigan
| | - Samantha B. Kemp
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kailee M. Hartway
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Hospital, Detroit, Michigan
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan
| | - Jacob T. Mitchell
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Yuesong Wu
- Department of Statistics and Probability, Michigan State University, East Lansing, Michigan
| | - Samuel D. Zwernik
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Hospital, Detroit, Michigan
| | - Daniel James Salas-Escabillas
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Hospital, Detroit, Michigan
- Department of Cancer Biology, University of Michigan, Ann Arbor, Michigan
| | - Sydney Brender
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Hospital, Detroit, Michigan
| | - Madison George
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Hospital, Detroit, Michigan
| | - Yetunde Makinwa
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Hospital, Detroit, Michigan
| | - Thais Stockdale
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Hospital, Detroit, Michigan
| | | | - Rohit G. Reddy
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Hospital, Detroit, Michigan
| | - Daniel W. Long
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Hospital, Detroit, Michigan
| | - Allison Wombwell
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Hospital, Detroit, Michigan
| | - Julie M. Clark
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Hospital, Detroit, Michigan
| | - Albert M. Levin
- Department of Public Health Sciences, Center for Bioinformatics, Henry Ford Health, Detroit, Michigan
| | - David Kwon
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Hospital, Detroit, Michigan
| | - Ling Huang
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Hospital, Detroit, Michigan
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan
- Department of Oncology, Wayne State University, Detroit, Michigan
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Ralph Francescone
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Hospital, Detroit, Michigan
| | | | - Ben Z. Stanger
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Adam Alessio
- Medical Imaging and Data Integration Lab, Department of Computational Mathematics, Science, and Engineering, Michigan State University, East Lansing, Michigan
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan
- Department of Radiology, MSU, Michigan State University, East Lansing, Michigan
| | - Andrew M. Waters
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Yuehua Cui
- Department of Statistics and Probability, Michigan State University, East Lansing, Michigan
| | - Elana J. Fertig
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Applied Mathematics and Statistics, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland
- The Skip Viragh Center for Clinical and Translational Research, Baltimore, Maryland
| | - Luciane T. Kagohara
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Applied Mathematics and Statistics, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Brian Theisen
- Department of Pathology, Henry Ford Health, Detroit, Michigan
| | - Howard C. Crawford
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Hospital, Detroit, Michigan
- Department of Cancer Biology, University of Michigan, Ann Arbor, Michigan
- Department of Oncology, Wayne State University, Detroit, Michigan
| | - Nina G. Steele
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Hospital, Detroit, Michigan
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan
- Department of Oncology, Wayne State University, Detroit, Michigan
| |
Collapse
|
11
|
Yamamoto N, Dobersch S, Loveless I, Samraj AN, Jang GH, Haraguchi M, Kang LI, Ruzinova MB, Vij KR, Mudd JL, Walsh T, Safyan RA, Chiorean EG, Hingorani SR, Bolton NM, Li L, Fields RC, DeNardo DG, Notta F, Crawford HC, Steele NG, Kugel S. HMGA2 Expression Predicts Subtype, Survival, and Treatment Outcome in Pancreatic Ductal Adenocarcinoma. Clin Cancer Res 2025; 31:733-745. [PMID: 39680021 PMCID: PMC11967372 DOI: 10.1158/1078-0432.ccr-24-2200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/17/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
PURPOSE The purpose of this study was to establish HMGA2 as a marker of basal-like disease in pancreatic ductal adenocarcinoma (PDAC) and explore its use as a biomarker for prognosis and treatment resistance. EXPERIMENTAL DESIGN We identified high-mobility group A2 (HMGA2) protein expression in basal PDAC cells in a single-cell RNA sequencing (RNA-seq) atlas of 172 patient samples. We then analyzed HMGA2 expression, along with expression of the classic marker GATA-binding factor 6 (GATA6), in a cohort of 580 PDAC samples with multiplex IHC. We further supplemented these data with an additional 30 diverse patient samples and multiple independent single-cell RNA-seq databases. RESULTS We found that expression of HMGA2, but not previously described basal markers cytokeratins 5 or 17, predicted overall survival in our cohort. Combining HMGA2 and GATA6 statuses allowed for the identification of two key study groups: an HMGA2+/GATA6- cohort with worse survival, low tumor-infiltrating CD8+ T cells, increased FAP+ fibroblasts, and poorer response to gemcitabine-based chemotherapies (n = 94, median survival = 11.2 months after surgery) and an HMGA2-/GATA6+ cohort with improved survival, increased CD8+ T-cell infiltrate, decreased FAP+ fibroblasts, and improved survival with gemcitabine-based chemotherapy (n = 198, median survival = 21.7 months after surgery). HMGA2 was also prognostic for overall survival in RNA-seq from an independent cohort. CONCLUSIONS IHC stratification of primary tumors by HMGA2 and GATA6 statuses in pancreatic cancer is associated with differential outcomes, survival following chemotherapy, and tumor microenvironments. As a nuclear marker for basal disease, HMGA2 complements GATA6 to identify disease subtypes in PDAC.
Collapse
Affiliation(s)
- Naomi Yamamoto
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Program in Molecular and Cellular Biology, University of Washington, Seattle, WA 98195, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Stephanie Dobersch
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Ian Loveless
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI 48202, USA
| | - Annie N. Samraj
- Laboratory Medicine & Pathology, University of Washington, Seattle, WA 98195, USA
| | - Gun Ho Jang
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Miki Haraguchi
- Experimental Histopathology, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Liang-I Kang
- Departments of Medicine and Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marianna B. Ruzinova
- Surgical Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kiran R. Vij
- Departments of Medicine and Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jacqueline L. Mudd
- Surgical Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Thomas Walsh
- Surgical Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rachael A. Safyan
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Hematology and Oncology, University of Washington School of Medicine, Seattle, WA 98195 USA
| | - E. Gabriela Chiorean
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Hematology and Oncology, University of Washington School of Medicine, Seattle, WA 98195 USA
| | - Sunil R. Hingorani
- Division of Hematology and Oncology, Department of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, Pancreatic Cancer Center of Excellence, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Nathan M. Bolton
- Laboratory of Translational Cancer Research, Tom & Gayle Benson Cancer Center, Ochsner Clinic Foundation, New Orleans, LA 70124, USA
| | - Li Li
- Laboratory of Translational Cancer Research, Tom & Gayle Benson Cancer Center, Ochsner Clinic Foundation, New Orleans, LA 70124, USA
| | - Ryan C. Fields
- Surgical Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David G. DeNardo
- Departments of Medicine and Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Faiyaz Notta
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Howard C. Crawford
- Department of Surgery, Henry Ford Health System, Detroit, MI 48202, USA
- Department of Pathology, Wayne State University, Detroit, MI 48202, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
- Department of Oncology, Wayne State University, Detroit, MI 48202, USA
| | - Nina G. Steele
- Department of Surgery, Henry Ford Health System, Detroit, MI 48202, USA
- Department of Pathology, Wayne State University, Detroit, MI 48202, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
- Department of Oncology, Wayne State University, Detroit, MI 48202, USA
| | - Sita Kugel
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
12
|
Famili-Youth EHH, Famili-Youth A, Yang D, Siddique A, Wu EY, Liu W, Resnick MB, Chen Q, Brodsky AS. Aberrant expression of collagen type X in solid tumor stroma is associated with EMT, immunosuppressive and pro-metastatic pathways, bone marrow stromal cell signatures, and poor survival prognosis. BMC Cancer 2025; 25:247. [PMID: 39939916 PMCID: PMC11823173 DOI: 10.1186/s12885-025-13641-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 02/04/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Collagen type X (ColXα1, encoded by COL10A1) is expressed specifically in the cartilage-to-bone transition, in bone marrow cells, and in osteoarthritic (OA) cartilage. We have previously shown that ColXα1 is expressed in breast tumor stroma, correlates with tumor-infiltrating lymphocytes, and predicts poor adjuvant therapy outcomes in ER+/HER2+ breast cancer. However, the underlying molecular mechanisms for these effects are unknown. In this study, we performed bioinformatic analysis of COL10A1-associated gene modules in breast and pancreatic cancer as well as in cells from bone marrow and OA cartilage. These findings provide important insights into the mechanisms of transcriptional and extracellular matrix changes which impact the local stromal microenvironment and tumor progression. METHODS Immunohistochemistry was performed to examine collagen type X expression in solid tumors. WGCNA was used to generate COL10A1-associated gene networks in breast and pancreatic tumor cohorts using RNA-Seq data from The Cancer Genome Atlas. Computational analysis was employed to assess the impact of these gene networks on development and progression of cancer and OA. Data processing and statistical analysis was performed using R and various publicly-available computational tools. RESULTS Expression of COL10A1 and its associated gene networks highlights inflammatory and immunosuppressive microenvironments, which identify aggressive breast and pancreatic tumors and contribute to metastatic potential in a sex-dependent manner. Both cancer types are enriched in stroma, and COL10A1 implicates bone marrow-derived fibroblasts as contributors to the epithelial-to-mesenchymal transition (EMT) in these tumors. Heightened expression of COL10A1 and its associated gene networks is correlated with poorer patient outcomes in both breast and pancreatic cancer. Common transcriptional changes and chondrogenic activity are shared between cancer and OA cartilage, suggesting that similar microenvironmental alterations may underlie both diseases. CONCLUSIONS COL10A1-associated gene networks may hold substantial value as regulators and biomarkers of aggressive tumor phenotypes with implications for therapy development and clinical outcomes. Identification of tumors which exhibit high expression of COL10A1 and its associated genes may reveal the presence of bone marrow-derived stromal microenvironments with heightened EMT capacity and metastatic potential. Our analysis may enable more effective risk assessment and more precise treatment of patients with breast and pancreatic cancer.
Collapse
Affiliation(s)
- Elliot H H Famili-Youth
- Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA.
| | - Aryana Famili-Youth
- Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA.
| | - Dongfang Yang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Ayesha Siddique
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Elizabeth Y Wu
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Wenguang Liu
- Department of Orthopedics, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Present address: School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, Shaanxi, China
| | - Murray B Resnick
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Qian Chen
- Department of Orthopedics, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA.
| | - Alexander S Brodsky
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Center for Computational Molecular Biology, Brown University, Providence, RI, USA
| |
Collapse
|
13
|
Yagi T, Kagawa S, Nogi S, Taniguchi A, Yoshimoto M, Suemori K, Nagai Y, Fujita S, Kuroda S, Kikuchi S, Kakiuchi Y, Teraishi F, Takagi K, Ohara T, Tazawa H, Fujiwara T. Cancer-associated fibroblasts promote pro-tumor functions of neutrophils in pancreatic cancer via IL-8: potential suppression by pirfenidone. Cancer Immunol Immunother 2025; 74:96. [PMID: 39904796 PMCID: PMC11794937 DOI: 10.1007/s00262-025-03946-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND The mechanisms by which neutrophils acquire pro-tumor properties remain poorly understood. In pancreatic cancer, cancer-associated fibroblasts (CAFs) may interact with neutrophils, directing them to promote tumor progression. METHODS To validate the association between CAFs and neutrophils, the localization of neutrophils was examined in clinically resected pancreatic cancer specimens. CAFs were produced by culturing in cancer-conditioned media, and the effects of these CAFs on neutrophils were examined. In vitro migration and invasion assays assess the effect of CAF-activated neutrophils on cancer cells. The factors secreted by the activated neutrophils were also explored. Finally, pirfenidone (PFD) was tested to determine whether it could suppress the pro-tumor functions of activated neutrophils. RESULTS In pancreatic cancer specimens, neutrophils tended to co-localize with IL-6-positive CAFs. Neutrophils co-cultured with CAFs increased migratory capacity and prolonged life span. CAF-affected neutrophils enhance the migratory and invasive activities of pancreatic cancer cells. IL-8 is the most upregulated cytokine secreted by the neutrophils. PFD suppresses IL-8 secretion from CAF-stimulated neutrophils and mitigates the malignant traits of pancreatic cancer cells. CONCLUSION CAFs activate neutrophils and enhance the malignant phenotype of pancreatic cancer. The interactions between cancer cells, CAFs, and neutrophils can be disrupted by PFD, highlighting a potential therapeutic approach.
Collapse
Affiliation(s)
- Tomohiko Yagi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Shunsuke Kagawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan.
| | - Shohei Nogi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Atsuki Taniguchi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Masashi Yoshimoto
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Kanto Suemori
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Yasuo Nagai
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Shuto Fujita
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Shinji Kuroda
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
- Minimally Invasive Therapy Center, Okayama University Hospital, Okayama, Japan
| | - Satoru Kikuchi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Yoshihiko Kakiuchi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
- Minimally Invasive Therapy Center, Okayama University Hospital, Okayama, Japan
| | - Fuminori Teraishi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
- Minimally Invasive Therapy Center, Okayama University Hospital, Okayama, Japan
| | - Kosei Takagi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Toshiaki Ohara
- Departments of Pathology and Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroshi Tazawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| |
Collapse
|
14
|
Kai J, Liu X, Wu M, Liu P, Lin M, Yang H, Zhao Q. Technological advances in clinical individualized medication for cancer therapy: from genes to whole organism. Per Med 2025; 22:45-58. [PMID: 39764674 DOI: 10.1080/17410541.2024.2447224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 12/23/2024] [Indexed: 02/13/2025]
Abstract
Efforts have been made to leverage technology to accurately identify tumor characteristics and predict how each cancer patient may respond to medications. This involves collecting data from various sources such as genomic data, histological information, functional drug profiling, and drug metabolism using techniques like polymerase chain reaction, sanger sequencing, next-generation sequencing, fluorescence in situ hybridization, immunohistochemistry staining, patient-derived tumor xenograft models, patient-derived organoid models, and therapeutic drug monitoring. The utilization of diverse detection technologies in clinical practice has made "individualized treatment" possible, but the desired level of accuracy has not been fully attained yet. Here, we briefly summarize the conventional and state-of-the-art technologies contributing to individualized medication in clinical settings, aiming to explore therapy options enhancing clinical outcomes.
Collapse
Affiliation(s)
- Jiejing Kai
- Department of Clinical Pharmacy, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xueling Liu
- Department of Clinical Pharmacy, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meijia Wu
- Department of Clinical Pharmacy, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pan Liu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Meihua Lin
- Department of Clinical Pharmacy, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongyu Yang
- Department of Clinical Pharmacy, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingwei Zhao
- Department of Clinical Pharmacy, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
15
|
Liu Y, Zhang K, Cai X, Zhou J, Cai Y, Gu Y, Xia T, Ye J. The role of IL‑17, IFN‑γ, 4‑1BBL and tumour‑infiltrating lymphocytes in the occurrence, development and prognosis of pancreatic cancer. Oncol Lett 2025; 29:88. [PMID: 39677412 PMCID: PMC11638937 DOI: 10.3892/ol.2024.14834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/14/2024] [Indexed: 12/17/2024] Open
Abstract
Immunotherapy has made progress in the treatment of tumours; however, in patients with pancreatic cancer, immunotherapy has not achieved effective results. The present study investigated changes in the immune microenvironment during tumour development and progression, and the relationship between the immune microenvironment and prognosis, to clarify the mechanism of immune escape in pancreatic cancer. A total of 40 patients with pancreatic cancer (including 22 with stage I-II disease and 18 with stage III-IV disease) and 20 patients with chronic pancreatitis were included in the present study. The expression of CD3, CD4, CD8, CD56, IFN-γ, IL-17 and 4-1BBL was assessed by immunohistochemistry, and the mRNA expression levels were detected by reverse transcription-quantitative PCR (RT-qPCR). The clinicopathological characteristics and prognoses of patients with pancreatic cancer were analysed to further explore the role of IL-17, IFN-γ, 4-1BBL and tumour-infiltrating lymphocytes in pancreatic cancer. Notably, the expression levels of CD3, CD8, CD56, IFN-γ and 4-1BBL in patients with stages I-II and III-IV cancer were lower than those in patients with chronic pancreatitis (P<0.05), especially in patients with stage III-IV cancer (P<0.05). In addition, the expression of IL-17 in patients with stages I-II and III-IV cancer was greater than in patients with chronic pancreatitis (P<0.05), especially in patients with stage III-IV cancer (P<0.05). The RT-qPCR results regarding CD3, CD4, CD8, CD56, IFN-γ and IL-17 were almost the same as those obtained from immunohistochemical analysis; however, the mRNA expression levels of 4-1BBL were not significantly different between stages I-II and III-IV. Furthermore, patients with pancreatic cancer with higher expression levels of CD3, CD8, CD56, IFN-γ and 4-1BBL exhibited longer survival, whereas those with higher expression of IL-17 had a shorter survival time. The expression levels of CD3, CD8, CD56, cytokines IL-17 and IFN-γ, and costimulatory molecule 4-1BBL were revealed to be related to the degree of differentiation, Tumour-Node-Metastasis staging and the prognosis of pancreatic cancer, and may serve as novel immunological indicators for evaluating the condition and treatment effectiveness in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Yingying Liu
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, P.R. China
| | - Ke Zhang
- Department of Gastroenterology, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu 215500, P.R. China
| | - Xiaodi Cai
- Department of Gastroenterology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jikai Zhou
- Department of Gastroenterology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yixuan Cai
- Department of Gastroenterology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yujie Gu
- Department of Gastroenterology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Tingting Xia
- Department of Gastroenterology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jianxin Ye
- Department of Gastroenterology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
16
|
Alver TN, Bergholtz H, Holm MB, Dorg LT, Skrede ML, Kure EH, Verbeke CS. Spatial Transcriptomics Reveals Cancer and Stromal Cell Heterogeneity Between Center and Invasive Front of Pancreatic Cancer. Mod Pathol 2025; 38:100726. [PMID: 39889965 DOI: 10.1016/j.modpat.2025.100726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 12/19/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
Intratumor heterogeneity is considered a major cause of treatment failure in pancreatic ductal adenocarcinoma (PDAC). In recent years, marked heterogeneity at the genomic and transcriptional level has been revealed, but the spatial distribution of the heterogeneous cell populations has not been considered. Yet, it is assumed that cancer cells at the invasive front are endowed with enhanced migratory and invasive properties, although evidence is scanty, and cancer-associated fibroblasts (CAFs) in this location have not been characterized. In this study, digital spatial profiling was used to compare the transcriptional profiles of cancer cells and CAFs in the tumor center versus the invasive front of human PDAC. Four well-differentiated PDACs with conventional morphology were investigated with the GeoMx system (Nanostring). Regions of interest were analyzed in the tumor center and at the invasive front using a whole transcriptome assay in the cancer cell and CAF segments separately. Three of the PDACs harbored mutated KRAS, whereas the fourth case was confirmed wild-type KRAS. Substantial inter-regional heterogeneity was identified, with increased activity of pathways associated with cellular stress (including TNFα-signaling via NFκB, hypoxia, P53 pathway), proliferation (MYC targets, mitotic spindle), glycolysis, and epithelial-mesenchymal transition (EMT) at the invasive front in both the cancer cell and CAF segments compared with the center of the tumor. Immunohistochemical validation on 17 PDACs of well, moderate, and poor differentiation confirmed significant inter-regional heterogeneity in the expression level of markers of EMT and glycolysis. The results of this study show that in PDAC, transcriptional profiles of both cancer cells and CAFs differ between the center of the tumor and the invasive front.
Collapse
Affiliation(s)
- Tine Norman Alver
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Norway.
| | - Helga Bergholtz
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Norway
| | - Maia Blomhoff Holm
- Department of Pathology, Oslo University Hospital, Rikshospitalet, Norway; Department of Pathology, Institute of Clinical Medicine, University of Oslo, Norway
| | - Linda Trobe Dorg
- Department of Pathology, Institute of Clinical Medicine, University of Oslo, Norway
| | | | - Elin Hegland Kure
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Norway
| | - Caroline Sophie Verbeke
- Department of Pathology, Oslo University Hospital, Rikshospitalet, Norway; Department of Pathology, Institute of Clinical Medicine, University of Oslo, Norway
| |
Collapse
|
17
|
Xia L, Guo X, Lu D, Jiang Y, Liang X, Shen Y, Lin J, Zhang L, Chen H, Jin J, Luan X, Zhang W. S100A13-driven interaction between pancreatic adenocarcinoma cells and cancer-associated fibroblasts promotes tumor progression through calcium signaling. Cell Commun Signal 2025; 23:51. [PMID: 39871271 PMCID: PMC11773924 DOI: 10.1186/s12964-025-02049-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 01/16/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) are key components of the pancreatic adenocarcinoma (PAAD) tumor microenvironment (TME), where they promote tumor progression and metastasis through immunosuppressive functions. Although significant progress has been made in understanding the crosstalk between cancer cells and CAFs, many underlying mechanisms remain unclear. Recent studies have highlighted the importance of calcium signaling in enhancing interactions between tumor cells and the surrounding stroma, with the S100 family of proteins serving as important regulators. While the roles of some S100 proteins have been extensively studied, others, such as S100A13, remain less well understood. METHODS Bioinformatic analysis was employed to predict the pathogenic potential of CAFs and S100A13. Stable S100A13 knockdown CAFs were generated using a short hairpin RNA system. Cellular viability and apoptosis rates were evaluated through CCK-8 and flow cytometry tests, respectively. Additionally, the wound healing and migration assays were conducted to assess the invasive and metastatic capabilities. Transcriptome analysis was conducted to identify differential gene expression and associated signaling pathways in PAAD cells derived from an indirect culture system. Furthermore, the protumoral role of S100A13 in PAAD was further verified using both 3D bioprinting and cell line-based xenograft tumor models. RESULTS In this study, we identified a strong association between S100A13, a calcium-binding protein, and CAFs in PAAD. Gene expression analysis revealed that S100A13 was highly expressed in CAFs and correlated with poor prognosis. Knockdown of S100A13 in CAFs reduced the metastatic potential of PAAD cells. In addition, S100A13 depletion impaired cell motility and calcium signaling pathways within the TME. Furthermore, silencing S100A13 in CAFs markedly slowed PAAD progression in both tumor spheroids and Balb/c nude mice. CONCLUSIONS Together, our findings underscore the critical role of CAFs-derived S100A13 in PAAD progression and suggest that targeting S100A13 may offer a promising therapeutic strategy for PAAD.
Collapse
Affiliation(s)
- Liuyuan Xia
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Xin Guo
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dong Lu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yixin Jiang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaohui Liang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yiwen Shen
- Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jiayi Lin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lijun Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hongzhuan Chen
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jinmei Jin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Xin Luan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Weidong Zhang
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China.
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100700, China.
| |
Collapse
|
18
|
Huang Y, Zhang R, Fan S, Shi M, Tang X, Wang X, Deng X. OSBPL10-CNBP axis mediates hypoxia-induced pancreatic cancer development. Biofactors 2025; 51:e2124. [PMID: 39329194 DOI: 10.1002/biof.2124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of malignancies with worst outcomes among digestive system tumors. Identification of novel biomarkers is of great significance for treatment researches and prognosis prediction of pancreatic cancer patients. Due to OSBPL10 known involvement in oncogenic activity in other tumors, we elucidated the mechanism underlying its contribution to pancreatic cancer progression. We employed data from the Gene Expression Omnibus database to detect the expression of OSBPL10 in normal and pancreatic cancer tissues. A series of assays were conducted to assess the impact of OSBPL10 on the proliferation and metastatic capacities of pancreatic cancer cells and the influence of OSBPL10 on macrophages were evaluated by Flow cytometry. In addition, Co-immunoprecipitation, mass spectrometry, and western blot assays were utilized to investigate the potential mechanisms of OSBPL10 activity. From our study, OSBPL10 is revealed to be upregulated in pancreatic cancer, with poor prognosis. The overexpression promotes malignant behaviors of pancreatic cancer cells and has an impact on tumor immune microenvironment by stimulating the transformation M1 macrophages into M2 macrophages. Mechanistically, hypoxia induces the expression of OSBPL10 through interaction between hypoxia-inducible factor 1-α and the promoter region of OSBPL10. Additionally, OSBPL10 directly bound to CNBP, mediating CNBP expression and ultimately regulating the proliferation and metastasis capacity of pancreatic cancer cells, as well as influencing macrophage polarization. The research emphasized the oncogenic role of OSBPL10 in pancreatic cancer, uncovering key mechanisms involving hypoxia, HIF-1α, and CNBP. The finding suggests that OSBPL10 is a novel biomarker in pancreatic cancer, making it a potential therapeutic target for intervention in this malignancy.
Collapse
Affiliation(s)
- Yishu Huang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Neoplasms Translational Medicine, Shanghai, China
| | - Ronghao Zhang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shuyang Fan
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
| | - Minmin Shi
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
| | - Xiaomei Tang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xinjing Wang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaxing Deng
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Naro C, Ruta V, Sette C. Splicing dysregulation: hallmark and therapeutic opportunity in pancreatic cancer. Trends Mol Med 2024:S1471-4914(24)00308-3. [PMID: 39648052 DOI: 10.1016/j.molmed.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 12/10/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer characterized by dismal prognosis. Late diagnosis, resistance to chemotherapy, and lack of efficacious targeted therapies render PDAC almost untreatable. Dysregulation of splicing, the process that excises the introns from nascent transcripts, is emerging as a hallmark of PDAC and a possible vulnerability of this devastating cancer. Splicing factors are deregulated in PDAC and contribute to all steps of tumorigenesis, from inflammation-related early events to metastasis and acquisition of chemoresistance. At the same time, splicing dysregulation offers a therapeutic opportunity to target cancer-specific vulnerabilities. We discuss mounting evidence that splicing plays a key role in PDAC and the opportunities that this essential process offers for developing new targeted therapies.
Collapse
Affiliation(s)
- Chiara Naro
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, 00168 Rome, Italy; Gemelli Science and Technology Park (GSTeP) Organoids Research Core Facility, Fondazione Policlinico A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Veronica Ruta
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Claudio Sette
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, 00168 Rome, Italy; Gemelli Science and Technology Park (GSTeP) Organoids Research Core Facility, Fondazione Policlinico A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy.
| |
Collapse
|
20
|
Hashimoto A, Hashimoto S. Plasticity and Tumor Microenvironment in Pancreatic Cancer: Genetic, Metabolic, and Immune Perspectives. Cancers (Basel) 2024; 16:4094. [PMID: 39682280 DOI: 10.3390/cancers16234094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer has long been believed to be a genetic disease caused by the accumulation of mutations in key genes involved in cellular processes. However, recent advances in sequencing technology have demonstrated that cells with cancer driver mutations are also present in normal tissues in response to aging, environmental damage, and chronic inflammation, suggesting that not only intrinsic factors within cancer cells, but also environmental alterations are important key factors in cancer development and progression. Pancreatic cancer tissue is mostly comprised of stromal cells and immune cells. The desmoplasmic microenvironment characteristic of pancreatic cancer is hypoxic and hypotrophic. Pancreatic cancer cells may adapt to this environment by rewiring their metabolism through epigenomic changes, enhancing intrinsic plasticity, creating an acidic and immunosuppressive tumor microenvironment, and inducing noncancerous cells to become tumor-promoting. In addition, pancreatic cancer has often metastasized to local and distant sites by the time of diagnosis, suggesting that a similar mechanism is operating from the precancerous stage. Here, we review key recent findings on how pancreatic cancers acquire plasticity, undergo metabolic reprogramming, and promote immunosuppressive microenvironment formation during their evolution. Furthermore, we present the following two signaling pathways that we have identified: one based on the small G-protein ARF6 driven by KRAS/TP53 mutations, and the other based on the RNA-binding protein Arid5a mediated by inflammatory cytokines, which promote both metabolic reprogramming and immune evasion in pancreatic cancer. Finally, the striking diversity among pancreatic cancers in the relative importance of mutational burden and the tumor microenvironment, their clinical relevance, and the potential for novel therapeutic strategies will be discussed.
Collapse
Affiliation(s)
- Ari Hashimoto
- Department of Molecular Biology, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Shigeru Hashimoto
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0818, Japan
| |
Collapse
|
21
|
Famili-Youth EHH, Famili-Youth A, Yang D, Siddique A, Wu EY, Liu W, Resnick MB, Chen Q, Brodsky AS. Aberrant expression of collagen type X in solid tumor stroma is associated with EMT, immunosuppressive and pro-metastatic pathways, bone marrow stromal cell signatures, and poor survival prognosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.621984. [PMID: 39605631 PMCID: PMC11601388 DOI: 10.1101/2024.11.13.621984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Background Collagen type X (ColXα1, encoded by COL10A1) is expressed specifically in the cartilage-to-bone transition, in bone marrow cells, and in osteoarthritic (OA) cartilage. We have previously shown that ColXα1 is expressed in breast tumor stroma, correlates with tumor-infiltrating lymphocytes, and predicts poor adjuvant therapy outcomes in ER+/HER2+ breast cancer. However, the underlying molecular mechanisms for these effects are unknown. In this study, we performed bioinformatic analysis of COL10A1-associated gene modules in breast and pancreatic cancer as well as in cells from bone marrow and OA cartilage. These findings provide important insights into the mechanisms of transcriptional and extracellular matrix changes which impact the local stromal microenvironment and tumor progression. Methods Immunohistochemistry was performed to examine collagen type X expression in solid tumors. WGCNA was used to generate COL10A1-associated gene networks in breast and pancreatic tumor cohorts using RNA-Seq data from The Cancer Genome Atlas. Computational analysis was employed to assess the impact of these gene networks on development and progression of cancer and OA. Data processing and statistical analysis was performed using R and various publicly-available computational tools. Results Expression of COL10A1 and its associated gene networks highlights inflammatory and immunosuppressive microenvironments, which identify aggressive breast and pancreatic tumors and contribute to metastatic potential in a sex-dependent manner. Both cancer types are enriched in stroma, and COL10A1 implicates bone marrow-derived fibroblasts as drivers of the epithelial-to-mesenchymal transition (EMT) in these tumors. Heightened expression of COL10A1 and its associated gene networks is correlated with poorer patient outcomes in both breast and pancreatic cancer. Common transcriptional changes and chondrogenic activity are shared between cancer and OA cartilage, suggesting that similar microenvironmental alterations may underlie both diseases. Conclusions COL10A1-associated gene networks may hold substantial value as regulators and biomarkers of aggressive tumor phenotypes with implications for therapy development and clinical outcomes. Identification of tumors which exhibit high expression of COL10A1 and its associated genes may reveal the presence of bone marrow-derived stromal microenvironments with heightened EMT capacity and metastatic potential. Our analysis may enable more effective risk assessment and more precise treatment of patients with breast and pancreatic cancer.
Collapse
Affiliation(s)
- Elliot H H Famili-Youth
- Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Aryana Famili-Youth
- Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Dongfang Yang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Ayesha Siddique
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Elizabeth Y Wu
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Wenguang Liu
- Department of Orthopedics, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Murray B Resnick
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Qian Chen
- Department of Orthopedics, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Alexander S Brodsky
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Center for Computational Molecular Biology, Brown University, Providence, RI, USA
| |
Collapse
|
22
|
Carpenter ES, Vendramini-Costa DB, Hasselluhn MC, Maitra A, Olive KP, Cukierman E, Pasca di Magliano M, Sherman MH. Pancreatic Cancer-Associated Fibroblasts: Where Do We Go from Here? Cancer Res 2024; 84:3505-3508. [PMID: 39283867 DOI: 10.1158/0008-5472.can-24-2860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 11/05/2024]
Abstract
Pancreatic ductal adenocarcinoma is a deadly disease and is projected to become the second leading cause of cancer-related death by 2030. A major hallmark is the exuberant host response comprising the tumor microenvironment, of which, cancer-associated fibroblasts (CAF) are a prevalent component. Despite the gains in understanding of their heterogeneity and functionality from CAF studies in recent years, there are many unanswered questions surrounding this diverse population of cells. Here, we summarize the views of several experts in the field, focusing on the current understanding of CAFs and challenges to address.
Collapse
Affiliation(s)
- Eileen S Carpenter
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Debora Barbosa Vendramini-Costa
- Department of Surgery, Henry Ford Health, Detroit, Michigan
- Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, Michigan
| | - Marie C Hasselluhn
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Anirban Maitra
- Department of Translational Molecular Pathology, Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kenneth P Olive
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Edna Cukierman
- Cancer Signaling and Microenvironment Program, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Temple Health, Philadelphia, Pennsylvania
| | - Marina Pasca di Magliano
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Mara H Sherman
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
23
|
Xia C, Qu JR, Jiao YP, Lu CQ, Zhao B, Ge RJ, Qiu Y, Cao BY, Yu Q, Xia TY, Meng XP, Song Y, Zhang LH, Long XY, Ye J, Ding ZM, Cai W, Ju SH. Signal enhancement ratio of multi-phase contrast-enhanced MRI: an imaging biomarker for survival in pancreatic adenocarcinoma. Eur Radiol 2024; 34:7460-7470. [PMID: 38750169 PMCID: PMC11519106 DOI: 10.1007/s00330-024-10746-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/07/2024] [Accepted: 03/30/2024] [Indexed: 10/29/2024]
Abstract
OBJECTIVES To evaluate signal enhancement ratio (SER) for tissue characterization and prognosis stratification in pancreatic adenocarcinoma (PDAC), with quantitative histopathological analysis (QHA) as the reference standard. METHODS This retrospective study included 277 PDAC patients who underwent multi-phase contrast-enhanced (CE) MRI and whole-slide imaging (WSI) from three centers (2015-2021). SER is defined as (SIlt - SIpre)/(SIea - SIpre), where SIpre, SIea, and SIlt represent the signal intensity of the tumor in pre-contrast, early-, and late post-contrast images, respectively. Deep-learning algorithms were implemented to quantify the stroma, epithelium, and lumen of PDAC on WSIs. Correlation, regression, and Bland-Altman analyses were utilized to investigate the associations between SER and QHA. The prognostic significance of SER on overall survival (OS) was evaluated using Cox regression analysis and Kaplan-Meier curves. RESULTS The internal dataset comprised 159 patients, which was further divided into training, validation, and internal test datasets (n = 60, 41, and 58, respectively). Sixty-five and 53 patients were included in two external test datasets. Excluding lumen, SER demonstrated significant correlations with stroma (r = 0.29-0.74, all p < 0.001) and epithelium (r = -0.23 to -0.71, all p < 0.001) across a wide post-injection time window (range, 25-300 s). Bland-Altman analysis revealed a small bias between SER and QHA for quantifying stroma/epithelium in individual training, validation (all within ± 2%), and three test datasets (all within ± 4%). Moreover, SER-predicted low stromal proportion was independently associated with worse OS (HR = 1.84 (1.17-2.91), p = 0.009) in training and validation datasets, which remained significant across three combined test datasets (HR = 1.73 (1.25-2.41), p = 0.001). CONCLUSION SER of multi-phase CE-MRI allows for tissue characterization and prognosis stratification in PDAC. CLINICAL RELEVANCE STATEMENT The signal enhancement ratio of multi-phase CE-MRI can serve as a novel imaging biomarker for characterizing tissue composition and holds the potential for improving patient stratification and therapy in PDAC. KEY POINTS Imaging biomarkers are needed to better characterize tumor tissue in pancreatic adenocarcinoma. Signal enhancement ratio (SER)-predicted stromal/epithelial proportion showed good agreement with histopathology measurements across three distinct centers. Signal enhancement ratio (SER)-predicted stromal proportion was demonstrated to be an independent prognostic factor for OS in PDAC.
Collapse
Affiliation(s)
- Cong Xia
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu, China
| | - Jin-Rong Qu
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Yi-Ping Jiao
- Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China
| | - Chun-Qiang Lu
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu, China
| | - Ben Zhao
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu, China
| | - Rong-Jun Ge
- School of Instrument Science and Engineering, Southeast University, Nanjing, China
| | - Yue Qiu
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu, China
| | - Bu-Yue Cao
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu, China
| | - Qian Yu
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu, China
| | - Tian-Yi Xia
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu, China
| | - Xiang-Pan Meng
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu, China
| | - Yang Song
- MR Scientific Marketing, Siemens Healthineers Ltd., Shanghai, China
| | - Li-Hua Zhang
- Department of Pathology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xue-Ying Long
- Department of Radiology, The Xiangya Hospital of Central South University, Changsha, China
| | - Jing Ye
- Department of Medical Imaging, Subei People's Hospital, Medical School of Yangzhou University, Yangzhou, China
| | - Zhi-Min Ding
- Department of Radiology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Wu Cai
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Sheng-Hong Ju
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu, China.
| |
Collapse
|
24
|
Mushtaq A, Iqbal MZ, Tang J, Sun W. The wonders of X-PDT: an advance route to cancer theranostics. J Nanobiotechnology 2024; 22:655. [PMID: 39456085 PMCID: PMC11520131 DOI: 10.1186/s12951-024-02931-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Global mortality data indicates cancer as the second-leading cause of death worldwide. Therefore, there's a pressing need to innovate effective treatments to address this significant medical and societal challenge. In recent years, X-ray-induced photodynamic therapy (X-PDT) has emerged as a promising advancement, revolutionizing traditional photodynamic therapy (PDT) for deeply entrenched malignancies by harnessing penetrating X-rays as external stimuli. Recent developments in X-ray photodynamic therapy have shown a trend toward minimizing radiation doses to remarkably low levels after the proof-of-concept demonstration. Early detection and real-time monitoring are crucial aspects of effective cancer treatment. Sophisticated X-ray imaging techniques have been enhanced by the introduction of X-ray luminescence nano-agents, alongside contrast nanomaterials based on X-ray attenuation. X-ray luminescence-based in vivo imaging offers excellent detection sensitivity and superior image quality in deep tissues at a reasonable cost, due to unhindered penetration and unimpeded auto-fluorescence of X-rays. This review emphasizes the significance of X-ray responsive theranostics, exploring their mechanism of action, feasibility, biocompatibility, and promising prospects in imaging-guided therapy for deep-seated tumors. Additionally, it discusses promising applications of X-PDT in treating breast cancer, liver cancer, lung cancer, skin cancer, and colorectal cancer.
Collapse
Affiliation(s)
- Asim Mushtaq
- Institute for Intelligent Bio/Chem Manufacturing (iBCM), ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311200, Zhejiang, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310007, China
| | - Muhammad Zubair Iqbal
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Jianbin Tang
- Institute for Intelligent Bio/Chem Manufacturing (iBCM), ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311200, Zhejiang, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310007, China
| | - Wenjing Sun
- Institute for Intelligent Bio/Chem Manufacturing (iBCM), ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311200, Zhejiang, China.
| |
Collapse
|
25
|
Picozzi VJ. Pancreatic cancer: new approaches to drug therapy. Int J Surg 2024; 110:6070-6080. [PMID: 38573111 PMCID: PMC11486970 DOI: 10.1097/js9.0000000000000877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/22/2023] [Indexed: 04/05/2024]
Abstract
Outcomes in pancreatic ductal adenocarcinoma (PDAC) remain poor due to a variety of biological, clinical, and societal factors. However, in recent years, PDAC has seen 1) increased precision of initial evaluation, 2) increased emphasis on supportive care, 3) deeper understanding of the translation biology of PDAC, especially as pertains to genomic alterations, and 4) foundational combination chemotherapy clinical trials across all disease stages. These advances have led to a wide range of new approaches to drug therapy for PDAC. Currently available drugs are showing added benefit, both by resequencing them with each other and also with respect to other therapeutic modalities. Molecular strategies are being developed to predict response to known therapeutic agents and to identify others. Additionally, a wide range of new drugs for PDAC are under development, including drugs which inhibit critical molecular pathways, drugs which attempt to capitalize on homologous repair deficiencies, immunotherapeutic approaches, antimetabolic agents, and drugs which attack the extracellular matrix which supports PDAC growth. These new approaches offer the promise of improved survival for future PDAC patients.
Collapse
|
26
|
Sozzi S, Manni I, Ercolani C, Diodoro MG, Bartolazzi A, Spallotta F, Piaggio G, Monteonofrio L, Soddu S, Rinaldo C, Valente D. Inactivation of HIPK2 attenuates KRAS G12D activity and prevents pancreatic tumorigenesis. J Exp Clin Cancer Res 2024; 43:265. [PMID: 39342278 PMCID: PMC11437985 DOI: 10.1186/s13046-024-03189-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/11/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) features KRAS mutations in approximately 90% of human cases and excessive stromal response, termed desmoplastic reaction. Oncogenic KRAS drives pancreatic carcinogenesis by acting on both epithelial cells and tumor microenvironment (TME). We have previously shown that Homeodomain-Interacting Protein Kinase 2 (HIPK2) cooperates with KRAS in sustaining ERK1/2 phosphorylation in human colorectal cancers. Here, we investigated whether HIPK2 contributes to oncogenic KRAS-driven tumorigenesis in vivo, in the onset of pancreatic cancer. METHODS We employed an extensively characterized model of KRASG12D-dependent preinvasive PDAC, the Pdx1-Cre;LSL-KRasG12D/+ (KC) mice. In these mice, HIPK2 was inhibited by genetic knockout in the pancreatic epithelial cells (KCH-/-) or by pharmacologic inactivation with the small molecule 5-IodoTubercidin (5-ITu). The development of preneoplastic acinar-to-ductal metaplasia (ADM), intraepithelial neoplasia (PanIN), and their associated desmoplastic reaction were analyzed. RESULTS In Hipk2-KO mice (KCH-/-), ERK phosphorylation was lowered, the appearance of ADM was slowed down, and both the number and pathologic grade of PanIN were reduced compared to Hipk2-WT KC mice. The pancreatic lesion phenotype in KCH-/- mice was characterized by abundant collagen fibers and reduced number of αSMA+ and pSTAT3+ desmoplastic cells. These features were reminiscent of the recently described human "deserted" sub-TME, poor in cells, rich in matrix, and associated with tumor differentiation. In contrast, the desmoplastic reaction of KC mice resembled the "reactive" sub-TME, rich in stromal cells and associated with tumor progression. These observations were confirmed by the pharmacologic inhibition of HIPK2 in KC mice. CONCLUSION This study demonstrates that HIPK2 inhibition weakens oncogenic KRAS activity and pancreatic tumorigenesis providing a rationale for testing HIPK2 inhibitors to mitigate the incidence of PDAC development in high-risk individuals.
Collapse
Affiliation(s)
- Silvia Sozzi
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Department of Science, Roma Tre University, Rome, Italy
| | - Isabella Manni
- SAFU Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Cristiana Ercolani
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maria Grazia Diodoro
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Armando Bartolazzi
- Pathology Research Laboratories, Sant'Andrea University Hospital, Rome, Italy
| | - Francesco Spallotta
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, Rome, Italy
| | - Giulia Piaggio
- SAFU Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Laura Monteonofrio
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Soddu
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Cinzia Rinaldo
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR), c/o Sapienza University, Rome, Italy.
| | - Davide Valente
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR), c/o Sapienza University, Rome, Italy.
| |
Collapse
|
27
|
Struth E, Labaf M, Karimnia V, Liu Y, Cramer G, Dahl JB, Slack FJ, Zarringhalam K, Celli JP. Drug resistant pancreatic cancer cells exhibit altered biophysical interactions with stromal fibroblasts in imaging studies of 3D co-culture models. Sci Rep 2024; 14:20698. [PMID: 39237667 PMCID: PMC11377574 DOI: 10.1038/s41598-024-71372-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 08/27/2024] [Indexed: 09/07/2024] Open
Abstract
Interactions between tumor and stromal cells are well known to play prominent roles in progression of pancreatic ductal adenocarcinoma (PDAC). As knowledge of stromal crosstalk in PDAC has evolved, it has become clear that cancer associated fibroblasts can play both tumor promoting and tumor suppressive roles through a combination of paracrine crosstalk and juxtacrine interactions involving direct physical contact. Another major contributor to dismal survival statistics for PDAC is development of resistance to chemotherapy drugs, though less is known about how the acquisition of chemoresistance impacts upon tumor-stromal crosstalk. Here, we use time lapse imaging and image analysis to study how co-culture geometry impacts interactions between epithelial and stromal cells. We show that extracellular matrix (ECM) overlay cultures in which stromal cells (pancreatic stellate cells, or normal human fibroblasts) are placed adjacent to PDAC cells (PANC1) result in direct heterotypic cell adhesions accompanied by dramatic fibroblast contractility. We analyze these interactions in co-cultures using particle image velocimetry (PIV) analysis to quantify cell velocities over the course of time lapse movie sequences. We further contrast co-cultures of PANC1 with those containing a drug resistant subline (PANC1-OR) previously established in our lab and find that heterotypic cell-cell interactions are suppressed in the latter relative to the parental line. We use RNA-seq and bioinformatics analysis to identify differential gene expression in PANC1 and PANC1-OR, which shows that negative regulation of cell adhesion molecules, consistent with increased epithelial mesenchymal transition (EMT), is also correlated with reduction in the hetrotypic cell-cell contact necessary for the contractile behavior observed in drug naïve cultures. Overall these findings elucidate the role of drug-resistance in inhibiting an avenue of stromal crosstalk which is associated with tumor suppression and also help to establish cell culture conditions useful for further mechanistic investigation.
Collapse
Affiliation(s)
- Eric Struth
- Department of Physics, University of Massachusetts Boston, Boston, MA, 02125, USA
| | - Maryam Labaf
- Department of Mathematics, University of Massachusetts Boston, Boston, MA, 02125, USA
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA, 02125, USA
| | - Vida Karimnia
- Department of Physics, University of Massachusetts Boston, Boston, MA, 02125, USA
| | - Yiran Liu
- Department of Physics, University of Massachusetts Boston, Boston, MA, 02125, USA
| | - Gwendolyn Cramer
- Department of Physics, University of Massachusetts Boston, Boston, MA, 02125, USA
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joanna B Dahl
- Department of Engineering, University of Massachusetts Boston, Boston, MA, 02125, USA
| | - Frank J Slack
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School Initiative for RNA Medicine, Boston, MA, 02115, USA
| | - Kourosh Zarringhalam
- Department of Mathematics, University of Massachusetts Boston, Boston, MA, 02125, USA
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA, 02125, USA
| | - Jonathan P Celli
- Department of Physics, University of Massachusetts Boston, Boston, MA, 02125, USA.
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA, 02125, USA.
| |
Collapse
|
28
|
Li H, Liu D, Li K, Wang Y, Zhang G, Qi L, Xie K. Pancreatic stellate cells and the interleukin family: Linking fibrosis and immunity to pancreatic ductal adenocarcinoma (Review). Mol Med Rep 2024; 30:159. [PMID: 38994764 PMCID: PMC11258612 DOI: 10.3892/mmr.2024.13283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/19/2024] [Indexed: 07/13/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive form of cancer with a low survival rate. A successful treatment strategy should not be limited to targeting cancer cells alone, but should adopt a more comprehensive approach, taking into account other influential factors. These include the extracellular matrix (ECM) and immune microenvironment, both of which are integral components of the tumor microenvironment. The present review describes the roles of pancreatic stellate cells, differentiated cancer‑associated fibroblasts and the interleukin family, either independently or in combination, in the progression of precursor lesions in pancreatic intraepithelial neoplasia and PDAC. These elements contribute to ECM deposition and immunosuppression in PDAC. Therapeutic strategies that integrate interleukin and/or stromal blockade for PDAC immunomodulation and fibrogenesis have yielded inconsistent results. A deeper comprehension of the intricate interplay between fibrosis, and immune responses could pave the way for more effective treatment targets, by elucidating the mechanisms and causes of ECM fibrosis during PDAC progression.
Collapse
Affiliation(s)
- Haichao Li
- Institute of Digestive Disease, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, P.R. China
| | - Donglian Liu
- Institute of Digestive Disease, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, P.R. China
| | - Kaishu Li
- Institute of Digestive Disease, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, P.R. China
| | - Yichen Wang
- Institute of Digestive Disease, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, P.R. China
| | - Gengqiang Zhang
- Institute of Digestive Disease, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, P.R. China
| | - Ling Qi
- Institute of Digestive Disease, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, P.R. China
| | - Keping Xie
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510000, P.R. China
| |
Collapse
|
29
|
Sgarminato V, Madrid-Wolff J, Boniface A, Ciardelli G, Tonda-Turo C, Moser C. 3D in vitromodeling of the exocrine pancreatic unit using tomographic volumetric bioprinting. Biofabrication 2024; 16:045034. [PMID: 39121863 DOI: 10.1088/1758-5090/ad6d8d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/09/2024] [Indexed: 08/12/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer, a leading cause of cancer-related deaths globally. Initial lesions of PDAC develop within the exocrine pancreas' functional units, with tumor progression driven by interactions between PDAC and stromal cells. Effective therapies require anatomically and functionally relevantin vitrohuman models of the pancreatic cancer microenvironment. We employed tomographic volumetric bioprinting, a novel biofabrication method, to create human fibroblast-laden constructs mimicking the tubuloacinar structures of the exocrine pancreas. Human pancreatic ductal epithelial (HPDE) cells overexpressing the KRAS oncogene (HPDE-KRAS) were seeded in the multiacinar cavity to replicate pathological tissue. HPDE cell growth and organization within the structure were assessed, demonstrating the formation of a thin epithelium covering the acini inner surfaces. Immunofluorescence assays showed significantly higher alpha smooth muscle actin (α-SMA) vs. F-actin expression in fibroblasts co-cultured with cancerous versus wild-type HPDE cells. Additionally,α-SMA expression increased over time and was higher in fibroblasts closer to HPDE cells. Elevated interleukin (IL)-6 levels were quantified in supernatants from co-cultures of stromal and HPDE-KRAS cells. These findings align with inflamed tumor-associated myofibroblast behavior, serving as relevant biomarkers to monitor early disease progression and target drug efficacy. To our knowledge, this is the first demonstration of a 3D bioprinted model of exocrine pancreas that recapitulates its true 3-dimensional microanatomy and shows tumor triggered inflammation.
Collapse
Affiliation(s)
- Viola Sgarminato
- Laboratory of Applied Photonics Devices, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Jorge Madrid-Wolff
- Laboratory of Applied Photonics Devices, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Antoine Boniface
- Laboratory of Applied Photonics Devices, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Gianluca Ciardelli
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Chiara Tonda-Turo
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Christophe Moser
- Laboratory of Applied Photonics Devices, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
30
|
Burchard PR, Ruffolo LI, Ullman NA, Dale BS, Dave YA, Hilty BK, Ye J, Georger M, Jewell R, Miller C, De Las Casas L, Jarolimek W, Perryman L, Byrne MM, Loria A, Marin C, Chávez Villa M, Yeh JJ, Belt BA, Linehan DC, Hernandez-Alejandro R. Pan-lysyl oxidase inhibition disrupts fibroinflammatory tumor stroma, rendering cholangiocarcinoma susceptible to chemotherapy. Hepatol Commun 2024; 8:e0502. [PMID: 39101793 PMCID: PMC11299993 DOI: 10.1097/hc9.0000000000000502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/11/2024] [Indexed: 08/06/2024] Open
Abstract
BACKGROUND Cholangiocarcinoma (CCA) features highly desmoplastic stroma that promotes structural and functional resistance to therapy. Lysyl oxidases (LOX, LOXL1-4) catalyze collagen cross-linking, thereby increasing stromal rigidity and facilitating therapeutic resistance. Here, we evaluate the role of lysyl oxidases in stromal desmoplasia and the effects of pan-lysyl oxidase (pan-LOX) inhibition in CCA. METHODS Resected CCA and normal liver specimens were analyzed from archival tissues. Spontaneous and orthotopic murine models of intrahepatic CCA (iCCA) were used to assess the impact of the pan-LOX inhibitor PXS-5505 in treatment and correlative studies. The functional role of pan-LOX inhibition was interrogated through in vivo and ex vivo assays. RESULTS All 5 lysyl oxidases are upregulated in CCA and reduced lysyl oxidase expression is correlated with an improved prognosis in resected patients with CCA. Spontaneous and orthotopic murine models of intrahepatic cholangiocarcinoma upregulate all 5 lysyl oxidase isoforms. Pan-LOX inhibition reversed mechanical compression of tumor vasculature, resulting in improved chemotherapeutic penetrance and cytotoxic efficacy. The combination of chemotherapy with pan-LOX inhibition increased damage-associated molecular pattern release, which was associated with improved antitumor T-cell responses. Pan-LOX inhibition downregulated macrophage invasive signatures in vitro, rendering tumor-associated macrophages more susceptible to chemotherapy. Mice bearing orthotopic and spontaneously occurring intrahepatic cholangiocarcinoma tumors exhibited delayed tumor growth and improved survival following a combination of pan-LOX inhibition with chemotherapy. CONCLUSIONS CCA upregulates all 5 lysyl oxidase isoforms, and pan-LOX inhibition reverses tumor-induced mechanical forces associated with chemotherapy resistance to improve chemotherapeutic efficacy and reprogram antitumor immune responses. Thus, combination therapy with pan-LOX inhibition represents an innovative therapeutic strategy in CCA.
Collapse
Affiliation(s)
- Paul R. Burchard
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Luis I. Ruffolo
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Nicholas A. Ullman
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Benjamin S. Dale
- Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | - Yatee A. Dave
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Bailey K. Hilty
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Jian Ye
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Mary Georger
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Rachel Jewell
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Christine Miller
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Luis De Las Casas
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | - Lara Perryman
- Drug Discovery, Syntara Ltd., Sydney, New South Wales, Australia
| | - Matthew M. Byrne
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Anthony Loria
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Chelsea Marin
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Mariana Chávez Villa
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Jen Jen Yeh
- Departments of Surgery and Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina System, Chapel Hill, North Carolina, USA
| | - Brian A. Belt
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - David C. Linehan
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Department of Surgery, Division of Surgical Oncology, University of Rochester Medical Center, Rochester, New York, USA
| | - Roberto Hernandez-Alejandro
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Division of Solid Organ Transplant Surgery, Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
31
|
Assouline B, Kahn R, Hodali L, Condiotti R, Engel Y, Elyada E, Mordechai-Heyn T, Pitarresi JR, Atias D, Steinberg E, Bidany-Mizrahi T, Forkosh E, Katz LH, Benny O, Golan T, Hofree M, Stewart SA, Atlan KA, Zamir G, Stanger BZ, Berger M, Ben-Porath I. Senescent cancer-associated fibroblasts in pancreatic adenocarcinoma restrict CD8 + T cell activation and limit responsiveness to immunotherapy in mice. Nat Commun 2024; 15:6162. [PMID: 39039076 PMCID: PMC11263607 DOI: 10.1038/s41467-024-50441-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 07/05/2024] [Indexed: 07/24/2024] Open
Abstract
Senescent cells within tumors and their stroma exert complex pro- and anti-tumorigenic functions. However, the identities and traits of these cells, and the potential for improving cancer therapy through their targeting, remain poorly characterized. Here, we identify a senescent subset within previously-defined cancer-associated fibroblasts (CAFs) in pancreatic ductal adenocarcinomas (PDAC) and in premalignant lesions in mice and humans. Senescent CAFs isolated from mouse and humans expressed elevated levels of immune-regulatory genes. Depletion of senescent CAFs, either genetically or using the Bcl-2 inhibitor ABT-199 (venetoclax), increased the proportion of activated CD8+ T cells in mouse pancreatic carcinomas, whereas induction of CAF senescence had the opposite effect. Combining ABT-199 with an immune checkpoint therapy regimen significantly reduced mouse tumor burden. These results indicate that senescent CAFs in PDAC stroma limit the numbers of activated cytotoxic CD8+ T cells, and suggest that their targeted elimination through senolytic treatment may enhance immunotherapy.
Collapse
Grants
- R01 CA217208 NCI NIH HHS
- R01 CA276512 NCI NIH HHS
- R00 CA252153 NCI NIH HHS
- 2621/18 Israel Science Foundation (ISF)
- R01 CA252225 NCI NIH HHS
- R01 AG059244 NIA NIH HHS
- Israel Cancer Research Fund (Israel Cancer Research Fund, Inc.)
- This study was supported by grants from the Israel Science Foundation - Broad Institute program (2621/18, I.B.-P.), Israel Precision Medicine Partnership (3755/21, I.B.-P.), Israel Science Foundation Mid-Career Program (1923/22, I.B.-P.), the Israel Ministry of Science and Technology DKFZ-MOST program (4062, I.B.-P.), the Chief Scientist of the Israel Ministry of Health (3-15017, I.B.-P.), the Alex U. Soyka Program (I.B.-P., B.A., R.K., L.H.), the Israel Cancer Research Fund International Collaboration Program (I.B.-P)
Collapse
Affiliation(s)
- Benjamin Assouline
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rachel Kahn
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lutfi Hodali
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Reba Condiotti
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yarden Engel
- The Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ela Elyada
- Department of Biochemistry, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tzlil Mordechai-Heyn
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Surgery, Hadassah Medical Center, and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jason R Pitarresi
- Division of Hematology-Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Molecular Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Dikla Atias
- Pancreatic Cancer Translational Research Laboratory, Oncology Institute, Sheba Medical Center, Tel Hashomer, Israel, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eliana Steinberg
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tirza Bidany-Mizrahi
- The Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Esther Forkosh
- Department of Gastroenterology, Hadassah Medical Center, and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lior H Katz
- Department of Gastroenterology, Hadassah Medical Center, and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ofra Benny
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Talia Golan
- Pancreatic Cancer Translational Research Laboratory, Oncology Institute, Sheba Medical Center, Tel Hashomer, Israel, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Matan Hofree
- The Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sheila A Stewart
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Karine A Atlan
- Department of Pathology, Hadassah Medical Center, and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gideon Zamir
- Department of Surgery, Hadassah Medical Center, and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ben Z Stanger
- Department of Medicine and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael Berger
- The Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ittai Ben-Porath
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
32
|
Struth E, Labaf M, Karimnia V, Liu Y, Cramer G, Dahl JB, Slack FJ, Zarringhalam K, Celli JP. Drug resistant pancreatic cancer cells exhibit altered biophysical interactions with stromal fibroblasts in imaging studies of 3D co-culture models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.14.602133. [PMID: 39071263 PMCID: PMC11275726 DOI: 10.1101/2024.07.14.602133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Interactions between tumor and stromal cells are well known to play a prominent roles in progression of pancreatic ductal adenocarcinoma (PDAC). As knowledge of stromal crosstalk in PDAC has evolved, it has become clear that cancer associated fibroblasts can play both tumor promoting and tumor suppressive roles through a combination of paracrine crosstalk and juxtacrine interactions involving direct physical contact. Another major contributor to dismal survival statistics for PDAC is development of resistance to chemotherapy drugs. Though less is known about how the acquisition of chemoresistance impacts upon tumor-stromal crosstalk. Here, we use 3D co-culture geometries to recapitulate juxtacrine interactions between epithelial and stromal cells. In particular, extracellular matrix (ECM) overlay cultures in which stromal cells (pancreatic stellate cells, or normal human fibroblasts) are placed adjacent to PDAC cells (PANC1), result in direct heterotypic cell adhesions accompanied by dramatic fibroblast contractility which leads to highly condensed macroscopic multicellular aggregates as detected using particle image velocimetry (PIV) analysis to quantify cell velocities over the course of time lapse movie sequences. To investigate how drug resistance impacts these juxtacrine interactions we contrast cultures in which PANC1 are substituted with a drug resistant subline (PANC1-OR) previously established in our lab. We find that heterotypic cell-cell interactions are highly suppressed in drug-resistant cells relative to the parental PANC1 cells. To investigate further we conduct RNA-seq and bioinformatics analysis to identify differential gene expression in PANC1 and PANC1-OR, which shows that negative regulation of cell adhesion molecules, consistent with increased epithelial mesenchymal transition (EMT), is also consistent with loss of hetrotypic cell-cell contact necessary for the contractile behavior observed in drug naïve cultures. Overall these findings elucidate the role of drug-resistance in inhibiting an avenue of stromal crosstalk which is associated with tumor suppression and also help to establish cell culture conditions useful for further mechanistic investigation.
Collapse
|
33
|
Elebo N, Abdel-Shafy EA, Omoshoro-Jones JAO, Nsingwane Z, Hussein AAA, Smith M, Candy G, Cacciatore S, Fru P, Nweke EE. Comparative immune profiling of pancreatic ductal adenocarcinoma progression among South African patients. BMC Cancer 2024; 24:809. [PMID: 38973003 PMCID: PMC11229237 DOI: 10.1186/s12885-024-12595-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 07/02/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND Pancreatic Ductal Adenocarcinoma (PDAC) is an aggressive cancer characterized by an immunosuppressive microenvironment. Patients from specific ethnicities and population groups have poorer prognoses than others. Therefore, a better understanding of the immune landscape in such groups is necessary for disease elucidation, predicting patient outcomes and therapeutic targeting. This study investigated the expression of circulating key immune cell markers in South African PDAC patients of African ancestry. METHODS Blood samples were obtained from a total of 6 healthy volunteers (HC), 6 Chronic Pancreatitis (CP) and 34 PDAC patients consisting of 22 resectable (RPC), 8 locally advanced (LAPC) and 4 metastatic (MPC). Real-time Quantitative Polymerase Chain reactions (RT-qPCR), Metabolomics, Enzyme-Linked Immunosorbent Assay (ELISA), Reactive Oxygen Species (ROS), and Immunophenotyping assays were conducted. Statistical analysis was conducted in R (v 4.3.2). Additional analysis of single-cell RNA data from 20 patients (16 PDAC and 4 controls) was conducted to interrogate the distribution of T-cell and Natural Killer cell populations. RESULTS Granulocyte and neutrophil levels were significantly elevated while lymphocytes decreased with PDAC severity. The total percentages of CD3 T-cell subpopulations (helper and double negative T-cells) decreased when compared to HC. Although both NK (p = 0.014) and NKT (p < 0.001) cell levels increased as the disease progressed, their subsets: NK CD56dimCD16- (p = 0.024) and NKTs CD56+ (p = 0.008) cell levels reduced significantly. Of note is the negative association of NK CD56dimCD16- (p < 0.001) cell levels with survival time. The gene expression analyses showed no statistically significant correlation when comparing the PDAC groups with the controls. The inflammatory status of PDAC was assessed by ROS levels of serum which were elevated in CP (p = 0.025), (RPC (p = 0.003) and LAPC (p = 0.008)) while no significant change was observed in MPC, compared to the HC group. ROS was shown to be positively correlated with GlycA (R = 0.45, p = 0.0096). Single-cell analyses showed a significant difference in the ratio of NKT cells per total cell counts in LAPC (p < 0.001) and MPC (p < 0.001) groups compared with HC, confirming observations in our sample group. CONCLUSION The expression of these immune cell markers observed in this pilot study provides insight into their potential roles in tumour progression in the patient group and suggests their potential utility in the development of immunotherapeutic strategies.
Collapse
Affiliation(s)
- Nnenna Elebo
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2193, South Africa
- Bioinformatics Unit, International Centre for Genetic Engineering and Biotechnology, Observatory, Cape Town, 7925, South Africa
| | - Ebtesam A Abdel-Shafy
- Bioinformatics Unit, International Centre for Genetic Engineering and Biotechnology, Observatory, Cape Town, 7925, South Africa
- National Research Centre, Cairo, Egypt
| | - Jones A O Omoshoro-Jones
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2193, South Africa
- Hepatopancreatobiliary Unit, Department of Surgery, Chris Hani-Baragwanath Academic Hospital, Soweto Johannesburg, South Africa
| | - Zanele Nsingwane
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2193, South Africa
| | - Ahmed A A Hussein
- Bioinformatics Unit, International Centre for Genetic Engineering and Biotechnology, Observatory, Cape Town, 7925, South Africa
- Theodore Bilharz Research Institute, Giza, Egypt
| | - Martin Smith
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2193, South Africa
- Hepatopancreatobiliary Unit, Department of Surgery, Chris Hani-Baragwanath Academic Hospital, Soweto Johannesburg, South Africa
| | - Geoffrey Candy
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2193, South Africa
| | - Stefano Cacciatore
- Bioinformatics Unit, International Centre for Genetic Engineering and Biotechnology, Observatory, Cape Town, 7925, South Africa
| | - Pascaline Fru
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2193, South Africa
| | - Ekene Emmanuel Nweke
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2193, South Africa.
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Florida, Roodepoort, South Africa.
| |
Collapse
|
34
|
Belle JI, Sen D, Baer JM, Liu X, Lander VE, Ye J, Sells BE, Knolhoff BL, Faiz A, Kang LI, Qian G, Fields RC, Ding L, Kim H, Provenzano PP, Stewart SA, DeNardo DG. Senescence Defines a Distinct Subset of Myofibroblasts That Orchestrates Immunosuppression in Pancreatic Cancer. Cancer Discov 2024; 14:1324-1355. [PMID: 38683144 DOI: 10.1158/2159-8290.cd-23-0428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 01/29/2024] [Accepted: 03/08/2024] [Indexed: 05/01/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) therapeutic resistance is largely attributed to a unique tumor microenvironment embedded with an abundance of cancer-associated fibroblasts (CAF). Distinct CAF populations were recently identified, but the phenotypic drivers and specific impact of CAF heterogeneity remain unclear. In this study, we identify a subpopulation of senescent myofibroblastic CAFs (SenCAF) in mouse and human PDAC. These SenCAFs are a phenotypically distinct subset of myofibroblastic CAFs that localize near tumor ducts and accumulate with PDAC progression. To assess the impact of endogenous SenCAFs in PDAC, we used an LSL-KRASG12D;p53flox;p48-CRE;INK-ATTAC (KPPC-IA) mouse model of spontaneous PDAC with inducible senescent cell depletion. Depletion of senescent stromal cells in genetic and pharmacologic PDAC models relieved immune suppression by macrophages, delayed tumor progression, and increased responsiveness to chemotherapy. Collectively, our findings demonstrate that SenCAFs promote PDAC progression and immune cell dysfunction. Significance: CAF heterogeneity in PDAC remains poorly understood. In this study, we identify a novel subpopulation of senescent CAFs that promotes PDAC progression and immunosuppression. Targeting CAF senescence in combination therapies could increase tumor vulnerability to chemo or immunotherapy. See related article by Ye et al., p. 1302.
Collapse
Affiliation(s)
- Jad I Belle
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Devashish Sen
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - John M Baer
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Xiuting Liu
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Varintra E Lander
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Jiayu Ye
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Blake E Sells
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Brett L Knolhoff
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Ahmad Faiz
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Liang-I Kang
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Guhan Qian
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
- Department of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Ryan C Fields
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Li Ding
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Hyun Kim
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Paolo P Provenzano
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
- Department of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Sheila A Stewart
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - David G DeNardo
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
35
|
Chen C, Jiang YP, You I, Gray NS, Lin RZ. Down-Regulation of AKT Proteins Slows the Growth of Mutant-KRAS Pancreatic Tumors. Cells 2024; 13:1061. [PMID: 38920688 PMCID: PMC11202146 DOI: 10.3390/cells13121061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
Serine/threonine kinase AKT isoforms play a well-established role in cell metabolism and growth. Most pancreatic adenocarcinomas (PDACs) harbor activation mutations of KRAS, which activates the PI3K/AKT signaling pathway. However, AKT inhibitors are not effective in the treatment of pancreatic cancer. To better understand the role of AKT signaling in mutant-KRAS pancreatic tumors, this study utilized proteolysis-targeting chimeras (PROTACs) and CRISPR-Cas9-genome editing to investigate AKT proteins. The PROTAC down-regulation of AKT proteins markedly slowed the growth of three pancreatic tumor cell lines harboring mutant KRAS. In contrast, the inhibition of AKT kinase activity alone had very little effect on the growth of these cell lines. The concurrent genetic deletion of all AKT isoforms (AKT1, AKT2, and AKT3) in the KPC (KrasG12D; Trp53R172H; Pdx1-Cre) pancreatic cancer cell line also dramatically slowed its growth in vitro and when orthotopically implanted in syngeneic mice. Surprisingly, insulin-like growth factor-1 (IGF-1), but not epidermal growth factor (EGF), restored KPC cell growth in serum-deprived conditions, and the IGF-1 growth stimulation effect was AKT-dependent. The RNA-seq analysis of AKT1/2/3-deficient KPC cells suggested that reduced cholesterol synthesis may be responsible for the decreased response to IGF-1 stimulation. These results indicate that the presence of all three AKT isoforms supports pancreatic tumor cell growth, and the pharmacological degradation of AKT proteins may be more effective than AKT catalytic inhibitors for treating pancreatic cancer.
Collapse
Affiliation(s)
- Chuankai Chen
- Department of Physiology & Biophysics, Stony Brook University, Stony Brook, NY 11794, USA; (C.C.); (Y.-P.J.)
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY 11790, USA
| | - Ya-Ping Jiang
- Department of Physiology & Biophysics, Stony Brook University, Stony Brook, NY 11794, USA; (C.C.); (Y.-P.J.)
| | - Inchul You
- Department of Chemical and Systems Biology, ChEM-H, and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA; (I.Y.); (N.S.G.)
| | - Nathanael S. Gray
- Department of Chemical and Systems Biology, ChEM-H, and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA; (I.Y.); (N.S.G.)
| | - Richard Z. Lin
- Department of Physiology & Biophysics, Stony Brook University, Stony Brook, NY 11794, USA; (C.C.); (Y.-P.J.)
- Northport VA Medical Center, Northport, NY 11768, USA
| |
Collapse
|
36
|
Chen C, Jiang YP, You I, Gray NS, Lin RZ. Down-regulation of AKT proteins slows the growth of mutant-KRAS pancreatic tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592345. [PMID: 38746217 PMCID: PMC11092743 DOI: 10.1101/2024.05.03.592345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Serine/threonine kinase AKT isoforms play a well-established role in cell metabolism and growth. Most pancreatic adenocarcinoma (PDAC) harbors activation mutations of KRAS, which activates the PI3K/AKT signaling pathway. However, AKT inhibitors are not effective in the treatment of pancreatic cancer. To better understand the role of AKT signaling in mutant-KRAS pancreatic tumors, this study utilizes proteolysis-targeting chimeras (PROTACs) and CRISPR-Cas9-genome editing to investigate AKT proteins. PROTAC down-regulation of AKT proteins markedly slowed the growth of three pancreatic tumor cell lines harboring mutant KRAS. In contrast, inhibition of AKT kinase activity alone had very little effect on the growth of these cell lines. Concurrent genetic deletion of all AKT isoforms (AKT1, AKT2, and AKT3) in the KPC (KrasG12D; Trp53R172H; Pdx1-Cre) pancreatic cancer cell line also dramatically slowed its growth in vitro and when orthotopically implanted in syngeneic mice. Surprisingly, insulin-like growth factor-1 (IGF-1), but not epidermal growth factor (EGF), restored KPC cell growth in serum-deprived conditions and the IGF-1 growth stimulation effect was AKT dependent. RNA-seq analysis of AKT1/2/3-deficient KPC cells suggested that reduced cholesterol synthesis may be responsible for the decreased response to IGF-1 stimulation. These results indicate that the presence of all three AKT isoforms supports pancreatic tumor cell growth and pharmacological degradation of AKT proteins may be more effective than AKT catalytic inhibitors for treating pancreatic cancer.
Collapse
Affiliation(s)
- Chuankai Chen
- Department of Physiology & Biophysics, Stony Brook University, Stony Brook, New York, USA
- Graduate Program in Genetics, Stony Brook University, New York, USA
| | - Ya-Ping Jiang
- Department of Physiology & Biophysics, Stony Brook University, Stony Brook, New York, USA
| | - Inchul You
- Department of Chemical and Systems Biology, ChEM-H, and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, California 94305, USA
| | - Nathanael S. Gray
- Department of Chemical and Systems Biology, ChEM-H, and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, California 94305, USA
| | - Richard Z. Lin
- Department of Physiology & Biophysics, Stony Brook University, Stony Brook, New York, USA
- Northport VA Medical Center, Northport, New York, USA
| |
Collapse
|
37
|
An M, Mehta A, Min BH, Heo YJ, Wright SJ, Parikh M, Bi L, Lee H, Kim TJ, Lee SY, Moon J, Park RJ, Strickland MR, Park WY, Kang WK, Kim KM, Kim ST, Klempner SJ, Lee J. Early Immune Remodeling Steers Clinical Response to First-Line Chemoimmunotherapy in Advanced Gastric Cancer. Cancer Discov 2024; 14:766-785. [PMID: 38319303 PMCID: PMC11061611 DOI: 10.1158/2159-8290.cd-23-0857] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/28/2023] [Accepted: 02/02/2024] [Indexed: 02/07/2024]
Abstract
Adding anti-programmed cell death protein 1 (anti-PD-1) to 5-fluorouracil (5-FU)/platinum improves survival in some advanced gastroesophageal adenocarcinomas (GEA). To understand the effects of chemotherapy and immunotherapy, we conducted a phase II first-line trial (n = 47) sequentially adding pembrolizumab to 5-FU/platinum in advanced GEA. Using serial biopsy of the primary tumor at baseline, after one cycle of 5-FU/platinum, and after the addition of pembrolizumab, we transcriptionally profiled 358,067 single cells to identify evolving multicellular tumor microenvironment (TME) networks. Chemotherapy induced early on-treatment multicellular hubs with tumor-reactive T-cell and M1-like macrophage interactions in slow progressors. Faster progression featured increased MUC5A and MSLN containing treatment resistance programs in tumor cells and M2-like macrophages with immunosuppressive stromal interactions. After pembrolizumab, we observed increased CD8 T-cell infiltration and development of an immunity hub involving tumor-reactive CXCL13 T-cell program and epithelial interferon-stimulated gene programs. Strategies to drive increases in antitumor immune hub formation could expand the portion of patients benefiting from anti-PD-1 approaches. SIGNIFICANCE The benefit of 5-FU/platinum with anti-PD-1 in first-line advanced gastric cancer is limited to patient subgroups. Using a trial with sequential anti-PD-1, we show coordinated induction of multicellular TME hubs informs the ability of anti-PD-1 to potentiate T cell-driven responses. Differential TME hub development highlights features that underlie clinical outcomes. This article is featured in Selected Articles from This Issue, p. 695.
Collapse
Affiliation(s)
- Minae An
- Experimental Therapeutics Development Center, Samsung Medical Center, Seoul, Korea
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Arnav Mehta
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Byung Hoon Min
- Department of Medicine, Division of Gastroenterology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | - Samuel J. Wright
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Milan Parikh
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Lynn Bi
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Hyuk Lee
- Department of Medicine, Division of Gastroenterology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Tae Jun Kim
- Department of Medicine, Division of Gastroenterology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Song-Yi Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeonghyeon Moon
- Departments of Neurology and Immunology, Yale School of Medicine, New Haven, Connecticut
| | - Ryan J. Park
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Division of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Matthew R. Strickland
- Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | | | - Won Ki Kang
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seung Tae Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Samuel J. Klempner
- Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Jeeyun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
38
|
Wehrli M, Guinn S, Birocchi F, Kuo A, Sun Y, Larson RC, Almazan AJ, Scarfò I, Bouffard AA, Bailey SR, Anekal PV, Llopis PM, Nieman LT, Song Y, Xu KH, Berger TR, Kann MC, Leick MB, Silva H, Salas-Benito D, Kienka T, Grauwet K, Armstrong TD, Zhang R, Zhu Q, Fu J, Schmidts A, Korell F, Jan M, Choi BD, Liss AS, Boland GM, Ting DT, Burkhart RA, Jenkins RW, Zheng L, Jaffee EM, Zimmerman JW, Maus MV. Mesothelin CAR T Cells Secreting Anti-FAP/Anti-CD3 Molecules Efficiently Target Pancreatic Adenocarcinoma and its Stroma. Clin Cancer Res 2024; 30:1859-1877. [PMID: 38393682 PMCID: PMC11062832 DOI: 10.1158/1078-0432.ccr-23-3841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
PURPOSE Targeting solid tumors with chimeric antigen receptor (CAR) T cells remains challenging due to heterogenous target antigen expression, antigen escape, and the immunosuppressive tumor microenvironment (TME). Pancreatic cancer is characterized by a thick stroma generated by cancer-associated fibroblasts (CAF), which may contribute to the limited efficacy of mesothelin-directed CAR T cells in early-phase clinical trials. To provide a more favorable TME for CAR T cells to target pancreatic ductal adenocarcinoma (PDAC), we generated T cells with an antimesothelin CAR and a secreted T-cell-engaging molecule (TEAM) that targets CAF through fibroblast activation protein (FAP) and engages T cells through CD3 (termed mesoFAP CAR-TEAM cells). EXPERIMENTAL DESIGN Using a suite of in vitro, in vivo, and ex vivo patient-derived models containing cancer cells and CAF, we examined the ability of mesoFAP CAR-TEAM cells to target PDAC cells and CAF within the TME. We developed and used patient-derived ex vivo models, including patient-derived organoids with patient-matched CAF and patient-derived organotypic tumor spheroids. RESULTS We demonstrated specific and significant binding of the TEAM to its respective antigens (CD3 and FAP) when released from mesothelin-targeting CAR T cells, leading to T-cell activation and cytotoxicity of the target cell. MesoFAP CAR-TEAM cells were superior in eliminating PDAC and CAF compared with T cells engineered to target either antigen alone in our ex vivo patient-derived models and in mouse models of PDAC with primary or metastatic liver tumors. CONCLUSIONS CAR-TEAM cells enable modification of tumor stroma, leading to increased elimination of PDAC tumors. This approach represents a promising treatment option for pancreatic cancer.
Collapse
Affiliation(s)
- Marc Wehrli
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Samantha Guinn
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Filippo Birocchi
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Adam Kuo
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Yi Sun
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Rebecca C. Larson
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Antonio J. Almazan
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Irene Scarfò
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Amanda A. Bouffard
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Stefanie R. Bailey
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | | | | | - Linda T. Nieman
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Yuhui Song
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Katherine H. Xu
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Trisha R. Berger
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Michael C. Kann
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Mark B. Leick
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Blood and Marrow Transplant Program, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Harrison Silva
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Diego Salas-Benito
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Tamina Kienka
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Korneel Grauwet
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Todd D. Armstrong
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Rui Zhang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Qingfeng Zhu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Juan Fu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Andrea Schmidts
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Felix Korell
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Max Jan
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School; Boston, MA, USA
| | - Bryan D. Choi
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School; Boston, MA, USA
| | - Andrew S. Liss
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Genevieve M. Boland
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School; Boston, MA, USA
| | - David T. Ting
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Richard A. Burkhart
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Russell W. Jenkins
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Elizabeth M. Jaffee
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Jacquelyn W. Zimmerman
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Marcela V. Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Blood and Marrow Transplant Program, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| |
Collapse
|
39
|
Bhartiya D, Raouf S, Pansare K, Tripathi A, Tripathi A. Initiation of Cancer: The Journey From Mutations in Somatic Cells to Epigenetic Changes in Tissue-resident VSELs. Stem Cell Rev Rep 2024; 20:857-880. [PMID: 38457060 DOI: 10.1007/s12015-024-10694-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2024] [Indexed: 03/09/2024]
Abstract
Multiple theories exist to explain cancer initiation, although a consensus on this is crucial for developing effective therapies. 'Somatic mutation theory' suggests that mutations in somatic cells during DNA repair initiates cancer but this concept has several attached paradoxes. Research efforts to identify quiescent cancer stem cells (CSCs) that survive therapy and result in metastasis and recurrence have remained futile. In solid cancers, CSCs are suggested to appear during epithelial-mesenchymal transition by the dedifferentiation and reprogramming of epithelial cells. Pluripotent and quiescent very small embryonic-like stem cells (VSELs) exist in multiple tissues but remain elusive owing to their small size and scarce nature. VSELs are developmentally connected to primordial germ cells, undergo rare, asymmetrical cell divisions and are responsible for the regular turnover of cells to maintain tissue homeostasis throughout life. VSELs are directly vulnerable to extrinsic endocrine insults because they express gonadal and gonadotropin hormone receptors. VSELs undergo epigenetic changes due to endocrine insults and transform into CSCs. CSCs exhibit genomic instability and develop mutations due to errors during DNA replication while undergoing excessive proliferation and clonal expansion to form spheroids. Thus tissue-resident VSELs offer a connection between extrinsic insults and variations in cancer incidence reported in various body tissues. To conclude, cancer is indeed a stem cell disease with mutations occurring as a consequence. In addition to immunotherapy, targeting mutations, and Lgr5 + organoids for developing new therapeutics, targeting CSCs (epigenetically altered VSELs) by improving their niche and epigenetic status could serve as a promising strategy to treat cancer.
Collapse
Affiliation(s)
- Deepa Bhartiya
- Epigeneres Biotech Pvt Ltd, Todi Mill Compound, Senapati Bapat Marg, Lower Parel, 400013, Mumbai, India.
| | | | - Kshama Pansare
- Epigeneres Biotech Pvt Ltd, Todi Mill Compound, Senapati Bapat Marg, Lower Parel, 400013, Mumbai, India
| | - Anish Tripathi
- Epigeneres Biotech Pvt Ltd, Todi Mill Compound, Senapati Bapat Marg, Lower Parel, 400013, Mumbai, India
| | - Ashish Tripathi
- Epigeneres Biotech Pvt Ltd, Todi Mill Compound, Senapati Bapat Marg, Lower Parel, 400013, Mumbai, India
- 23Ikigai Pte Ltd, 30 Cecil Street, #21-08 Prudentsial Tower, Singapore, 049712, Singapore
| |
Collapse
|
40
|
Parte S, Kaur AB, Nimmakayala RK, Ogunleye AO, Chirravuri R, Vengoji R, Leon F, Nallasamy P, Rauth S, Alsafwani ZW, Lele S, Cox JL, Bhat I, Singh S, Batra SK, Ponnusamy MP. Cancer-Associated Fibroblast Induces Acinar-to-Ductal Cell Transdifferentiation and Pancreatic Cancer Initiation Via LAMA5/ITGA4 Axis. Gastroenterology 2024; 166:842-858.e5. [PMID: 38154529 PMCID: PMC11694316 DOI: 10.1053/j.gastro.2023.12.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 12/09/2023] [Accepted: 12/19/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND & AIMS Pancreatic ductal adenocarcinoma (PDAC) is characterized by desmoplastic stroma surrounding most tumors. Activated stromal fibroblasts, namely cancer-associated fibroblasts (CAFs), play a major role in PDAC progression. We analyzed whether CAFs influence acinar cells and impact PDAC initiation, that is, acinar-to-ductal metaplasia (ADM). ADM connection with PDAC pathophysiology is indicated, but not yet established. We hypothesized that CAF secretome might play a significant role in ADM in PDAC initiation. METHODS Mouse and human acinar cell organoids, acinar cells cocultured with CAFs and exposed to CAF-conditioned media, acinar cell explants, and CAF cocultures were examined by means of quantitative reverse transcription polymerase chain reaction, RNA sequencing, immunoblotting, and confocal microscopy. Data from liquid chromatography with tandem mass spectrometry analysis of CAF-conditioned medium and RNA sequencing data of acinar cells post-conditioned medium exposure were integrated using bioinformatics tools to identify the molecular mechanism for CAF-induced ADM. Using confocal microscopy, immunoblotting, and quantitative reverse transcription polymerase chain reaction analysis, we validated the depletion of a key signaling axis in the cell line, acinar explant coculture, and mouse cancer-associated fibroblasts (mCAFs). RESULTS A close association of acino-ductal markers (Ulex europaeus agglutinin 1, amylase, cytokeratin-19) and mCAFs (α-smooth muscle actin) in LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx1Cre (KPC) and LSL-KrasG12D/+; Pdx1Cre (KC) autochthonous progression tumor tissue was observed. Caerulein treatment-induced mCAFs increased cytokeratin-19 and decreased amylase in wild-type and KC pancreas. Likewise, acinar-mCAF cocultures revealed the induction of ductal transdifferentiation in cell line, acinar-organoid, and explant coculture formats in WT and KC mice pancreas. Proteomic and transcriptomic data integration revealed a novel laminin α5/integrinα4/stat3 axis responsible for CAF-mediated acinar-to-ductal cell transdifferentiation. CONCLUSIONS Results collectively suggest the first evidence for CAF-influenced acino-ductal phenotypic switchover, thus highlighting the tumor microenvironment role in pancreatic carcinogenesis inception.
Collapse
Affiliation(s)
- Seema Parte
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska
| | - Annant B Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska
| | - Rama Krishna Nimmakayala
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska
| | - Ayoola O Ogunleye
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska
| | - Ramakanth Chirravuri
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska
| | - Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska
| | - Frank Leon
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska
| | - Palanisamy Nallasamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska
| | - Sanchita Rauth
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska
| | - Zahraa Wajih Alsafwani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska
| | - Subodh Lele
- Department of Pathology and Microbiology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska
| | - Jesse L Cox
- Department of Pathology and Microbiology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska
| | - Ishfaq Bhat
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Nebraska Medical Center at Omaha, Omaha, Nebraksa
| | - Shailender Singh
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Nebraska Medical Center at Omaha, Omaha, Nebraksa
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center at Omaha, Omaha, Nebraska.
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center at Omaha, Omaha, Nebraska.
| |
Collapse
|
41
|
Uehara M, Domoto T, Takenaka S, Takeuchi O, Shimasaki T, Miyashita T, Minamoto T. Glycogen synthase kinase 3β: the nexus of chemoresistance, invasive capacity, and cancer stemness in pancreatic cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:4. [PMID: 38318525 PMCID: PMC10838383 DOI: 10.20517/cdr.2023.84] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/20/2023] [Accepted: 01/17/2024] [Indexed: 02/07/2024]
Abstract
The treatment of pancreatic cancer remains a significant clinical challenge due to the limited number of patients eligible for curative (R0) surgery, failures in the clinical development of targeted and immune therapies, and the pervasive acquisition of chemotherapeutic resistance. Refractory pancreatic cancer is typified by high invasiveness and resistance to therapy, with both attributes related to tumor cell stemness. These malignant characteristics mutually enhance each other, leading to rapid cancer progression. Over the past two decades, numerous studies have produced evidence of the pivotal role of glycogen synthase kinase (GSK)3β in the progression of over 25 different cancer types, including pancreatic cancer. In this review, we synthesize the current knowledge on the pathological roles of aberrant GSK3β in supporting tumor cell proliferation and invasion, as well as its contribution to gemcitabine resistance in pancreatic cancer. Importantly, we discuss the central role of GSK3β as a molecular hub that mechanistically connects chemoresistance, tumor cell invasion, and stemness in pancreatic cancer. We also discuss the involvement of GSK3β in the formation of desmoplastic tumor stroma and in promoting anti-cancer immune evasion, both of which constitute major obstacles to successful cancer treatment. Overall, GSK3β has characteristics of a promising therapeutic target to overcome chemoresistance in pancreatic cancer.
Collapse
Affiliation(s)
- Masahiro Uehara
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Authors contributed equally
| | - Takahiro Domoto
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Authors contributed equally
| | - Satoshi Takenaka
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
- Department of Surgery, Toyama City Hospital, Toyama 939-8511, Japan
| | - Osamu Takeuchi
- Biomedical Laboratory, Department of Research, Kitasato University Kitasato Institute Hospital, Tokyo 108-8642, Japan
| | - Takeo Shimasaki
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Medical Research Institute, Kanazawa Medical University, Uchinada 920-0293, Japan
| | - Tomoharu Miyashita
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
- Department of Surgery, Toyama City Hospital, Toyama 939-8511, Japan
| | - Toshinari Minamoto
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
| |
Collapse
|
42
|
Luo W, Wen T, Qu X. Tumor immune microenvironment-based therapies in pancreatic ductal adenocarcinoma: time to update the concept. J Exp Clin Cancer Res 2024; 43:8. [PMID: 38167055 PMCID: PMC10759657 DOI: 10.1186/s13046-023-02935-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal solid tumors. The tumor immune microenvironment (TIME) formed by interactions among cancer cells, immune cells, cancer-associated fibroblasts (CAF), and extracellular matrix (ECM) components drives PDAC in a more immunosuppressive direction: this is a major cause of therapy resistance and poor prognosis. In recent years, research has advanced our understanding of the signaling mechanism by which TIME components interact with the tumor and the evolution of immunophenotyping. Through revolutionary technologies such as single-cell sequencing, we have gone from simply classifying PDACs as "cold" and "hot" to a more comprehensive approach of immunophenotyping that considers all the cells and matrix components. This is key to improving the clinical efficacy of PDAC treatments. In this review, we elaborate on various TIME components in PDAC, the signaling mechanisms underlying their interactions, and the latest research into PDAC immunophenotyping. A deep understanding of these network interactions will contribute to the effective combination of TIME-based therapeutic approaches, such as immune checkpoint inhibitors (ICI), adoptive cell therapy, therapies targeting myeloid cells, CAF reprogramming, and stromal normalization. By selecting the appropriate integrated therapies based on precise immunophenotyping, significant advances in the future treatment of PDAC are possible.
Collapse
Affiliation(s)
- Wenyu Luo
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, 110001, Liaoning, China
| | - Ti Wen
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, 110001, Liaoning, China.
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
43
|
Costa V, Giovannetti E, Lonardo E. Revolutionizing Cancer Treatment: Unveiling New Frontiers by Targeting the (Un)Usual Suspects. Cancers (Basel) 2023; 16:132. [PMID: 38201558 PMCID: PMC10778478 DOI: 10.3390/cancers16010132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
This Special Issue includes original articles and reviews on both established and innovative approaches to cancer targeting, showcased at the 29th IGB Workshop titled "Targeting the (un)usual suspects in cancer" "https://29thigbworkshop [...].
Collapse
Affiliation(s)
- Valerio Costa
- Institute of Genetics and Biophysics (IGB), National Research Council of Italy (CNR), 80131 Naples, Italy;
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam UMC, VU University, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands;
- Fondazione Pisana per la Scienza, San Giuliano Terme, 56124 Pisa, Italy
| | - Enza Lonardo
- Institute of Genetics and Biophysics (IGB), National Research Council of Italy (CNR), 80131 Naples, Italy;
| |
Collapse
|
44
|
Affiliation(s)
| | - Claus Jorgensen
- Systems Oncology, Cancer Research UK Manchester Institute, Manchester, UK
| |
Collapse
|
45
|
Rogers S, Charles A, Thomas RM. The Prospect of Harnessing the Microbiome to Improve Immunotherapeutic Response in Pancreatic Cancer. Cancers (Basel) 2023; 15:5708. [PMID: 38136254 PMCID: PMC10741649 DOI: 10.3390/cancers15245708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/24/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Pancreatic ductal adenocarcinoma cancer (PDAC) is projected to become the second leading cause of cancer-related death in the United States by 2030. Patients are often diagnosed with advanced disease, which explains the dismal 5-year median overall survival rate of ~12%. Immunotherapy has been successful in improving outcomes in the past decade for a variety of malignancies, including gastrointestinal cancers. However, PDAC is historically an immunologically "cold" tumor, one with an immunosuppressive environment and with restricted entry of immune cells that have limited the success of immunotherapy in these tumors. The microbiome, the intricate community of microorganisms present on and within humans, has been shown to contribute to many cancers, including PDAC. Recently, its role in tumor immunology and response to immunotherapy has generated much interest. Herein, the current state of the interaction of the microbiome and immunotherapy in PDAC is discussed with a focus on needed areas of study in order to harness the immune system to combat pancreatic cancer.
Collapse
Affiliation(s)
- Sherise Rogers
- Department of Medicine, Division of Hematology and Oncology, University of Florida College of Medicine, Gainesville, FL 32610, USA;
| | - Angel Charles
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA;
| | - Ryan M. Thomas
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA;
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL 32603, USA
| |
Collapse
|
46
|
Kessler L, Hirmas N, Pabst KM, Hamacher R, Ferdinandus J, Schaarschmidt BM, Milosevic A, Nader M, Umutlu L, Uhl W, Reinacher-Schick A, Lugnier C, Witte D, Niedergethmann M, Herrmann K, Fendler WP, Siveke JT. 68Ga-Labeled Fibroblast Activation Protein Inhibitor ( 68Ga-FAPI) PET for Pancreatic Adenocarcinoma: Data from the 68Ga-FAPI PET Observational Trial. J Nucl Med 2023; 64:1910-1917. [PMID: 37973185 DOI: 10.2967/jnumed.122.264827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 09/27/2023] [Indexed: 11/19/2023] Open
Abstract
The fibroblast activation protein (FAP) is highly expressed on carcinoma-associated fibroblasts in the stroma of pancreatic cancer and thus is a promising target for imaging and therapy. Preliminary data on PET imaging with radiolabeled FAP inhibitors (FAPIs) demonstrate superior tumor detection. Here we assess the accuracy of FAP-directed PET in patients with pancreatic cancer. Methods: Of 64 patients with suspected or proven pancreatic cancer, 62 (97%) were included in the data analysis of the 68Ga-FAPI PET observational trial (NCT04571086). All of these patients underwent contrast-enhanced CT, and 38 patients additionally underwent 18F-FDG PET. The primary study endpoint was the association of 68Ga-FAPI PET uptake intensity and histopathologic FAP expression. Secondary endpoints were detection rate, diagnostic performance, interreader reproducibility, and change in management. Datasets were interpreted by 2 masked readers. Results: The primary endpoint was met: The association between 68Ga-FAPI SUVmax and histopathologic FAP expression was significant (Spearman r, 0.48; P = 0.04). For histopathology-validated lesions, 68Ga-FAPI PET showed high sensitivity and positive predictive values (PPVs) on per-patient (sensitivity, 100%; PPV, 96.3%) and per-region (sensitivity, 100%; PPV, 97.0%) bases. In a head-to-head comparison versus 18F-FDG or contrast-enhanced CT, 68Ga-FAPI detected more tumor on a per-lesion (84.7% vs. 46.5% vs. 52.9%), per-patient (97.4% vs. 73.7% vs. 92.1%), or per-region (32.6% vs. 18.8% vs. 23.7%) basis, respectively. 68Ga-FAPI PET readers showed substantial overall agreement on the basis of the Fleiss κ: primary κ, 0.77 (range, 0.66-0.88). Minor and major changes in clinical management occurred in 5 patients (8.4%) after 68Ga-FAPI PET. Conclusion: We confirmed an association of 68Ga-FAPI PET SUVmax and histopathologic FAP expression in pancreatic cancer patients. Additionally, we found high detection rate and diagnostic accuracy, superior to those of 18F-FDG PET/CT. 68Ga-FAPI might become a powerful diagnostic tool for pancreatic cancer work-up.
Collapse
Affiliation(s)
- Lukas Kessler
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK) (Partner Site University Hospital Essen) and German Cancer Research Center (DKFZ), Essen, Germany
| | - Nader Hirmas
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK) (Partner Site University Hospital Essen) and German Cancer Research Center (DKFZ), Essen, Germany
| | - Kim M Pabst
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Rainer Hamacher
- German Cancer Consortium (DKTK) (Partner Site University Hospital Essen) and German Cancer Research Center (DKFZ), Essen, Germany
- Department of Medical Oncology, West German Cancer Center, University of Duisburg-Essen, Essen, Germany
| | - Justin Ferdinandus
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Benedikt M Schaarschmidt
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK) (Partner Site University Hospital Essen) and German Cancer Research Center (DKFZ), Essen, Germany
| | - Aleksandar Milosevic
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK) (Partner Site University Hospital Essen) and German Cancer Research Center (DKFZ), Essen, Germany
| | - Michael Nader
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Lale Umutlu
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK) (Partner Site University Hospital Essen) and German Cancer Research Center (DKFZ), Essen, Germany
| | - Waldemar Uhl
- Department of General and Visceral Surgery, St. Josef Hospital Bochum, Ruhr-University Bochum, Bochum, Germany
| | - Anke Reinacher-Schick
- Department of Hematology and Oncology with Palliative Care, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Celine Lugnier
- Department of General and Visceral Surgery, Alfried Krupp Hospital, Essen, Germany
| | - David Witte
- Department of General and Visceral Surgery, Alfried Krupp Hospital, Essen, Germany
| | - Marco Niedergethmann
- Department of General and Visceral Surgery, Alfried Krupp Hospital, Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK) (Partner Site University Hospital Essen) and German Cancer Research Center (DKFZ), Essen, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany;
- German Cancer Consortium (DKTK) (Partner Site University Hospital Essen) and German Cancer Research Center (DKFZ), Essen, Germany
| | - Jens T Siveke
- German Cancer Consortium (DKTK) (Partner Site University Hospital Essen) and German Cancer Research Center (DKFZ), Essen, Germany;
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; and
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK) (Partner Site University Hospital Essen) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
47
|
Zhang X, Liang B, Huang Y, Meng H, Li Z, Du J, Zhou L, Zhong Y, Wang B, Lin X, Yu G, Chen X, Lu W, Chen Z, Yang X, Huang Z. Behind the Indolent Facade: Uncovering the Molecular Features and Malignancy Potential in Lung Minimally Invasive Adenocarcinoma by Single-Cell Transcriptomics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303753. [PMID: 37991139 PMCID: PMC10754125 DOI: 10.1002/advs.202303753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/28/2023] [Indexed: 11/23/2023]
Abstract
The increased use of low-dose computed tomography screening has led to more frequent detection of early stage lung tumors, including minimally invasive adenocarcinoma (MIA). To unravel the intricacies of tumor cells and the immune microenvironment in MIA, this study performs a comprehensive single-cell transcriptomic analysis and profiles the transcriptomes of 156,447 cells from fresh paired MIA and invasive adenocarcinoma (IA) tumor samples, peripheral blood mononuclear cells, and adjacent normal tissue samples from three patients with synchronous multiple primary lung adenocarcinoma. This study highlights a connection and heterogeneity between the tumor ecosystem of MIA and IA. MIA tumor cells exhibited high expression of aquaporin-1 and angiotensin II receptor type 2 and a basal-like molecular character. Furthermore, it identifies that cathepsin B+ tumor-associated macrophages may over-activate CD8+ T cells in MIA, leading to an enrichment of granzyme K+ senescent CD8+ T cells, indicating the possibility of malignant progression behind the indolent appearance of MIA. These findings are further validated in 34 MIA and 35 IA samples by multiplexed immunofluorescence. These findings provide valuable insights into the mechanisms that maintain the indolent nature and prompt tumor progression of MIA and can be used to develop more effective therapeutic targets and strategies for MIA patients.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Thoracic SurgeryThe First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory HealthGuangzhou510140China
| | - Boxuan Liang
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Yuji Huang
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Hao Meng
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Zhiming Li
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Jiaxin Du
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Lang Zhou
- Department of BioinformaticsSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Yizhou Zhong
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Bo Wang
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Xi Lin
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Guangchuang Yu
- Department of BioinformaticsSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Xuewei Chen
- Department of Thoracic SurgeryThe First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory HealthGuangzhou510140China
| | - Weixiang Lu
- Department of Thoracic SurgeryThe First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory HealthGuangzhou510140China
| | - Zhe‐Sheng Chen
- College of Pharmacy and Health SciencesSt. John's UniversityQueensNY11439USA
| | - Xingfen Yang
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Zhenlie Huang
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| |
Collapse
|
48
|
Zheng S, Tian Q, Yuan Y, Sun S, Li T, Xia R, He R, Luo Y, Lin Q, Fu Z, Zhou Y, Chen R, Hu C. Extracellular vesicle-packaged circBIRC6 from cancer-associated fibroblasts induce platinum resistance via SUMOylation modulation in pancreatic cancer. J Exp Clin Cancer Res 2023; 42:324. [PMID: 38012734 PMCID: PMC10683239 DOI: 10.1186/s13046-023-02854-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/07/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) play pivotal roles in chemoresistance of pancreatic ductal adenocarcinoma (PDAC). However, the underlying mechanisms are poorly understood. Revealing the cross-talk network between tumor stroma and pancreatic cancer and developing effective strategies against oxaliplatin resistance are highly desired in the clinic. METHODS High-throughput sequence was used to screened the key circRNAs transmitted by extracellular vesicles (EVs) from CAFs to pancreatic cancer cells. The associations between EV-packaged circBIRC6 and chemotherapy responsiveness were validated in a cohort of 82 cases of advanced PDAC patients. Then, the effects of EV-packaged circBIRC6 on CAF-induced oxaliplatin resistance were investigated by flow cytometry, colony formation, viability of pancreatic cancer organoids in vitro and by xenograft models in vivo. RNA pulldown, RNA immunoprecipitation, and sites mutation assays were used to reveal the underlying mechanism. RESULTS We identified a circRNA, circBIRC6, is significantly upregulated in CAF-derived EVs and is positively associated with oxaliplatin-based chemoresistance. In vitro and in vivo functional assays showed that CAF-derived EV-packaged circBIRC6 enhance oxaliplatin resistance of pancreatic cancer cells and organoids via regulating the non-homologous end joining (NHEJ) dependent DNA repair. Mechanistically, circBIRC6 directly binds with XRCC4 and enhanced the interaction of XRCC4 with SUMO1 at the lysine 115 residue, which facilitated XRCC4 chromatin localization. XRCC4K115R mutation dramatically abrogated the EV-packaged circBIRC6 induced effect. Moreover, combination of antisense oligonucleotide inhibitors against circBIRC6 with Olaparib dramatically suppressed chemoresistance in patient-derived xenograft models. CONCLUSIONS Our study revealed that EV-packaged circBIRC6 confer oxaliplatin resistance in PDAC by mediating SUMOylation of XRCC4, introducing a promising predictive and therapeutic target for PDAC on oxaliplatin resistance.
Collapse
Affiliation(s)
- Shangyou Zheng
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Qing Tian
- School of medicine, South China University of Technology, Guangzhou, 510006, Guangdong Province, China
| | - Yuan Yuan
- Guangdong cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Shuxin Sun
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Tingting Li
- School of medicine, South China University of Technology, Guangzhou, 510006, Guangdong Province, China
| | - Renpeng Xia
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Rihua He
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China
- Shantou University Medical College, Shantou, 515041, Guangdong province, China
| | - Yuming Luo
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Qing Lin
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Zhiqiang Fu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China
- Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Yu Zhou
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China.
| | - Rufu Chen
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China.
- School of medicine, South China University of Technology, Guangzhou, 510006, Guangdong Province, China.
- Guangdong cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China.
| | - Chonghui Hu
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
49
|
Wu C, Zheng C, Chen S, He Z, Hua H, Sun C, Yu C. FOXQ1 promotes pancreatic cancer cell proliferation, tumor stemness, invasion and metastasis through regulation of LDHA-mediated aerobic glycolysis. Cell Death Dis 2023; 14:699. [PMID: 37875474 PMCID: PMC10598070 DOI: 10.1038/s41419-023-06207-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/19/2023] [Accepted: 09/29/2023] [Indexed: 10/26/2023]
Abstract
Pancreatic cancer (PC), a gastrointestinal tract malignant tumor, has a poor prognosis due to early metastasis and limited response to chemotherapy. Therefore, identifying novel therapeutic approaches for PC is critical. Epithelial-mesenchymal transition (EMT) is known as the vital progress in PC development, we constructed the EMT-related prognosis model to screen out that FOXQ1 probably involving in the EMT regulation. FOXQ1 has been linked to the malignant process in a number of cancers. However, its function in PC is unknown. In our work, the expression of FOXQ1 was elevated in PC tissues, and a high level of FOXQ1 in PC was linked to patients' poor prognosis. FOXQ1 overexpression promoted aerobic glycolysis and enhanced PC cell proliferation, tumor stemness, invasion, and metastasis. Whereas, FOXQ1 silencing showed the reverse effect. Furthermore, mechanistic studies indicated that FOXQ1 promotes LDHA transcription, and thus modulates aerobic glycolysis to enhance PC cell proliferation, tumor stemness, invasion, and metastasis by increasing LDHA expression. Therefore, these novel data suggest that FOXQ1 may be a possible therapeutic target in PC.
Collapse
Affiliation(s)
- Changhao Wu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, 550001, Guiyang, China
- College of Clinical Medicine, Guizhou Medical University, 550001, Guiyang, China
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, 550001, Guiyang, China
- Key Laboratory of Liver, Gallbladder, Pancreas and Spleen of Guizhou Medical University, 550001, Guiyang, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, 550004, Guiyang, Guizhou, China
| | - Chenglong Zheng
- Department of Hepatobiliary Surgery, Shenzhen Key Laboratory, Shenzhen University General Hospital, 518000, Shenzhen, China
| | - Shiyu Chen
- Department of Hepatic-Biliary-Pancreatic Surgery, South China Hospital, Medical School, Shenzhen University, 518116, Shenzhen, China
| | - Zhiwei He
- Department of Hepatobiliary Surgery, Shenzhen Key Laboratory, Shenzhen University General Hospital, 518000, Shenzhen, China
| | - Hao Hua
- College of Clinical Medicine, Guizhou Medical University, 550001, Guiyang, China
- Key Laboratory of Liver, Gallbladder, Pancreas and Spleen of Guizhou Medical University, 550001, Guiyang, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, 550004, Guiyang, Guizhou, China
| | - Chengyi Sun
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, 550001, Guiyang, China
- College of Clinical Medicine, Guizhou Medical University, 550001, Guiyang, China
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, 550001, Guiyang, China
- Key Laboratory of Liver, Gallbladder, Pancreas and Spleen of Guizhou Medical University, 550001, Guiyang, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, 550004, Guiyang, Guizhou, China
| | - Chao Yu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, 550001, Guiyang, China.
- College of Clinical Medicine, Guizhou Medical University, 550001, Guiyang, China.
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, 550001, Guiyang, China.
- Key Laboratory of Liver, Gallbladder, Pancreas and Spleen of Guizhou Medical University, 550001, Guiyang, China.
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, 550004, Guiyang, Guizhou, China.
| |
Collapse
|
50
|
Sun Y, Yao L, Man C, Gao Z, He R, Fan Y. Development and validation of cuproptosis-related lncRNAs associated with pancreatic cancer immune microenvironment based on single-cell. Front Immunol 2023; 14:1220760. [PMID: 37822927 PMCID: PMC10563513 DOI: 10.3389/fimmu.2023.1220760] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/12/2023] [Indexed: 10/13/2023] Open
Abstract
Background Cuproptosis, a novel mode of cell death associated with the tricarboxylic acid (TCA) cycle, is relevant to the development of cancer. However, the impact of single-cell-based Cuproptosis-associated lncRNAs on the Tumor immune microenvironment (TIME) of Pancreatic adenocarcinoma (PAAD) and its potential value for individualized immunotherapy has not been clarified. Methods 14 immune-related CRGs were screened by exploring the interaction between differentially expressed Immune-Related Genes (IRGs) and Cuproptosis-Related Genes (CRGs) in PAAD. Next, the expression amount and expression distribution of CRGs in single-cell samples were analyzed by focusing on 7-CRGs with significant expressions. On the one hand, MAP2K2, SOD1, and VEGFA, which were significantly differentially expressed between PAAD sites and normal tissues adjacent to them, were subjected to immunohistochemical validation and immune landscape analysis. On the other hand, from these 7-CRGs, prognostic signatures of lncRNAs were established by co-expression and LASSO-COX regression analysis, and their prognostic value and immune relevance were assessed. In addition, this study not only validated the hub CRGs and the lncRNAs constituting the signature in a PAAD animal model treated with immunotherapy-based combination therapy using immunohistochemistry and qRT-PCR but also explored the potential value of the combination of targeted, chemotherapy and immunotherapy. Results Based on the screening of 7-CRGs significantly expressed in a PAAD single-cell cohort and their co-expressed Cuproptosis-Related lncRNAs (CRIs), this study constructed a prognostic signature of 4-CRIs named CIR-score. A Nomogram integrating the CIR-score and clinical risk factors was constructed on this basis to predict the individualized survival of patients. Moreover, high and low-risk groups classified according to the median of signatures exhibited significant differences in clinical prognosis, immune landscape, bioenrichment, tumor burden, and drug sensitivity. And the immunohistochemical and qRT-PCR results of different mouse PAAD treatment strategies were consistent with the trend of inter-group variability in drug sensitivity of hub CRGs and CIR-score. The combination of immunotherapy, targeted therapy, and chemotherapy exhibited a better tumor suppression effect. Conclusion CIR-score, as a Cuproptosis-related TIME-specific prognostic signature based on PAAD single cells, not only predicts the prognosis and immune landscape of PAAD patients but also provides a new strategy for individualized immunotherapy-based combination therapy.
Collapse
Affiliation(s)
- Yimeng Sun
- Cancer Institute, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Lin Yao
- Cancer Institute, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Changfeng Man
- Cancer Institute, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhenjun Gao
- Department of Gastroenterology, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Rong He
- Cancer Institute, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yu Fan
- Cancer Institute, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|