1
|
Wang SM, Zhang MF, Pan QH, Yu TF, Lei RL, Li QJ. Causal Relationships Between Gut Microbiota, Immune Cell and Pancreatic Cancer: A Two-Step, Two-Sample Mendelian Randomization Study. World J Oncol 2024; 15:922-928. [PMID: 39697429 PMCID: PMC11650607 DOI: 10.14740/wjon1960] [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: 08/25/2024] [Accepted: 10/17/2024] [Indexed: 12/20/2024] Open
Abstract
Background Gut microbiota (GM) is associated with both the occurrence and development of pancreatic cancer (PC), and immune cells potentially play a role in this process. This study sought to evaluate the causative effect of GM on PC and to ascertain possible immune cell mediators. Methods The study primarily employed a two-step, two-sample Mendelian randomization (MR) analysis to explore the causal relationship between GM and PC within the European population, placing particular emphasis on the application of the inverse variance weighted (IVW) approach. Additionally, mediation analysis was conducted to explore the potential influence of immune cells as mediators. Results The MR analysis revealed a significant association between Geminocystis and the risk of PC. Increased abundance of Geminocystis was positively associated with the risk of PC (odds ratio (OR): 2.580, 95% confidence interval (CI): 1.050 - 6.342). The validity of the outcomes was also verified by the sensitivity analysis. The mediation MR analysis showed that the B-cell absolute count served as a partial intermediary in the causal link between Geminocystis and the risk of PC, contributing to 15.321% of the mediating impact. Conclusion This MR study demonstrated that Geminocystis has a causal relationship with PC and potentially mediates B-cell absolute count in the TBNK panel.
Collapse
Affiliation(s)
- Si Ming Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- These authors contributed equally to this article
| | - Ming Feng Zhang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- These authors contributed equally to this article
| | - Qian Hui Pan
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- These authors contributed equally to this article
| | - Ting Feng Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Rui Lin Lei
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Gynecological Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qing Jian Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
2
|
van Eijck CWF, Ju J, van 't Land FR, Verheij M, Li Y, Stubbs A, Doukas M, Lila K, Heij LR, Wiltberger G, Alonso L, Malats N, Groot Koerkamp B, Vietsch EE, van Eijck CHJ. The tumor immune microenvironment in resected treatment-naive pancreatic cancer patients with long-term survival. Pancreatology 2024; 24:1057-1065. [PMID: 39218754 DOI: 10.1016/j.pan.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/27/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers worldwide. Presently, only a fraction of patients undergo successful surgical resection, the most effective treatment. Enhancing treatment strategies necessitates a deep comprehension of the factors underlying extended survival after surgical resection in patients. METHODS This study aims to identify the important factors of PDAC patients' long-term survival with metatranscriptomics and multiplex immunofluorescence (IF) staining analyses. Specifically, differences in tumor immune microenvironment (TIME) were investigated between treatment-naïve PDAC short-term survivors (STS, overall survival <6 months) and long-term survivors (LTS, overall survival >5 years). RESULTS As a result, we detected 589 over-expressed genes, including HOXB9, CDA, and HOXB8, and 507 under-expressed genes, including AMY2B, SCARA5, and SLC2A2 in LTS. Most of the Reactome overbiological pathways enriched in our data were over-expressed in LTS, such as RHO GTPase Effectors and Cell Cycle Checkpoints. Eleven microbiomes significantly differed between LTS and STS, including Sphingopyxis and Capnocytophaga. Importantly, we demonstrate that the TIME profile with an increased abundance of memory B cells and the reduction of M0 and pro-tumoral M2 macrophages are associated with a good prognosis in PDAC. CONCLUSIONS In this study, we delved into the TIME with metatranscriptomics and IF staining analyses to understand the prerequisite of prolonged survival in PDAC patients. In LTS, several biological pathways were overexpressed, and specific microbiomes were identified. Furthermore, apparent differences in driven immune factors were found that provide valuable insights into developing new treatment strategies.
Collapse
Affiliation(s)
- Casper W F van Eijck
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
| | - Jie Ju
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Freek R van 't Land
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands
| | - Maaike Verheij
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands
| | - Yunlei Li
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Andrew Stubbs
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Michael Doukas
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Karishma Lila
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Lara R Heij
- Institute of Pathology, Medical Center University Duisburg-Essen, Essen, Germany; Department of Surgery and Transplantation, University Hospital Essen, Germany; Department of General, Gastrointestinal, Hepatobiliary and Transplant Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Georg Wiltberger
- Department of General, Gastrointestinal, Hepatobiliary and Transplant Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Lola Alonso
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Bas Groot Koerkamp
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands
| | - Eveline E Vietsch
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands
| | - Casper H J van Eijck
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| |
Collapse
|
3
|
Guo JL, Lopez DM, Mascharak S, Foster DS, Khan A, Davitt MF, Nguyen AT, Burcham AR, Chinta MS, Guardino NJ, Griffin M, Miller E, Januszyk M, Raghavan SS, Longacre TA, Delitto DJ, Norton JA, Longaker MT. Hematoxylin and Eosin Architecture Uncovers Clinically Divergent Niches in Pancreatic Cancer. Tissue Eng Part A 2024; 30:605-613. [PMID: 38874979 DOI: 10.1089/ten.tea.2024.0039] [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] [Indexed: 06/15/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents one of the only cancers with an increasing incidence rate and is often associated with intra- and peri-tumoral scarring, referred to as desmoplasia. This scarring is highly heterogeneous in extracellular matrix (ECM) architecture and plays complex roles in both tumor biology and clinical outcomes that are not yet fully understood. Using hematoxylin and eosin (H&E), a routine histological stain utilized in existing clinical workflows, we quantified ECM architecture in 85 patient samples to assess relationships between desmoplastic architecture and clinical outcomes such as survival time and disease recurrence. By utilizing unsupervised machine learning to summarize a latent space across 147 local (e.g., fiber length, solidity) and global (e.g., fiber branching, porosity) H&E-based features, we identified a continuum of histological architectures that were associated with differences in both survival and recurrence. Furthermore, we mapped H&E architectures to a CO-Detection by indEXing (CODEX) reference atlas, revealing localized cell- and protein-based niches associated with outcome-positive versus outcome-negative scarring in the tumor microenvironment. Overall, our study utilizes standard H&E staining to uncover clinically relevant associations between desmoplastic organization and PDAC outcomes, offering a translatable pipeline to support prognostic decision-making and a blueprint of spatial-biological factors for modeling by tissue engineering methods.
Collapse
Affiliation(s)
- Jason L Guo
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - David M Lopez
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Shamik Mascharak
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Deshka S Foster
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Anum Khan
- Cell Sciences Imaging Facility, Stanford University, Stanford, California, USA
| | - Michael F Davitt
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Alan T Nguyen
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Austin R Burcham
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Malini S Chinta
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Nicholas J Guardino
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Michelle Griffin
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Elisabeth Miller
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Michael Januszyk
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Shyam S Raghavan
- Department of Pathology, University of Colorado Anschutz Medical Center, Aurora, Colorado, USA
| | - Teri A Longacre
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Daniel J Delitto
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Jeffrey A Norton
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Michael T Longaker
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
4
|
Poyia F, Neophytou CM, Christodoulou MI, Papageorgis P. The Role of Tumor Microenvironment in Pancreatic Cancer Immunotherapy: Current Status and Future Perspectives. Int J Mol Sci 2024; 25:9555. [PMID: 39273502 PMCID: PMC11395109 DOI: 10.3390/ijms25179555] [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: 07/26/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Pancreatic cancer comprises different subtypes, where most cases include ductal adenocarcinoma (PDAC). It is one of the deadliest tumor types, with a poor prognosis. In the majority of patients, the disease has already spread by the time of diagnosis, making full recovery unlikely and increasing mortality risk. Despite developments in its detection and management, including chemotherapy, radiotherapy, and targeted therapies as well as advances in immunotherapy, only in about 13% of PDAC patients does the overall survival exceed 5 years. This may be attributed, at least in part, to the highly desmoplastic tumor microenvironment (TME) that acts as a barrier limiting perfusion, drug delivery, and immune cell infiltration and contributes to the establishment of immunologically 'cold' conditions. Therefore, there is an urgent need to unravel the complexity of the TME that promotes PDAC progression and decipher the mechanisms of pancreatic tumors' resistance to immunotherapy. In this review, we provide an overview of the major cellular and non-cellular components of PDAC TME, as well as their biological interplays. We also discuss the current state of PDAC therapeutic treatments and focus on ongoing and future immunotherapy efforts and multimodal treatments aiming at remodeling the TME to improve therapeutic efficacy.
Collapse
Affiliation(s)
- Fotini Poyia
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 2404, Cyprus
| | - Christiana M Neophytou
- Apoptosis and Cancer Chemoresistance Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 2404, Cyprus
| | - Maria-Ioanna Christodoulou
- Tumor Immunology and Biomarkers Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 2404, Cyprus
| | - Panagiotis Papageorgis
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 2404, Cyprus
| |
Collapse
|
5
|
Xie W, Lu J, Chen Y, Wang X, Lu H, Li Q, Jin N, He J, Ou L, Ni J, Shen Y, Shao L. TCL1A-expressing B cells are critical for tertiary lymphoid structure formation and the prognosis of oral squamous cell carcinoma. J Transl Med 2024; 22:477. [PMID: 38764038 PMCID: PMC11103841 DOI: 10.1186/s12967-024-05292-7] [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: 01/28/2024] [Accepted: 05/11/2024] [Indexed: 05/21/2024] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is a malignant tumor with a poor prognosis. Traditional treatments have limited effectiveness. Regulation of the immune response represents a promising new approach for OSCC treatment. B cells are among the most abundant immune cells in OSCC. However, the role of B cells in OSCC treatment has not been fully elucidated. METHODS Single-cell RNA sequencing analysis of 13 tissues and 8 adjacent normal tissues from OSCC patients was performed to explore differences in B-cell gene expression between OSCC tissues and normal tissues. We further investigated the relationship between differentially expressed genes and the immune response to OSCC. We utilized tissue microarray data for 146 OSCC clinical samples and RNA sequencing data of 359 OSCC samples from The Cancer Genome Atlas (TCGA) to investigate the role of T-cell leukemia 1 A (TCL1A) in OSCC prognosis. Multiplex immunohistochemistry (mIHC) was employed to investigate the spatial distribution of TCL1A in OSCC tissues. We then investigated the effect of TCL1A on B-cell proliferation and trogocytosis. Finally, lentiviral transduction was performed to induce TCL1A overexpression in B lymphoblastoid cell lines (BLCLs) to verify the function of TCL1A. RESULTS Our findings revealed that TCL1A was predominantly expressed in B cells and was associated with a better prognosis in OSCC patients. Additionally, we found that TCL1A-expressing B cells are located at the periphery of lymphatic follicles and are associated with tertiary lymphoid structures (TLS) formation in OSCC. Mechanistically, upregulation of TCL1A promoted the trogocytosis of B cells on dendritic cells by mediating the upregulation of CR2, thereby improving antigen-presenting ability. Moreover, the upregulation of TCL1A expression promoted the proliferation of B cells. CONCLUSION This study revealed the role of B-cell TCL1A expression in TLS formation and its effect on OSCC prognosis. These findings highlight TCL1A as a novel target for OSCC immunotherapy.
Collapse
Affiliation(s)
- Wenqiang Xie
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, PR China
| | - Jinjin Lu
- Department of Periodontics, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510182, Guangdong, PR China
| | - Yichen Chen
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, PR China
| | - Xi Wang
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, 510055, PR China
| | - Huanzi Lu
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, 510055, PR China
| | - Qunxing Li
- Department of Stomatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Nianqiang Jin
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, PR China
| | - Jiankang He
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, PR China
| | - Lingling Ou
- Department of Periodontics, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510182, Guangdong, PR China
| | - Jia Ni
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, PR China
| | - Yuqin Shen
- Department of Periodontics, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510182, Guangdong, PR China
| | - Longquan Shao
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, PR China.
| |
Collapse
|
6
|
van Eijck CWF, Sabroso-Lasa S, Strijk GJ, Mustafa DAM, Fellah A, Koerkamp BG, Malats N, van Eijck CHJ. A liquid biomarker signature of inflammatory proteins accurately predicts early pancreatic cancer progression during FOLFIRINOX chemotherapy. Neoplasia 2024; 49:100975. [PMID: 38335839 PMCID: PMC10873733 DOI: 10.1016/j.neo.2024.100975] [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: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is often treated with FOLFIRINOX, a chemotherapy associated with high toxicity rates and variable efficacy. Therefore, it is crucial to identify patients at risk of early progression during treatment. This study aims to explore the potential of a multi-omics biomarker for predicting early PDAC progression by employing an in-depth mathematical modeling approach. METHODS Blood samples were collected from 58 PDAC patients undergoing FOLFIRINOX before and after the first cycle. These samples underwent gene (GEP) and inflammatory protein expression profiling (IPEP). We explored the predictive potential of exclusively IPEP through Stepwise (Backward) Multivariate Logistic Regression modeling. Additionally, we integrated GEP and IPEP using Bayesian Kernel Regression modeling, aiming to enhance predictive performance. Ultimately, the FOLFIRINOX IPEP (FFX-IPEP) signature was developed. RESULTS Our findings revealed that proteins exhibited superior predictive accuracy than genes. Consequently, the FFX-IPEP signature consisted of six proteins: AMN, BANK1, IL1RL2, ITGB6, MYO9B, and PRSS8. The signature effectively identified patients transitioning from disease control to progression early during FOLFIRINOX, achieving remarkable predictive accuracy with an AUC of 0.89 in an independent test set. Importantly, the FFX-IPEP signature outperformed the conventional CA19-9 tumor marker. CONCLUSIONS Our six-protein FFX-IPEP signature holds solid potential as a liquid biomarker for the early prediction of PDAC progression during toxic FOLFIRINOX chemotherapy. Further validation in an external cohort is crucial to confirm the utility of the FFX-IPEP signature. Future studies should expand to predict progression under different chemotherapies to enhance the guidance of personalized treatment selection in PDAC.
Collapse
Affiliation(s)
- Casper W F van Eijck
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain.
| | - Sergio Sabroso-Lasa
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain; Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), Madrid, Spain
| | - Gaby J Strijk
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dana A M Mustafa
- Department of Clinical Bioinformatics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Amine Fellah
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain; Centro de Investigación Biomédica en Red-Cáncer (CIBERONC), Madrid, Spain
| | - Casper H J van Eijck
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain.
| |
Collapse
|
7
|
MINESHIGE T, TANAKA Y, WATANABE K, TAGAWA M, TOMIHARI M, KOBAYASHI Y. Histologic, immunohistochemical, and in situ hybridization study of myxoid stroma in feline oral squamous cell carcinoma. J Vet Med Sci 2024; 86:258-265. [PMID: 38233195 PMCID: PMC10963083 DOI: 10.1292/jvms.23-0356] [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: 08/23/2023] [Accepted: 12/26/2023] [Indexed: 01/19/2024] Open
Abstract
Oral squamous cell carcinoma (oSCC) is a highly invasive malignant neoplasm in cats. Recently, tumor stroma, known as tumor microenvironments, have been considered to play an essential role in tumor progression. However, their role in feline squamous cell carcinoma (SCC) remains unclear. This study aimed to reveal the cancer microenvironment of feline oSCC and evaluate the pathological mechanisms of progression. We used 19 samples from 17 cats with oSCC, which were examined using light microscopy, immunohistochemistry, and in situ hybridization (RNAscope®). Feline oSCCs had two types of stroma, namely fibrotic and myxoid stromal reaction patterns, which were easily distinguished using hematoxylin-eosin staining. The myxoid stroma was rich in hyaluronic acid, which seems to be produced by neoplastic cells. Furthermore, the presence of myxoid stroma was correlated with histological parameters, including the appearance of cancer-associated fibroblasts and tumor budding. Periostin protein expression was also frequently observed in the stroma of feline oSCC and was significantly more common in the myxoid stromal reaction pattern group than in the fibrotic group. Positive signals for periostin mRNA were detected in stromal cancer-associated fibroblasts. This study indicates that the interaction between neoplastic cells and stromal reaction pattern components, such as hyaluronic acid and periostin, may be involved in tumor malignancy. Therefore, we propose that focus be placed not only on the tumor tissue but also on the characterization of the stroma for analyzing feline oSCC.
Collapse
Affiliation(s)
- Takayuki MINESHIGE
- Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
- Present address: School of Veterinary Medicine, Azabu University, Kanagawa, Japan
| | - Yusuke TANAKA
- Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
| | - Kenichi WATANABE
- Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
| | - Michihito TAGAWA
- Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
- Veterinary Medical Center, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
- Department of Veterinary Associated Science, Okayama University of Science, Ehime, Japan
| | - Mizuki TOMIHARI
- Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
- Veterinary Medical Center, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
- Department of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Yoshiyasu KOBAYASHI
- Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
| |
Collapse
|
8
|
Maimela PWM, Smith M, Nel AJM, Bernam SDP, Jonas EG, Blackburn JM. Humoral immunoprofiling identifies novel biomarkers and an immune suppressive autoantibody phenotype at the site of disease in pancreatic ductal adenocarcinoma. Front Oncol 2024; 14:1330419. [PMID: 38450186 PMCID: PMC10917065 DOI: 10.3389/fonc.2024.1330419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/22/2024] [Indexed: 03/08/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a heterogeneous cancer, with minimal response to therapeutic intervention and with 85% of cases diagnosed at an advanced stage due to lack of early symptoms, highlighting the importance of understanding PDAC immunology in greater detail. Here, we applied an immunoproteomic approach to investigate autoantibody responses against cancer-testis and tumor-associated antigens in PDAC using a high-throughput multiplexed protein microarray platform, comparing humoral immune responses in serum and at the site of disease in order to shed new light on immune responses in the tumor microenvironment. We simultaneously quantified serum or tissue IgG and IgA antibody isotypes and subclasses in a cohort of PDAC, disease control and healthy patients, observing inter alia that subclass utilization in tumor tissue samples was predominantly immune suppressive IgG4 and inflammatory IgA2, contrasting with predominant IgG3 and IgA1 subclass utilization in matched sera and implying local autoantibody production at the site of disease in an immune-tolerant environment. By comparison, serum autoantibody subclass profiling for the disease controls identified IgG4, IgG1, and IgA1 as the abundant subclasses. Combinatorial analysis of serum autoantibody responses identified panels of candidate biomarkers. The top IgG panel included ACVR2B, GAGE1, LEMD1, MAGEB1 and PAGE1 (sensitivity, specificity and AUC values of 0.933, 0.767 and 0.906). Conversely, the top IgA panel included AURKA, GAGE1, MAGEA10, PLEKHA5 and XAGE3aV1 (sensitivity, specificity, and AUC values of 1.000, 0.800, and 0.954). Assessment of antigen-specific serum autoantibody glycoforms revealed abundant sialylation on IgA in PDAC, consistent with an immune suppressive IgA response to disease.
Collapse
Affiliation(s)
- Pamela Winnie M. Maimela
- Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Muneerah Smith
- Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Andrew J. M. Nel
- Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | | | - Eduard G. Jonas
- Department of Surgery, Gastroenterology Unit, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Jonathan M. Blackburn
- Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Sengenics Corporation, Kuala Lumpur, Malaysia
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
9
|
Sebastian A, Martin KA, Peran I, Hum NR, Leon NF, Amiri B, Wilson SP, Coleman MA, Wheeler EK, Byers SW, Loots GG. Loss of Cadherin-11 in pancreatic ductal adenocarcinoma alters tumor-immune microenvironment. Front Oncol 2023; 13:1286861. [PMID: 37954069 PMCID: PMC10639148 DOI: 10.3389/fonc.2023.1286861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/12/2023] [Indexed: 11/14/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the top five deadliest forms of cancer with very few treatment options. The 5-year survival rate for PDAC is 10% following diagnosis. Cadherin 11 (Cdh11), a cell-to-cell adhesion molecule, has been suggested to promote tumor growth and immunosuppression in PDAC, and Cdh11 inhibition significantly extended survival in mice with PDAC. However, the mechanisms by which Cdh11 deficiency influences PDAC progression and anti-tumor immune responses have yet to be fully elucidated. To investigate Cdh11-deficiency induced changes in PDAC tumor microenvironment (TME), we crossed p48-Cre; LSL-KrasG12D/+; LSL-Trp53R172H/+ (KPC) mice with Cdh11+/- mice and performed single-cell RNA sequencing (scRNA-seq) of the non-immune (CD45-) and immune (CD45+) compartment of KPC tumor-bearing Cdh11 proficient (KPC-Cdh11+/+) and Cdh11 deficient (KPC-Cdh11+/-) mice. Our analysis showed that Cdh11 is expressed primarily in cancer-associated fibroblasts (CAFs) and at low levels in epithelial cells undergoing epithelial-to-mesenchymal transition (EMT). Cdh11 deficiency altered the molecular profile of CAFs, leading to a decrease in the expression of myofibroblast markers such as Acta2 and Tagln and cytokines such as Il6, Il33 and Midkine (Mdk). We also observed a significant decrease in the presence of monocytes/macrophages and neutrophils in KPC-Cdh11+/- tumors while the proportion of T cells was increased. Additionally, myeloid lineage cells from Cdh11-deficient tumors had reduced expression of immunosuppressive cytokines that have previously been shown to play a role in immune suppression. In summary, our data suggests that Cdh11 deficiency significantly alters the fibroblast and immune microenvironments and contributes to the reduction of immunosuppressive cytokines, leading to an increase in anti-tumor immunity and enhanced survival.
Collapse
Affiliation(s)
- Aimy Sebastian
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Kelly A. Martin
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Ivana Peran
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University Medical Center, Washington, DC, United States
| | - Nicholas R. Hum
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Nicole F. Leon
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Beheshta Amiri
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Stephen P. Wilson
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Matthew A. Coleman
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Elizabeth K. Wheeler
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Stephen W. Byers
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University Medical Center, Washington, DC, United States
| | - Gabriela G. Loots
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
- University of California Davis Health, Department of Orthopaedic Surgery, Sacramento, CA, United States
| |
Collapse
|
10
|
Li O, Li L, Sheng Y, Ke K, Wu J, Mou Y, Liu M, Jin W. Biological characteristics of pancreatic ductal adenocarcinoma: Initiation to malignancy, intracellular to extracellular. Cancer Lett 2023; 574:216391. [PMID: 37714257 DOI: 10.1016/j.canlet.2023.216391] [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: 05/28/2023] [Revised: 09/04/2023] [Accepted: 09/10/2023] [Indexed: 09/17/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly life-threatening tumour with a low early-detection rate, rapid progression and a tendency to develop resistance to chemotherapy. Therefore, understanding the regulatory mechanisms underlying the initiation, development and metastasis of pancreatic cancer is necessary for enhancing therapeutic effectiveness. In this review, we summarised single-gene mutations (including KRAS, CDKN2A, TP53, SMAD4 and some other less prevalent mutations), epigenetic changes (including DNA methylation, histone modifications and RNA interference) and large chromosome alterations (such as copy number variations, chromosome rearrangements and chromothripsis) associated with PDAC. In addition, we discussed variations in signalling pathways that act as intermediate oncogenic factors in PDAC, including PI3K/AKT, MAPK/ERK, Hippo and TGF-β signalling pathways. The focus of this review was to investigate alterations in the microenvironment of PDAC, particularly the role of immunosuppressive cells, cancer-associated fibroblasts, lymphocytes, other para-cancerous cells and tumour extracellular matrix in tumour progression. Peripheral axons innervating the pancreas have been reported to play a crucial role in the development of cancer. In addition, tumour cells can influence the behaviour of neighbouring non-tumour cells by secreting certain factors, both locally and at a distance. In this review, we elucidated the alterations in intracellular molecules and the extracellular environment that occur during the progression of PDAC. Altogether, this review may enhance the understanding of the biological characteristics of PDAC and guide the development of more precise treatment strategies.
Collapse
Affiliation(s)
- Ou Li
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Li Li
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yunru Sheng
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Kun Ke
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jianzhang Wu
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yiping Mou
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Mingyang Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center, China; National Clinical Research Center for Cancer, China; Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Weiwei Jin
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
11
|
Guo J, Wang S, Gao Q. An integrated overview of the immunosuppression features in the tumor microenvironment of pancreatic cancer. Front Immunol 2023; 14:1258538. [PMID: 37771596 PMCID: PMC10523014 DOI: 10.3389/fimmu.2023.1258538] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 08/29/2023] [Indexed: 09/30/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies. It is characterized by a complex and immunosuppressive tumor microenvironment (TME), which is primarily composed of tumor cells, stromal cells, immune cells, and acellular components. The cross-interactions and -regulations among various cell types in the TME have been recognized to profoundly shape the immunosuppression features that meaningfully affect PDAC biology and treatment outcomes. In this review, we first summarize five cellular composition modules by integrating the cellular (sub)types, phenotypes, and functions in PDAC TME. Then we discuss an integrated overview of the cross-module regulations as a determinant of the immunosuppressive TME in PDAC. We also briefly highlight TME-targeted strategies that potentially improve PDAC therapy.
Collapse
Affiliation(s)
- Jinglong Guo
- Department of Cardiovascular Disease, the First Hospital of Jilin University, Changchun, China
| | - Siyue Wang
- Baylor College of Medicine, One Baylor Plaza, Houston, TX, United States
| | - Qi Gao
- Department of Cardiovascular Disease, the First Hospital of Jilin University, Changchun, China
| |
Collapse
|
12
|
Nie Y, Xu L, Bai Z, Liu Y, Wang S, Zeng Q, Gao X, Xia X, Chang D. Prognostic utility of TME-associated genes in pancreatic cancer. Front Genet 2023; 14:1218774. [PMID: 37727377 PMCID: PMC10505756 DOI: 10.3389/fgene.2023.1218774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/15/2023] [Indexed: 09/21/2023] Open
Abstract
Background: Pancreatic cancer (PC) is a deadly disease. The tumor microenvironment (TME) participates in PC oncogenesis. This study focuses on the assessment of the prognostic and treatment utility of TME-associated genes in PC. Methods: After obtaining the differentially expressed TME-related genes, univariate and multivariate Cox analyses and least absolute shrinkage and selection operator (LASSO) were performed to identify genes related to prognosis, and a risk model was established to evaluate risk scores, based on The Cancer Genome Atlas (TCGA) data set, and it was validated by external data sets from the Gene Expression Omnibus (GEO) and Clinical Proteomic Tumor Analysis Consortium (CPTAC). Multiomics analyses were adopted to explore the potential mechanisms, discover novel treatment targets, and assess the sensitivities of immunotherapy and chemotherapy. Results: Five TME-associated genes, namely, FERMT1, CARD9, IL20RB, MET, and MMP3, were identified and a risk score formula constructed. Next, their mRNA expressions were verified in cancer and normal pancreatic cells. Multiple algorithms confirmed that the risk model displayed a reliable ability of prognosis prediction and was an independent prognostic factor, indicating that high-risk patients had poor outcomes. Immunocyte infiltration, gene set enrichment analysis (GSEA), and single-cell analysis all showed a strong relationship between immune mechanism and low-risk samples. The risk score could predict the sensitivity of immunotherapy and some chemotherapy regimens, which included oxaliplatin and irinotecan. Various latent treatment targets (LAG3, TIGIT, and ARID1A) were addressed by mutation landscape based on the risk model. Conclusion: The risk model based on TME-related genes can reflect the prognosis of PC patients and functions as a novel set of biomarkers for PC therapy.
Collapse
Affiliation(s)
- Yuanhua Nie
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Longwen Xu
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Zilong Bai
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yaoyao Liu
- Geneplus-Beijing, Co., Ltd., Beijing, China
| | - Shilong Wang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Qingnuo Zeng
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xuan Gao
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- GenePlus- Shenzhen Clinical Laboratory, Shenzhen, China
| | | | - Dongmin Chang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
13
|
Zou X, Guan C, Gao J, Shi W, Cui Y, Zhong X. Tertiary lymphoid structures in pancreatic cancer: a new target for immunotherapy. Front Immunol 2023; 14:1222719. [PMID: 37529035 PMCID: PMC10388371 DOI: 10.3389/fimmu.2023.1222719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 06/26/2023] [Indexed: 08/03/2023] Open
Abstract
Pancreatic cancer (PC) is extremely malignant and shows limited response to available immunotherapies due to the hypoxic and immunosuppressive nature of its tumor microenvironment (TME). The aggregation of immune cells (B cells, T cells, dendritic cells, etc.), which is induced in various chronic inflammatory settings such as infection, inflammation, and tumors, is known as the tertiary lymphoid structure (TLS). Several studies have shown that TLSs can be found in both intra- and peritumor tissues of PC. The role of TLSs in peritumor tissues in tumors remains unclear, though intratumoral TLSs are known to play an active role in a variety of tumors, including PC. The formation of intratumoral TLSs in PC is associated with a good prognosis. In addition, TLSs can be used as an indicator to assess the effectiveness of treatment. Targeted induction of TLS formation may become a new avenue of immunotherapy for PC. This review summarizes the formation, characteristics, relevant clinical outcomes, and clinical applications of TLSs in the pancreatic TME. We aim to provide new ideas for future immunotherapy of PC.
Collapse
Affiliation(s)
- Xinlei Zou
- Department of Hepatopancreatobiary Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Canghai Guan
- Department of Hepatopancreatobiary Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jianjun Gao
- Department of Hepatopancreatobiary Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wujiang Shi
- Department of Hepatopancreatobiary Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yunfu Cui
- Department of Hepatopancreatobiary Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangyu Zhong
- Department of Hepatopancreatobiary Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
14
|
Khabipov A, Trung DN, van der Linde J, Miebach L, Lenz M, Erne F, von Bernstorff W, Schulze T, Kersting S, Bekeschus S, Partecke LI. CCR4 Blockade Diminishes Intratumoral Macrophage Recruitment and Augments Survival of Syngeneic Pancreatic Cancer-Bearing Mice. Biomedicines 2023; 11:1517. [PMID: 37371612 DOI: 10.3390/biomedicines11061517] [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: 04/18/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 06/29/2023] Open
Abstract
Pancreatic cancer is known for its tumor microenvironment (TME), which is rich in stromal and immune cells supporting cancer growth and therapy resistance. In particular, tumor-associated macrophages (TAMs) are known for their angiogenesis- and metastasis-promoting properties, which lead to the failure of conventional therapies for pancreatic cancer. Hence, treatment options targeting TAMs are needed. The C-C chemokine receptor type 4 (CCR4) is critical for immune cell recruitment into the TME, and in this paper we explore the effects of its genetic or immunotherapeutic blockade in pancreatic-cancer-bearing mice. Murine PDA6606 pancreatic cancer cells and murine peritoneal macrophages were used for in vitro migration assays. In vivo, a syngeneic, orthotropic pancreatic cancer model was established. Tumor growth and survival were monitored under prophylactic and therapeutic application of a CCR4 antagonist (AF-399/420/18025) in wildtype (CCR4wt) and CCR4-knockout (CCR4-/-) mice. Immune infiltration was monitored in tumor tissue sections and via flow cytometry of lysed tumors. PDA6606 cells induced less migration in CCR4-/- than in CCR4wt macrophages in vitro. Pancreatic TAM infiltration was higher, and survival was reduced in CCR4wt mice compared to CCR4-/- mice. Antagonizing CCR4 in wildtype mice revealed similar results as in CCR4-/- mice without antagonization. Prophylactic CCR4 antagonist application in wildtype mice was more efficient than therapeutic antagonization. CCR4 seems to be critically involved in TAM generation and tumor progression in pancreatic cancer. CCR4 blockade may help prolong the relapse-free period after curative surgery in pancreatic cancer and improve prognosis.
Collapse
Affiliation(s)
- Aydar Khabipov
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Dung Nguyen Trung
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Julia van der Linde
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Lea Miebach
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Maik Lenz
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Felix Erne
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Wolfram von Bernstorff
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Tobias Schulze
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Stephan Kersting
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Sander Bekeschus
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
- Clinic and Policlinic for Dermatology and Venerology, Rostock University Medical Center, Strempelstr. 13, 18057 Rostock, Germany
| | - Lars Ivo Partecke
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
- Department of General, Visceral, and Thoracic Surgery, Helios Clinic Schleswig, St. Jurgener Str. 1-3, 24837 Schleswig, Germany
| |
Collapse
|
15
|
Montagne JM, Jaffee EM, Fertig EJ. Multiomics Empowers Predictive Pancreatic Cancer Immunotherapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:859-868. [PMID: 36947820 PMCID: PMC10236355 DOI: 10.4049/jimmunol.2200660] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/23/2022] [Indexed: 03/24/2023]
Abstract
Advances in cancer immunotherapy, particularly immune checkpoint inhibitors, have dramatically improved the prognosis for patients with metastatic melanoma and other previously incurable cancers. However, patients with pancreatic ductal adenocarcinoma (PDAC) generally do not respond to these therapies. PDAC is exceptionally difficult to treat because of its often late stage at diagnosis, modest mutation burden, and notoriously complex and immunosuppressive tumor microenvironment. Simultaneously interrogating features of cancer, immune, and other cellular components of the PDAC tumor microenvironment is therefore crucial for identifying biomarkers of immunotherapeutic resistance and response. Notably, single-cell and multiomics technologies, along with the analytical tools for interpreting corresponding data, are facilitating discoveries of the systems-level cellular and molecular interactions contributing to the overall resistance of PDAC to immunotherapy. Thus, in this review, we will explore how multiomics and single-cell analyses provide the unprecedented opportunity to identify biomarkers of resistance and response to successfully sensitize PDAC to immunotherapy.
Collapse
Affiliation(s)
- Janelle M Montagne
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Elizabeth M Jaffee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg Kimmel Immunology Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Elana J Fertig
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
16
|
Kou YQ, Yang YP, Pan ZJ, Du SS, Yuan WN, He K, Nie B. Prognostic-Related Biomarkers in Pancreatic Ductal Adenocarcinoma Correlating with Immune Infiltrates Based on Proteomics. Med Sci Monit 2023; 29:e938785. [PMID: 36905103 PMCID: PMC10015732 DOI: 10.12659/msm.938785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) accounts for 85% of pancreatic carcinoma cases. Patients with PDAC have a poor prognosis. The lack of reliable prognostic biomarkers makes treatment challenging for patients with PDAC. Using a bioinformatics database, we sought to identify prognostic biomarkers for PDAC. MATERIAL AND METHODS Using proteomic analysis of the Clinical Proteomics Tumor Analysis Consortium (CPTAC) database, we were able to identify core differential proteins between early and advanced pancreatic ductal adenocarcinoma tissue, and then we used survival analysis, Cox regression analysis, and area under the ROC curves to screen for more significant differential proteins. Additionally, the Kaplan-Meier plotter database was utilized to determine the relationship between prognosis and immune infiltration in PDAC. RESULTS We identified 378 differential proteins in early (n=78) and advanced stages (n=47) of PDAC (P<0.05). PLG, COPS5, FYN, ITGB3, IRF3, and SPTA1 served as independent prognostic factors of patients with PDAC. Patients with higher COPS5 expression had shorter overall survival (OS) and recurrence-free survival, and those with higher PLG, ITGB3, and SPTA1, and lower FYN and IRF3 expression had shorter OS. More importantly, COPS5, IRF3 were negatively associated with macrophages and NK cells, but PLG, FYN, ITGB3, and SPTA1 were positively related to the expression of CD8+ T cells and B cells. COPS5 affected the prognosis of PDAC patients by acting on B cells, CD8+ T cells, macrophages, and NK cells immune infiltration, while PLG, FYN, ITGB3, IRF3, and SPTA1 affected PDAC patient prognosis through some immune cells. CONCLUSIONS PLG, COPS5, FYN, IRF3, ITGB3 and SPTA1 could be potential immunotherapeutic targets and valuable prognostic biomarkers of PDAC.
Collapse
Affiliation(s)
- Yan-Qi Kou
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China (mainland)
| | - Yu-Ping Yang
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China (mainland)
| | - Zhao-Jie Pan
- Department of Gastrointestinal Endoscopy, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China (mainland)
| | - Shen-Shen Du
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China (mainland)
| | - Wei-Nan Yuan
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China (mainland)
| | - Kun He
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China (mainland)
| | - Biao Nie
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China (mainland)
| |
Collapse
|
17
|
Messex JK, Liou GY. Impact of Immune Cells in the Tumor Microenvironment of Prostate Cancer Metastasis. Life (Basel) 2023; 13:333. [PMID: 36836690 PMCID: PMC9967893 DOI: 10.3390/life13020333] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/17/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
Prostate cancer is the most prevalent type of cancer in senior American men. Currently, the five-year survival rate after the initial diagnosis of prostate cancer is close to 100%. However, it is also the second leading cause of cancer death in senior men due to the dissemination of prostate cancer cells outside of the prostate causing growth in other organs, known as metastatic prostate cancer. The tumor microenvironment (TME) plays a critical role in the development, progression and metastasis of prostate cancer. One of the major components of the TME contains various types of immune cells, often recruited by cancer cells to the cancer formation areas. The interactions among prostate cancer cells and the infiltrating immune cells affect the outcome of prostate cancer. Here, we summarize the mechanisms various infiltrating immune cells use to regulate prostate cancer metastasis and possibly lead to the development of treatment strategies. Furthermore, the information here may also give rise to preventative strategies that focus on targeting the TME of prostate cancer patients.
Collapse
Affiliation(s)
- Justin K. Messex
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| | - Geou-Yarh Liou
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
- Department of Biological Sciences, Clark Atlanta University, Atlanta, GA 30314, USA
| |
Collapse
|
18
|
Senturk ZN, Akdag I, Deniz B, Sayi-Yazgan A. Pancreatic cancer: Emerging field of regulatory B-cell-targeted immunotherapies. Front Immunol 2023; 14:1152551. [PMID: 37033931 PMCID: PMC10076755 DOI: 10.3389/fimmu.2023.1152551] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/10/2023] [Indexed: 04/11/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common type of pancreatic cancer, is characterized by a high mortality rate and poor prognosis. Current treatments for PDAC, are ineffective due to a prominent immunosuppressive PDAC tumor microenvironment (TME). Although B lymphocytes are highly infiltrated into PDAC, the importance of B lymphocytes in tumorigenesis is largely neglected. B cells play a dual role in the PDAC tumor microenvironment, acting as either anti-tumorigenic or pro-tumorigenic depending on where they are localized. Tumor-infiltrating B cells, which reside in ectopic lymph nodes, namely tertiary lymphoid structures (TLS), produce anti-tumor antibodies and present tumor antigens to T cells to contribute to cancer immunosurveillance. Alternatively, regulatory B cells (Bregs), dispersed inside the TME, contribute to the dampening of anti-tumor immune responses by secreting anti-inflammatory cytokines (IL-10 and IL-35), which promote tumor growth and metastasis. Determining the role of Bregs in the PDAC microenvironment is thus becoming increasingly attractive for developing novel immunotherapeutic approaches. In this minireview, we shed light on the emerging role of B cells in PDAC development and progression, with an emphasis on regulatory B cells (Bregs). Furthermore, we discussed the potential link of Bregs to immunotherapies in PDAC. These current findings will help us in understanding the full potential of B cells in immunotherapy.
Collapse
|
19
|
Luong T, Golivi Y, Nagaraju GP, El-Rayes BF. Fibroblast heterogeneity in pancreatic ductal adenocarcinoma: Perspectives in immunotherapy. Cytokine Growth Factor Rev 2022; 68:107-115. [PMID: 36096869 DOI: 10.1016/j.cytogfr.2022.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 01/30/2023]
Abstract
Cancer-associated fibroblasts (CAFs), the key component in pancreatic tumor microenvironment (TME), originate from many sources and are naturally heterogeneous in phenotype and function. Numerous studies have identified their crucial role in promoting tumorigenesis through many routes including fostering cancer proliferation, angiogenesis, invasion, and metastasis. Conversely, research also indicates that subsets of CAFs express anti-tumor activity. These dual effects reflect the complexity of CAF heterogeneity and their interactions with other cells and factors in pancreatic TME. A critical component in this environment is infiltrated immune cells and immune mediators, which can communicate with CAFs. The crosstalk occurs via the production of various cytokines, chemokines, and other mediators and shapes the immunological state in TME. Comprehensive studies of the crosstalk between CAFs and tumor immune environment, particularly internal mechanisms interlinking CAFs and immune effectors, may provide new approaches for pancreatic ductal adenocarcinoma (PDAC) treatments. In this review, we explore the characteristics of CAFs, describe the interplay among CAFs, infiltrated immune cells, other mediators, and provide an overview of recent CAF-target therapies, their limitations, and potential research directions in CAF in the context of PDAC.
Collapse
Affiliation(s)
- Tha Luong
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35201, USA
| | - Yuvasri Golivi
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35201, USA
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35201, USA.
| | - Bassel F El-Rayes
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35201, USA.
| |
Collapse
|
20
|
Acid-sensing ion channel 1: potential therapeutic target for tumor. Biomed Pharmacother 2022; 155:113835. [DOI: 10.1016/j.biopha.2022.113835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/25/2022] [Accepted: 10/06/2022] [Indexed: 11/20/2022] Open
|
21
|
Skorupan N, Palestino Dominguez M, Ricci SL, Alewine C. Clinical Strategies Targeting the Tumor Microenvironment of Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:4209. [PMID: 36077755 PMCID: PMC9454553 DOI: 10.3390/cancers14174209] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 12/04/2022] Open
Abstract
Pancreatic cancer has a complex tumor microenvironment which engages in extensive crosstalk between cancer cells, cancer-associated fibroblasts, and immune cells. Many of these interactions contribute to tumor resistance to anti-cancer therapies. Here, new therapeutic strategies designed to modulate the cancer-associated fibroblast and immune compartments of pancreatic ductal adenocarcinomas are described and clinical trials of novel therapeutics are discussed. Continued advances in our understanding of the pancreatic cancer tumor microenvironment are generating stromal and immune-modulating therapeutics that may improve patient responses to anti-tumor treatment.
Collapse
Affiliation(s)
- Nebojsa Skorupan
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Medical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mayrel Palestino Dominguez
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Samuel L. Ricci
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christine Alewine
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
22
|
Jia W, Zhang T, Yao Q, Li J, Nie Y, Lei X, Mao Z, Wang Y, Shi W, Song W. Tertiary Lymphatic Structures in Primary Hepatic Carcinoma: Controversy Cannot Overshadow Hope. Front Immunol 2022; 13:870458. [PMID: 35844587 PMCID: PMC9278517 DOI: 10.3389/fimmu.2022.870458] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Tertiary lymphoid structures (TLSs) are organized aggregates of immune cells found in the tumor microenvironment. TLS can influence primary hepatic carcinoma (PHC) occurrence and have an active role in cancer. TLS can promote or inhibit the growth of PHC depending on their location, and although available findings are controversial, they suggest that TLS have a protective role in PHC tissues and a non-protective role in paracancerous tissues. In addition, the cellular composition of TLS can also influence the outcome of PHC. As an immunity marker, TLS can act as a marker of immunotherapy to predict its effect and help to identify patients who will respond well to immunotherapy. Modulation of TLS formation through the use of chemokines/cytokines, immunotherapy, or induction of high endothelial vein to interfere with tumor growth has been studied extensively in PHC and other cancers. In addition, new tools such as genetic interventions, cellular crosstalk, preoperative radiotherapy, and advances in materials science have been shown to influence the prognosis of malignant tumors by modulating TLS production. These can also be used to develop PHC treatment.
Collapse
Affiliation(s)
- Weili Jia
- Xi’an Medical University, Xi’an, China
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Tianchen Zhang
- Xi’an Medical University, Xi’an, China
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Qianyun Yao
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jianhui Li
- Xi’an Medical University, Xi’an, China
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Ye Nie
- Xi’an Medical University, Xi’an, China
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xinjun Lei
- Xi’an Medical University, Xi’an, China
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhenzhen Mao
- Xi’an Medical University, Xi’an, China
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yanfang Wang
- Xi’an Medical University, Xi’an, China
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Wen Shi
- Xi’an Medical University, Xi’an, China
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Wenjie Song
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Wenjie Song,
| |
Collapse
|