1
|
Ma C, Zhu W, Hu X, Wu D, Zhao X, Du Y, Kong X. Acinar cells modulate the tumor microenvironment through the promotion of M1 macrophage polarization via macrophage endocytosis in pancreatic cancer. Discov Oncol 2025; 16:489. [PMID: 40198509 PMCID: PMC11979042 DOI: 10.1007/s12672-025-02244-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/26/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDA) is a highly aggressive and fatal cancer. M1 macrophages are generally considered to have anti-tumor properties, capable of suppressing tumor growth and metastasis by secreting pro-inflammatory cytokines and enhancing the immune response. AIMS The objective of this research was to pinpoint crucial genes associated with M1 macrophages and search for a new way to activate the M1 phenotype of macrophages in PDA. METHODS The level of immune cell infiltration was assessed using CIBERSORT in TCGA-PAAD cohort and ICGC-PACA cohort. We performed weighted gene co-expression network analysis (WGCNA) to identify the module most correlated with M1 macrophages and we identified hub genes through protein-protein interaction (PPI) analyse. Through survival analysis, correlation analysis and single cell analysis, we obtained the relationship between hub genes and prognosis, and the relationship between key genes and immune cells, as well as its expression in various cells. RESULTS PRSS1 (Cationic trypsinogen) and CTRB1 (Chymosinogen B) were hub genes of the M1 macrophage-associated WGCNA module (211genes) and are closely related to the extension of survival time, which are also verified as cell growth-related genes by DepMap database. Through single-cell sequencing analysis, we determined that the expression levels of PRSS1 and CTRB1 in the acinar cells of tumor tissues were diminished. PRSS1 and CTRB1 are considered to be the signature genes of acinar cells. The proportion of acinar cells was also correlated with the infiltration of CD8T cells and M1 cells. Immunostaining revealed elevated expression levels of PRSS1 and CTRB1 in adjacent normal tissues. Cell line experiments confirmed that macrophages polarize towards M1 by engulfing pancreatic enzyme granules, thereby inhibiting the malignant phenotype of tumor cells. CONCLUSION Our findings highlight the critical role of acinar cells in modulating the immune microenvironment of pancreatic tumors by influencing macrophage polarization. This insight may provide novel opportunities for therapeutic interventions in cancer treatment.
Collapse
Affiliation(s)
- Congjia Ma
- Department of Gastroenterology, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China
| | - Wenbo Zhu
- Department of Gastroenterology, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China
| | - Xiulin Hu
- Department of Gastroenterology, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China
| | - Deli Wu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Soochow University, Soochow, China
| | - Xintong Zhao
- Department of Gastroenterology, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China
| | - Yiqi Du
- Department of Gastroenterology, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China.
| | - Xiangyu Kong
- Department of Gastroenterology, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China.
| |
Collapse
|
2
|
Kang N, Duan Q, Min X, Li T, Li Y, Gao J, Liu W. Multifaceted function of B cells in tumorigenesis. Front Med 2025; 19:297-317. [PMID: 40119025 DOI: 10.1007/s11684-025-1127-5] [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: 07/31/2024] [Accepted: 12/16/2024] [Indexed: 03/24/2025]
Abstract
B lymphocytes (B cells) play a complex and paradoxical role in tumorigenesis. They can recognize tumor-associated antigens, present these antigens to T cells, and produce antibodies that directly target and eliminate tumor cells. This makes B cells a potentially powerful ally in combating cancer. However, B cells also exhibit immunosuppressive functions, secreting cytokines like IL-10 or generating tumor-promoting antibodies that dampen the anti-tumor immune response, and some tumor cells have even been shown to exploit B cells to promote their growth and metastasis. This dual nature of B cells presents both opportunities and challenges for tumor immunotherapy. In this review, we summarize the mechanisms underlying the multifaceted functions of B cells and their current applications in cancer immunotherapy. Furthermore, we also explore the key issues and future directions in this field, emphasizing the need for further research to fully harness the anti-tumor potential of B cells in the fight against cancer.
Collapse
Affiliation(s)
- Na Kang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Qinghui Duan
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Xin Min
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Tong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Yuxin Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 100084, China
- School of Management and Engineering, Nanjing University, Nanjing, 210008, China
- Postdoctoral Workstation, Govtor Capital Co., Ltd., Nanjing, 210013, China
| | - Ji Gao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Wanli Liu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 100084, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China.
| |
Collapse
|
3
|
Breaza GM, Closca RM, Cindrea AC, Hut FE, Cretu O, Sima LV, Rakitovan M, Zara F. Immunohistochemical Evaluation of the Tumor Immune Microenvironment in Pancreatic Ductal Adenocarcinoma. Diagnostics (Basel) 2025; 15:646. [PMID: 40075893 PMCID: PMC11899021 DOI: 10.3390/diagnostics15050646] [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: 02/07/2025] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Background: Pancreatic ductal adenocarcinoma is an aggressive neoplasm with a complex carcinogenesis process that must be understood through the interactions between tumor cells and tumor microenvironment cells. Methods: This study was retrospective with a chronological extension period of 16 years and included 56 cases of pancreatic ductal adenocarcinoma. This study identified, quantified, and correlated the cells of the tumor immune microenvironment in pancreatic ductal adenocarcinoma with major prognostic factors as well as overall survival, using an extensive panel of immunohistochemical markers. Results: Three tumor immunotypes were identified: subtype A (hot immunotype), subtype B (intermediate immunotype), and subtype C (cold immunotype). Patients with immunotype C exhibit considerably higher rates of both pancreatic fistulas and acute pancreatitis. Immunotypes B and C significantly increased the risk of this complication by factors of 3.68 (p = 0.002) and 3.94 (p = 0.001), respectively. The estimated probabilities of fistula formation for each immunotype are as follows: 2.5% for immunotype A, 25% for immunotype B, and 28% for immunotype C. There was a statistically significant difference in median survival times according to tumor immunotype (p < 0.001). Specifically, patients with immunotype C tumors had a median survival time of only 120.5 days, compared to 553.5 days for those with immunotype A and 331.5 for immunotype B tumors. Conclusions: The identification of the immunotype of pancreatic ductal adenocarcinoma can be a predictive factor for the occurrence of complications such as pancreatic fistula as well as for overall survival.
Collapse
Affiliation(s)
- Gelu Mihai Breaza
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
| | - Raluca Maria Closca
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- Department of Pathology, Emergency City Hospital, 300254 Timisoara, Romania
| | - Alexandru Cristian Cindrea
- Department of Surgery, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania;
- Emergency Department, Emergency Clinical Municipal Hospital, 300079 Timisoara, Romania
| | - Florin Emil Hut
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
- Center for Hepato-Bilio-Pancreatic Surgery, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania
| | - Octavian Cretu
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
| | - Laurentiu Vasile Sima
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
| | - Marina Rakitovan
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- Oro-Maxillo-Facial Surgery Clinic, Emergency City Hospital, 300062 Timisoara, Romania
| | - Flavia Zara
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- Department of Pathology, Emergency City Hospital, 300254 Timisoara, Romania
| |
Collapse
|
4
|
Sherpally D, Manne A. Advancing Immunotherapy in Pancreatic Cancer: A Brief Review of Emerging Adoptive Cell Therapies. Cancers (Basel) 2025; 17:589. [PMID: 40002184 PMCID: PMC11853216 DOI: 10.3390/cancers17040589] [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: 12/24/2024] [Revised: 01/28/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Pancreatic cancer has the lowest 5-year survival rate (13%) among major cancers and is the third leading cause of cancer-related deaths in the United States. The high lethality of this cancer is attributed to its insidious onset, late-stage diagnosis, rapid progression, and limited treatment options. Addressing these challenges requires a deeper understanding of the complex tumor microenvironment to identify novel therapeutic targets. Newer approaches like adoptive cell therapy have shown remarkable success in treating hematological malignancies, but their application in solid tumors, particularly pancreatic cancer, is still in the early stages of development. ACT broadly involves isolating immune cells (T lymphocytes, Natural Killer cells, and macrophages) from the patient, followed by genetic engineering to enhance and mount a specific anti-tumor response. Various ACT modalities are under investigation for pancreatic cancer, including chimeric antigen receptor T cells (CAR-T), chimeric antigen receptor NK cells (CAR-NK), tumor-infiltrating lymphocytes (TIL), T-cell receptor (TCR)-engineered T cells, and cytokine-induced killer cells (CIK). Major hurdles have been identifying actionable tumor antigens and delivering focused cellular therapies to overcome the immunosuppressive and dense fibrotic stroma surrounding the pancreatic cancer. Further studies are needed to explore the limitations faced by cellular therapy in pancreatic cancer and identify novel combination treatment approaches in order to improve clinical outcomes.
Collapse
Affiliation(s)
- Deepak Sherpally
- Department of Internal Medicine, New York Medical College, Metropolitan, New York, NY 10029, USA
| | - Ashish Manne
- Department of Internal Medicine, Division of Medical Oncology, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA;
| |
Collapse
|
5
|
Tanisaka Y, Ryozawa S, Mizuide M, Fujita A, Jinushi R, Watanabe R, Sato R, Yasuda M. The correlation between tumoral CD8 expression and clinical course in patients with unresectable pancreatic cancer using tissue samples acquired by endoscopic ultrasound-guided tissue acquisition. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2025; 32:132-138. [PMID: 39328021 DOI: 10.1002/jhbp.12072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
BACKGROUND The aim of this study was to evaluate the correlation between tumoral CD8 expression and the clinical course in patients with unresectable pancreatic ductal adenocarcinoma (PDAC) using tissue samples acquired by endoscopic ultrasound-guided tissue acquisition (EUS-TA). METHODS Patients with unresectable PDAC who underwent EUS-TA prior to treatment between September 2017 and October 2021 were included. The localization of the CD8-positive areas was qualitatively evaluated. We divided the patients into high and low groups based on the median percentage of CD8-positive areas. The correlation between the number of CD8-positive areas and overall survival was assessed. Furthermore, the response to chemotherapy was assessed in patients who underwent chemotherapy. RESULTS A total of 169 patients were included in the analysis. The median overall survival was 171 days (95% confidence interval [CI]: 86-401). The median CD8-positive area was 0.10% (95% CI: 0.05-0.26). The median overall survival in the high (≥0.1%) and low (<0.1%) CD8-positive groups were 156 and 213.5 days, respectively (p = .33). The number of CD8-positive areas was not correlated with the overall survival and response to chemotherapy (p = .69). CONCLUSIONS Tissue samples acquired using EUS-TA from patients with unresectable PDAC showed that CD8 expression did not affect the clinical course of patients.
Collapse
Affiliation(s)
- Yuki Tanisaka
- Department of Gastroenterology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Shomei Ryozawa
- Department of Gastroenterology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Masafumi Mizuide
- Department of Gastroenterology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Akashi Fujita
- Department of Gastroenterology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Ryuhei Jinushi
- Department of Gastroenterology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Ryuichi Watanabe
- Department of Gastroenterology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Ryo Sato
- Department of Gastroenterology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Masanori Yasuda
- Department of Pathology, Saitama Medical University International Medical Center, Hidaka, Japan
| |
Collapse
|
6
|
Cheng M, Liu J, Liang Y, Xu J, Ma L, Liang J. Tissue-Resident Memory T Cells in Tumor Immunity and Immunotherapy of Digestive System Tumors. Immunol Invest 2025:1-22. [PMID: 39840686 DOI: 10.1080/08820139.2024.2447780] [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: 01/23/2025]
Abstract
Background: Tissue-resident memory T (TRM) cells possess unique abilities to migrate, establish themselves in tissues, and monitor peripheral tissues without circulating. They are crucial in providing long-lasting and local immune protection against surface infections. TRMs demonstrate distinct phenotypic and functional characteristics compared to central memory T (Tcm) cells and effector memory T (Tem) cells.Methods: We reviewed a large number of literature to explore the physiological and functional roles of tissue-resident memory T cells, as well as the link between TRM cells and the development and prognosis of digestive tract tumors. We also investigated the association between TRM cells, intestinal flora, and metabolites.Results: Recent studies have implicated TRMs in the immune response against tumors, making them a potential target for cancer therapy. However, research specifically focused on gastrointestinal tumors is limited.Conclusion: This review aims to compile and assess the most recent data on the role of TRM cells in gastrointestinal tumor immunity. Additionally, it explores recent advancements in immunotherapy and investigates how TRMs may influence intestinal flora and metabolites.
Collapse
Affiliation(s)
- Min Cheng
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| | - Jie Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Yue Liang
- Department of Breast Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of General Surgery (Breast Surgery), The First Affiliated Hospital of Shandong First Medical University (Shandong Provincial Qianfoshan Hospital), Jinan, China
| | - Jiamei Xu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| | - Lin Ma
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| | - Jing Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| |
Collapse
|
7
|
Zhang J, Feng Y, Li D, Shi D. Fungal influence on immune cells and inflammatory responses in the tumor microenvironment (Review). Oncol Lett 2025; 29:50. [PMID: 39564373 PMCID: PMC11574707 DOI: 10.3892/ol.2024.14796] [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: 04/09/2024] [Accepted: 07/25/2024] [Indexed: 11/21/2024] Open
Abstract
In recent years, a growing body of research has highlighted the significant influence of the microbiota on tumor immunity within the tumor microenvironment (TME). While much attention has been given to bacteria, emerging evidence suggests that fungi also play crucial roles in tumor development. The present review aimed to consolidate the latest findings on the mechanisms governing the interactions between fungi and the immune system or TME. By elucidating these intricate mechanisms, novel insights into the modulation of tumor immunity and therapeutic strategies may be uncovered. Ultimately, a deeper understanding of the interplay between fungi and the TME holds promise for the development of innovative management strategies and targeted drugs to enhance tumor therapy efficacy.
Collapse
Affiliation(s)
- Jinke Zhang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington DC 20057, USA
| | - Yahui Feng
- Laboratory of Medical Mycology, Jining No. 1 People's Hospital, Jining, Shandong 272001, P.R. China
| | - Dongmei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington DC 20057, USA
| | - Dongmei Shi
- Laboratory of Medical Mycology, Jining No. 1 People's Hospital, Jining, Shandong 272001, P.R. China
- Department of Dermatology, Jining No. 1 People's Hospital, Jining, Shandong 272001, P.R. China
| |
Collapse
|
8
|
Yaku H, Takahashi K, Okada H, Kobiyama K, Shiokawa M, Uza N, Kodama Y, Ishii KJ, Seno H. Near-infrared photoimmunotherapy as a complementary modality to in situ vaccine in a preclinical pancreatic cancer model. Biochem Biophys Res Commun 2024; 737:150534. [PMID: 39142137 DOI: 10.1016/j.bbrc.2024.150534] [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: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 08/16/2024]
Abstract
Pancreatic cancer is one of the most refractory malignancies. In situ vaccines (ISV), in which intratumorally injected immunostimulatory adjuvants activate innate immunity at the tumor site, utilize tumor-derived patient-specific antigens, thereby allowing for the development of vaccines in patients themselves. Near-infrared photoimmunotherapy (NIR-PIT) is a novel therapy that selectively kills cancer cells exclusively in the NIR-irradiated region. Extending our previous research showing that ISV using the unique nanoparticulate Toll-like receptor 9 (TLR9) ligand K3-SPG induced effective antitumor immunity, here we incorporated NIR-PIT into K3-SPG-ISV so that local tumor destruction by NIR-PIT augments the antitumor effect of ISV. In the mouse model of pancreatic cancer, the combination of K3-SPG-ISV and CD44-targeting NIR-PIT showed synergistic systemic antitumor effects and enhanced anti-programmed cell death-1 (PD-1) blockade. Mechanistically, strong intratumoral upregulation of interferon-related genes and dependency on CD8+ T cells were observed, suggesting the possible role of interferon and cytotoxic T cell responses in the induction of antitumor immunity. Importantly, this combination induced immunological memory in therapeutic and neoadjuvant settings. This study represents the first attempt to integrate NIR-PIT with ISV, offering a promising new direction for cancer immunotherapy, particularly for pancreatic cancer.
Collapse
Affiliation(s)
- Hiroaki Yaku
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ken Takahashi
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Division of Cancer Immunotherapy, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Hirokazu Okada
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Gastroenterology and Hepatology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Kouji Kobiyama
- Division of Vaccine Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center, The University of Tokyo, Tokyo, Japan
| | - Masahiro Shiokawa
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Norimitsu Uza
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Yuzo Kodama
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Ken J Ishii
- Division of Vaccine Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
9
|
Çorbacı K, Gurleyik MG, Gonultas A, Aker F, Gul MO, Tilki M. Evaluation of prognostic significance of histopathological characteristics and tumor-infiltrating lymphocytes for pancreatic cancer survival. Sci Rep 2024; 14:27392. [PMID: 39521901 PMCID: PMC11550438 DOI: 10.1038/s41598-024-79342-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024] Open
Abstract
With a 5-year survival of ˂ 10%, pancreatic cancer is one of the leading causes of cancer-related deaths. Given the role of the distribution of tumor-infiltrating lymphocyte (TILs) subtypes in the tumor and its microenvironment in predicting prognosis, the development of new targeted therapies based on T-cell adaptive response has gained considerable attention. This study aimed to examine the peritumoral spread of TILs and its relationship with other prognostic parameters and survival. This study included 60 patients with pancreatic cancer who had undergone surgery with follow-up between 2011 and 2021. Demographic characteristics, tumor histopathological features, peritumoral TILs counts, and intratumoral programmed cell death protein-1 (PD-1) and programmed death ligand - 1 (PD-L1) positivity were evaluated. Furthermore, overall survival and their efficacy in predicting survival according to TNM stage were analyzed. The number of cluster differentiation-3 positive (CD3 P) TILs increased with advancing pathological T stage. CD3 P and CD8 P TIL counts were higher in patients with peripancreatic fatty tissue invasion. Patients with PD-L1 positivity and higher TIL counts had better survival rates. PD-L1-negative patients with a low CD8 positive/total lymph node count (P/T) ratio had a longer survival. Moreover, patients with poorly differentiated tumors with low CD3 P/T and CD8 P/T ratios had a longer survival. The CD3 P/T and CD8 P/T ratios were compatible with the automatic and manual measurements. Age, tumor differentiation, N stage, and peritumoral TIL count and subtype, when evaluated together with the presence of PD-L1 in the tumor tissue, may have prognostic significance for survival in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Kadir Çorbacı
- General Surgery, Osmaneli Mustafa Selahattin Çetintaş State Hospital, Bilecik, Turkey.
| | - Meryem Gunay Gurleyik
- Department of General Surgery, Haydarpasa Numune Training and Research Hospital, University of Health Sciences, Istanbul, Turkey.
| | | | - Fugen Aker
- Department of Pathology, Haydarpasa Numune Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Mehmet Onur Gul
- Surgical Oncology, Malatya Training and Research Hospital, Malatya, Turkey
| | - Metin Tilki
- Department of General Surgery, Haydarpasa Numune Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
10
|
Rupp L, Dietsche I, Kießler M, Sommer U, Muckenhuber A, Steiger K, van Eijck CWF, Richter L, Istvanffy R, Jäger C, Friess H, van Eijck CHJ, Demir IE, Reyes CM, Schmitz M. Neoadjuvant chemotherapy is associated with suppression of the B cell-centered immune landscape in pancreatic ductal adenocarcinoma. Front Immunol 2024; 15:1378190. [PMID: 38629072 PMCID: PMC11018975 DOI: 10.3389/fimmu.2024.1378190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is typically diagnosed at advanced stages and associated with early distant metastasis and poor survival. Besides clinical factors, the tumor microenvironment (TME) emerged as a crucial determinant of patient survival and therapy response in many tumors, including PDAC. Thus, the presence of tumor-infiltrating lymphocytes and the formation of tertiary lymphoid structures (TLS) is associated with longer survival in PDAC. Although neoadjuvant therapy (NeoTx) has improved the management of locally advanced tumors, detailed insight into its effect on various TME components is limited. While a remodeling towards a proinflammatory state was reported for PDAC-infiltrating T cells, the effect of NeoTx on B cell subsets, including plasma cells, and TLS formation is widely unclear. We thus investigated the frequency, composition, and spatial distribution of PDAC-infiltrating B cells in primary resected (PR) versus neoadjuvant-treated patients using a novel multiplex immunohistochemistry panel. The NeoTx group displayed significantly lower frequencies of pan B cells, GC B cells, plasmablasts, and plasma cells, accompanied by a reduced abundance of TLS. This finding was supported by bulk RNA-sequencing analysis of an independent fresh frozen tissue cohort, which revealed that major B cell pathways were downregulated in the NeoTx group. We further observed that plasma cells frequently formed aggregates that localized close to TLS and that TLS+ patients displayed significantly higher plasma cell frequencies compared to TLS- patients in the PR group. Additionally, high densities of CD20+ intratumoral B cells were significantly associated with longer overall survival in the PR group. While CD20+ B cells held no prognostic value for NeoTx patients, an increased frequency of proliferating CD20+Ki67+ B cells emerged as an independent prognostic factor for longer survival in the NeoTx group. These results indicate that NeoTx differentially affects PDAC-infiltrating immune cells and may have detrimental effects on the existing B cell landscape and the formation of TLS. Gaining further insight into the underlying molecular mechanisms is crucial to overcome the intrinsic immunotherapy resistance of PDAC and develop novel strategies to improve the long-term outcome of PDAC patients.
Collapse
Affiliation(s)
- Luise Rupp
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Ina Dietsche
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Maximilian Kießler
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Neural Influences in Cancer (NIC), International Research Consortium, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Ulrich Sommer
- Institute of Pathology, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Alexander Muckenhuber
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Katja Steiger
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Casper W. F. van Eijck
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Leonard Richter
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Rouzanna Istvanffy
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Carsten Jäger
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Casper H. J. van Eijck
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Neural Influences in Cancer (NIC), International Research Consortium, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Department of General Surgery, Hepato-Pancreato-Biliary (HPB) Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Türkiye
- Else Kröner Clinician Scientist Professor for Translational Pancreatic Surgery, Technical University of Munich, Munich, Germany
| | - Carmen Mota Reyes
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Neural Influences in Cancer (NIC), International Research Consortium, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
11
|
Yamada Y, Yamamoto T, Tsutsumi C, Matsumoto T, Noguchi S, Shimada Y, Nakata K, Ohuchida K, Nakamura M, Oda Y. Immature stroma and high infiltration of CD15 + cells are predictive markers of poor prognosis in different subsets of patients with pancreatic cancer. Cancer Sci 2024; 115:1001-1013. [PMID: 38230840 PMCID: PMC10920995 DOI: 10.1111/cas.16060] [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: 09/06/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 01/18/2024] Open
Abstract
Preoperative treatment is commonly carried out for borderline resectable pancreatic ductal adenocarcinoma (PDAC). However, the relationship between the combination of immune cells in the tumor microenvironment and their intratumoral heterogeneity along with their association with histological findings remains unclear, especially in patients receiving preoperative chemotherapy. We aimed to explore the therapeutic strategies for patients with PDAC with poor prognosis after receiving chemotherapy based on histological and immunological microenvironmental classifications. We investigated the correlation between the prognosis and histological immune microenvironmental factors of patients who initially underwent surgery (n = 100) and were receiving gemcitabine plus nab-paclitaxel (GEM + nabPTX) as preoperative chemotherapy (n = 103). Immune profiles were generated based on immune cell infiltration into the tumor, and their correlation with patient outcomes and histological features was analyzed. Tumor-infiltrating neutrophils (TINs) were identified as independent poor prognostic factors using multivariate analysis in both surgery-first and preoperative chemotherapy groups. The patients were further classified into four groups based on immune cell infiltration into the tumor. Patients with high CD15 infiltration into the tumor and immature stroma at the cancer margins showed the worst prognosis in the preoperative chemotherapy group. The analysis of mRNA expression and immunohistochemical features revealed that CXCR2, the receptor for CXCL8, was correlated with disease-free and overall survival. We inferred that patients with immature stroma at the margins and high infiltration of CD15+ neutrophils within the tumor showed the worst prognosis and they could particularly benefit from treatment with inhibitors targeting CXCR2 or CXCL8.
Collapse
Affiliation(s)
- Yutaka Yamada
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Takeo Yamamoto
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Chikanori Tsutsumi
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Takashi Matsumoto
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Shoko Noguchi
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yuki Shimada
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Kohei Nakata
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Kenoki Ohuchida
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| |
Collapse
|
12
|
Szczepanski JM, Rudolf MA, Shi J. Clinical Evaluation of the Pancreatic Cancer Microenvironment: Opportunities and Challenges. Cancers (Basel) 2024; 16:794. [PMID: 38398185 PMCID: PMC10887250 DOI: 10.3390/cancers16040794] [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: 01/16/2024] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Advances in our understanding of pancreatic ductal adenocarcinoma (PDAC) and its tumor microenvironment (TME) have the potential to transform treatment for the hundreds of thousands of patients who are diagnosed each year. Whereas the clinical assessment of cancer cell genetics has grown increasingly sophisticated and personalized, current protocols to evaluate the TME have lagged, despite evidence that the TME can be heterogeneous within and between patients. Here, we outline current protocols for PDAC diagnosis and management, review novel biomarkers, and highlight potential opportunities and challenges when evaluating the PDAC TME as we prepare to translate emerging TME-directed therapies to the clinic.
Collapse
Affiliation(s)
| | | | - Jiaqi Shi
- Department of Pathology and Clinical Labs, University of Michigan, Ann Arbor, MI 48109, USA; (J.M.S.); (M.A.R.)
| |
Collapse
|
13
|
Katz MHG, Petroni GR, Bauer T, Reilley MJ, Wolpin BM, Stucky CC, Bekaii-Saab TS, Elias R, Merchant N, Dias Costa A, Lenehan P, Cardot-Ruffino V, Rodig S, Pfaff K, Dougan SK, Nowak JA, Varadhachary GR, Slingluff CL, Rahma O. Multicenter randomized controlled trial of neoadjuvant chemoradiotherapy alone or in combination with pembrolizumab in patients with resectable or borderline resectable pancreatic adenocarcinoma. J Immunother Cancer 2023; 11:e007586. [PMID: 38040420 PMCID: PMC10693876 DOI: 10.1136/jitc-2023-007586] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a challenging target for immunotherapy because it has an immunosuppressive tumor microenvironment. Neoadjuvant chemoradiotherapy can increase tumor-infiltrating lymphocyte (TIL) density, which may predict overall survival (OS). We hypothesized that adding programmed cell death protein 1 (PD-1) blockade to chemoradiotherapy would be well tolerated and increase TILs among patients with localized PDAC. METHODS Patients were randomized 2:1 to Arm A (receiving pembrolizumab plus chemoradiotherapy (capecitabine and external beam radiation)) or Arm B (receiving chemoradiotherapy alone) before anticipated pancreatectomy. Primary endpoints were (1) incidence and severity of adverse events during neoadjuvant therapy and (2) density of TILs in resected tumor specimens. TIL density was assessed using multiplexed immunofluorescence histologic examination. RESULTS Thirty-seven patients were randomized to Arms A (n=24) and B (n=13). Grade ≥3 adverse events related to neoadjuvant treatment were experienced by 9 (38%) and 4 (31%) patients in Arms A and B, respectively, with one patient experiencing dose-limiting toxicity in Arm A. Seventeen (71%) and 7 (54%) patients in Arms A and B, respectively, underwent pancreatectomy. Median CD8+ T-cell densities in Arms A and B were 67.4 (IQR: 39.2-141.8) and 37.9 (IQR: 22.9-173.4) cells/mm2, respectively. Arms showed no noticeable differences in density of CD8+Ki67+, CD4+, or CD4+FOXP3+ regulatory T cells; M1-like and M2-like macrophages; or granulocytes. Median OS durations were 27.8 (95% CI: 17.1 to NR) and 24.3 (95% CI: 12.6 to NR) months for Arms A and B, respectively. CONCLUSIONS Adding pembrolizumab to neoadjuvant chemoradiotherapy was safe. However, no convincing effect on CD8+ TILs was observed.
Collapse
Affiliation(s)
- Matthew H G Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gina R Petroni
- Division of Translational Research and Applied Statistics, Department of Public Health Sciences, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Todd Bauer
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Matthew J Reilley
- Division of Hematology and Oncology, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Tanios S Bekaii-Saab
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Mayo Clin, Phoenix, Arizona, USA
| | - Rawad Elias
- Hartford HealthCare Cancer Institute, Plainville, Connecticut, USA
| | - Nipun Merchant
- Department of Surgery, University of Miami, Coral Gables, Florida, USA
| | - Andressa Dias Costa
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Patrick Lenehan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | - Victoire Cardot-Ruffino
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | - Scott Rodig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | - Kathleen Pfaff
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Stephanie K Dougan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan Andrew Nowak
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | - Gauri R Varadhachary
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Craig L Slingluff
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Osama Rahma
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
14
|
van ‘t Land FR, Aziz MH, Michiels N, Mieog JSD, Bonsing BA, Luelmo SA, Homs MY, Groot Koerkamp B, Papageorgiou G, van Eijck CH. Increasing Systemic Immune-inflammation Index During Treatment in Patients With Advanced Pancreatic Cancer is Associated With Poor Survival: A Retrospective, Multicenter, Cohort Study. Ann Surg 2023; 278:1018-1023. [PMID: 37010512 PMCID: PMC10631500 DOI: 10.1097/sla.0000000000005865] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
BACKGROUND AND OBJECTIVES A high systemic immune-inflammation index (SIII) at diagnosis of various cancers, including pancreatic cancer, is associated with poor prognosis. The impact of FOLFIRINOX (5-fluorouracil, leucovorin, irinotecan, and oxaliplatin) chemotherapy or stereotactic body radiotherapy on this index is unknown. In addition, the prognostic value of changes in the SIII during treatment is unclear. In this retrospective analysis, we aimed to find answers regarding patients with advanced pancreatic cancer. METHODS Patients with advanced pancreatic cancer treated with FOLFIRINOX chemotherapy alone or with FOLFIRINOX chemotherapy followed by stereotactic body radiotherapy between 2015 and 2021 in 2 tertiary referral centers were included. Baseline characteristics, laboratory values at 3 time points during treatment, and survival outcomes were collected. The patient-specific evolutions of SIII and their association with mortality were assessed with joint models for longitudinal and time-to-event data. RESULTS Data of 141 patients were analyzed. At a median follow-up time of 23.0 months (95% CI: 14.6-31.3), 97 (69%) patients had died. Median overall survival was 13.2 months (95% CI: 11.0-15.5). During treatment with FOLFIRINOX, the log (SIII) was reduced by -0.588 (95% CI: -0.0978, -0.197; P = 0.003). One unit increase in log (SIII) increased the hazard ratio of dying by 1.604 (95% CI: 1.068-2.409; P = 0.023). CONCLUSIONS In addition to carbohydrate antigen 19-9, the SIII is a reliable biomarker in patients with advanced pancreatic cancer.
Collapse
Affiliation(s)
| | - Mohammad H. Aziz
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Nynke Michiels
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - J. Sven D. Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Bert A. Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Saskia A.C. Luelmo
- Department of Oncology, Leiden University Medical Center, the Netherlands
| | - Marjolein Y.V. Homs
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Grigorios Papageorgiou
- Department of Biostatistics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | | |
Collapse
|
15
|
Schardey J, Lu C, Neumann J, Wirth U, Li Q, Jiang T, Zimmermann P, Andrassy J, Bazhin AV, Werner J, Kühn F. Differential Immune Infiltration Profiles in Colitis-Associated Colorectal Cancer versus Sporadic Colorectal Cancer. Cancers (Basel) 2023; 15:4743. [PMID: 37835436 PMCID: PMC10571767 DOI: 10.3390/cancers15194743] [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: 08/13/2023] [Revised: 09/21/2023] [Accepted: 09/24/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Chronic inflammation is a significant factor in colorectal cancer (CRC) development, especially in colitis-associated CRC (CAC). T-cell exhaustion is known to influence inflammatory bowel disease (IBD) progression and antitumor immunity in IBD patients. This study aimed to identify unique immune infiltration characteristics in CAC patients. METHODS We studied 20 CAC and 20 sporadic CRC (sCRC) patients, who were matched by tumor stage, grade, and location. Immunohistochemical staining targeted various T-cell markers (CD3, CD4, CD8, and FOXP3), T-cell exhaustion markers (TOX and TIGIT), a B-cell marker (CD20), and a neutrophil marker (CD66b) in tumor and tumor-free mucosa from both groups. The quantification of the tumor immune stroma algorithm assessed immune-infiltrating cells. RESULTS CAC patients had significantly lower TOX+ cell infiltration than sCRC in tumors (p = 0.02) and paracancerous tissues (p < 0.01). Right-sided CAC showed increased infiltration of TOX+ cells (p = 0.01), FOXP3+ regulatory T-cells (p < 0.01), and CD20+ B-cells (p < 0.01) compared to left-sided CAC. In sCRC, higher tumor stages (III and IV) had significantly lower TIGIT+ infiltrate than stages I and II. In CAC, high CD3+ (p < 0.01) and CD20+ (p < 0.01) infiltrates correlated with improved overall survival. In sCRC, better survival was associated with decreased TIGIT+ cells (p < 0.038) and reduced CD8+ infiltrates (p = 0.02). CONCLUSION In CAC, high CD3+ and CD20+ infiltrates relate to improved survival, while this association is absent in sCRC. The study revealed marked differences in TIGIT and TOX expression, emphasizing distinctions between CAC and sCRC. T-cell exhaustion appears to have a different role in CAC development.
Collapse
Affiliation(s)
- Josefine Schardey
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians-University Hospital Munich, 81377 Munich, Germany
| | - Can Lu
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians-University Hospital Munich, 81377 Munich, Germany
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education & Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Zhejiang Provincial Clinical Research Center for CANCER & Cancer Center of Zhejiang University, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jens Neumann
- Department of Pathology, Ludwig-Maximilians University, 81377 Munich, Germany
| | - Ulrich Wirth
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians-University Hospital Munich, 81377 Munich, Germany
| | - Qiang Li
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians-University Hospital Munich, 81377 Munich, Germany
| | - Tianxiao Jiang
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians-University Hospital Munich, 81377 Munich, Germany
| | - Petra Zimmermann
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians-University Hospital Munich, 81377 Munich, Germany
| | - Joachim Andrassy
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians-University Hospital Munich, 81377 Munich, Germany
| | - Alexandr V. Bazhin
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians-University Hospital Munich, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Jens Werner
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians-University Hospital Munich, 81377 Munich, Germany
| | - Florian Kühn
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians-University Hospital Munich, 81377 Munich, Germany
| |
Collapse
|
16
|
Knipper K, Lyu SI, Goebel H, Damanakis AI, Zhao Y, Bruns CJ, Schmidt T, Kashkar H, Quaas A, Schiffmann LM, Popp FC. X-linked inhibitor of apoptosis protein is a prognostic marker for a favorable outcome in three identified subsets in resectable adenocarcinoma of the pancreas. J Cancer Res Clin Oncol 2023; 149:5531-5538. [PMID: 36472768 PMCID: PMC10356682 DOI: 10.1007/s00432-022-04476-2] [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: 09/19/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) is currently one of the leading causes of cancer death worldwide. Therefore, building further subgroups as well as enabling individual patient therapy and diagnostics are needed. X-linked inhibitor of apoptosis protein (XIAP) is known to modulate apoptotic and inflammatory pathways. Its expression was found to correlate with patients' survival in other tumor entities. This study aims to examine the role of XIAP in patients with PDAC in relation to the inflammatory microenvironment. METHODS The PANCALYZE multicenter study group included 257 patients with PDAC. Paraffin-embedded tumor samples were stained immunohistochemically for CD3, CD20, CD38, CD56, CD66b, CD117, and CD163 and XIAP. These stainings were further analyzed digitally with QuPath and survival analyses were done. RESULTS XIAP-positive patients with T-cell, respectively, neutrophil enriched tumors survived significantly longer compared to XIAP-negative patients (CD3: 37.6 vs. 24.6 months, p = 0.028; CD66b: 34.1 vs. 14.9 months, p = 0.027). Additionally, XIAP-positive patients showed better survival in the lymph node-negative population (48.4 vs. 24.2 months, p = 0.019). Regarding the total population, our findings did not show a correlation between XIAP expression and survival. In multivariate cox regression analyzes XIAP proves to be an independent factor for better survival in the identified subgroups (CD3: p = 0.043; CD66b: p = 0.012, N0: p = 0.040). CONCLUSION We found XIAP-positive subgroups with significantly better survival in patients with PDAC in T-cell-rich, neutrophil-rich, or lymph node-negative cohorts. This could lead to further individualized cancer treatment with less aggressive therapy protocols for XIAP-positive tumors or more intensive follow-up for XIAP-negative tumors.
Collapse
Affiliation(s)
- Karl Knipper
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Su Ir Lyu
- Faculty of Medicine, Institute of Pathology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Heike Goebel
- Faculty of Medicine, Institute of Pathology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Alexander I Damanakis
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Yue Zhao
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Christiane J Bruns
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Thomas Schmidt
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Hamid Kashkar
- Faculty of Medicine, Institute for Molecular Immunology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Faculty of Medicine, Institute of Pathology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Lars M Schiffmann
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Felix C Popp
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
17
|
Wang T, Zhang D, Tang D, Heng Y, Lu LM, Tao L. The role of systemic inflammatory response index (SIRI) and tumor-infiltrating lymphocytes (TILs) in the prognosis of patients with laryngeal squamous cell carcinoma. J Cancer Res Clin Oncol 2023; 149:5627-5636. [PMID: 36520215 DOI: 10.1007/s00432-022-04469-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/06/2022] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Systemic inflammatory response index (SIRI) values and tumor-infiltrating lymphocytes (TILs) are associated with the prognosis of various tumors. There is minimal evidence of those two as prognostic markers in laryngeal squamous cell carcinoma (LSCC). In this study, we aimed to examine the predictive value of SIRI and tumor-infiltrating CD3+/CD4+/CD8+ T cells in the prognosis of patients who underwent partial or total laryngectomy. STUDY DESIGN A total of 78 patients with LSCC who underwent total or partial laryngectomy at the Eye, Ear, Nose, and Throat Hospital of Fudan University between 2013 and 2015 were retrospectively analyzed. METHODS The tumor tissues of 78 LSCC patients were retrospectively evaluated using immunohistochemical staining for CD3+ /CD4+ /CD8+ -cells. The overall survival (OS) and disease-free survival (DFS) rates were recorded using the Kaplan-Meier method. RESULTS Patients with high immunoscore (IS) (3-4) had prolonged survival (P < 0.001 for OS). High SIRI values were independently associated with poorer OS and DFS (P = 0.018 for OS; P = 0.016 for DFS). CD8+ TILs and SIRI values showed a- negative association (P < 0.01). Patients with low SIRI values and high IS had better 5-year OS and DFS than those with high SIRI values and low IS (P < 0.001 for OS; P = 0.0014 for DFS). Patients with 'hot' tumor had a higher 5-year OS than those with 'excluded' or 'cold' phenotype. CONCLUSIONS The SIRI values and the density of TILs may help predict LSCC patients' outcomes after surgery. The combination of SIRI and IS may be a new component of the tumor, nodes, and metastases (TNM) classification of cancer and prognostic factor for T-cell-target immunotherapy.
Collapse
Affiliation(s)
- Tian Wang
- Department of Otolaryngology-HNS, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University School of Medicine, 83 Fenyang Road, Shanghai, 200031, People's Republic of China
| | - Duo Zhang
- Department of Otolaryngology-HNS, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University School of Medicine, 83 Fenyang Road, Shanghai, 200031, People's Republic of China
| | - Di Tang
- Department of Otolaryngology-HNS, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University School of Medicine, 83 Fenyang Road, Shanghai, 200031, People's Republic of China
| | - Yu Heng
- Department of Otolaryngology-HNS, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University School of Medicine, 83 Fenyang Road, Shanghai, 200031, People's Republic of China
| | - Li-Ming Lu
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, People's Republic of China.
| | - Lei Tao
- Department of Otolaryngology-HNS, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University School of Medicine, 83 Fenyang Road, Shanghai, 200031, People's Republic of China.
| |
Collapse
|
18
|
Giustarini G, Teng G, Pavesi A, Adriani G. Characterization of 3D heterocellular spheroids of pancreatic ductal adenocarcinoma for the study of cell interactions in the tumor immune microenvironment. Front Oncol 2023; 13:1156769. [PMID: 37519820 PMCID: PMC10375712 DOI: 10.3389/fonc.2023.1156769] [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: 02/01/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies nowadays. The available chemo- and immunotherapies are often ineffective in treating PDAC due to its immunosuppressive and highly desmoplastic tumor immune microenvironment (TIME), which is hardly reproduced in the existing preclinical models. The PDAC TIME results from a peculiar spatial organization between different cell types. For this reason, developing new human models recapitulating the tissue organization and cell heterogeneity of PDAC is highly desirable. We developed human 3D heterocellular tumor spheroids of PDAC formed by cancer cells, endothelial cells, pancreatic stellate cells (PSC), and monocytes. As a control, we formed spheroids using immortalized epithelial pancreatic ductal cells (non-cancerous spheroids) with cellular heterogeneity similar to the tumor spheroids. Normal spheroids containing endothelial cells formed a complex 3D endothelial network significantly compromised in tumor spheroids. Monocyte/macrophages within the 4-culture tumor spheroids were characterized by a higher expression of CD163, CD206, PD-L1, and CD40 than those in the non-cancerous spheroids suggesting their differentiation towards an immunosuppressive phenotype. The heterocellular tumor spheroids presented a hypoxic core populated with PSC and monocytes/macrophages. The 4-culture tumor spheroids were characterized by spatial proximity of PSC and monocytes to the endothelial cells and a cytokine signature with increased concentrations of CXCL10, CCL2, and IL-6, which have been observed in PDAC patients and associated with poor survival. Further, 4-culture tumor spheroids decreased the concentrations of T-cell chemoattracting cytokines, i.e., CCL4, CCL5, and CXCL9, when compared with the non-cancerous spheroids, revealing a critical immunosuppressive feature of the different types of cells forming the tumor spheroids. Our results showed that the 4-culture tumor spheroids better resembled some critical features of patients' PDAC TIME than monoculture tumor spheroids. Using the proposed human 3D spheroid model for therapy testing at the preclinical stage may reveal pitfalls of chemo- and immuno-therapies to help the development of better anti-tumor therapies.
Collapse
Affiliation(s)
- Giulio Giustarini
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Germaine Teng
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Andrea Pavesi
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Giulia Adriani
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| |
Collapse
|
19
|
Zong G, Wang X, Guo X, Zhao Q, Wang C, Shen S, Xiao W, Yang Q, Jiang W, Shen J, Wan R. NAT10-mediated AXL mRNA N4-acetylcytidine modification promotes pancreatic carcinoma progression. Exp Cell Res 2023; 428:113620. [PMID: 37156457 DOI: 10.1016/j.yexcr.2023.113620] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 04/27/2023] [Accepted: 04/29/2023] [Indexed: 05/10/2023]
Abstract
Although the patient's survival time in various cancers has significantly increased in recent decades, the overall 5-year survival rate of pancreatic ductal adenocarcinoma (PDAC) has remained virtually unchanged due to rapid progression and metastasis. While N-acetyltransferase 10 (NAT10) has been identified as a regulator of mRNA acetylation in many malignancies, its role in PDAC remains unclear. Here, we found that NAT10 mRNA and protein levels were upregulated in PDAC tissues. Increased NAT10 protein expression was significantly correlated with poor prognosis in PDAC patients. Through our experiments, we demonstrated that NAT10 acted as an oncogene to promote PDAC tumorigenesis and metastasis in vitro and in vivo. Mechanistically, NAT10 exerts its oncogenic effects by promoting mRNA stability of receptor tyrosine kinase AXL in an ac4C-dependent manner leading to increased AXL expression and further promoting PDAC cell proliferation and metastasis. Together, our findings highlight the critical of NAT10 in PDAC progression and reveal a novel epigenetic mechanism by which modified mRNA acetylation promotes PDAC metastasis.
Collapse
Affiliation(s)
- Guanzhao Zong
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xing Wang
- First Affiliated Hospital of Jiangxi Medical College, China
| | - Xingya Guo
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiuyan Zhao
- Department of Gastroenterology, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, China
| | - Chuanyang Wang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shien Shen
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenqin Xiao
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingqing Yang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiliang Jiang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Shen
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Rong Wan
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
20
|
Ogobuiro I, Baca Y, Ribeiro JR, Walker P, Wilson GC, Gulhati P, Marshall JL, Shroff RT, Spetzler D, Oberley MJ, Abbott DE, Kim HJ, Kooby DA, Maithel SK, Ahmad SA, Merchant NB, Xiu J, Hosein PJ, Datta J. Multi-omic characterization reveals a distinct molecular landscape in young-onset pancreatic cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.28.23287894. [PMID: 37034762 PMCID: PMC10081424 DOI: 10.1101/2023.03.28.23287894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Purpose Using a real-world database with matched genomic-transcriptomic molecular data, we sought to characterize the distinct molecular correlates underlying clinical differences between young-onset pancreatic cancer (YOPC; <50-yrs.) and average-onset pancreatic cancer (AOPC; ≥70-yrs.) patients. Methods We analyzed matched whole-transcriptome and DNA sequencing data from 2430 patient samples (YOPC, n=292; AOPC, n=2138) from the Caris Life Sciences database (Phoenix, AZ). Immune deconvolution was performed using the quanTIseq pipeline. Overall survival (OS) data was obtained from insurance claims (n=4928); Kaplan-Meier estimates were calculated for age-and molecularly-defined cohorts. Significance was determined as FDR-corrected P -values ( Q )<0.05. Results YOPC patients had higher proportions of mismatch repair-deficient (dMMR)/microsatellite instability-high (MSI-H), BRCA2 -mutant, and PALB2 -mutant tumors compared with AOPC patients, but fewer SMAD4-, RNF43-, CDKN2A- , and SF3B1- mutant tumors. Notably, YOPC patients demonstrated significantly lower incidence of KRAS mutations compared with AOPC patients (81.3% vs. 90.9%; Q =0.004). In the KRAS- wildtype subset (n=227), YOPC tumors demonstrated fewer TP53 mutations and were more likely driven by NRG1 and MET fusions, while BRAF fusions were exclusively observed in AOPC patients. Immune deconvolution revealed significant enrichment of natural killer (NK) cells, CD8 + T cells, monocytes, and M2 macrophages in YOPC patients relative to AOPC patients, which corresponded with lower rates of HLA-DPA1 homozygosity. There was an association with improved OS in YOPC patients compared with AOPC patients with KRAS -wildtype tumors (median 16.2 [YOPC- KRAS WT ] vs. 10.6 [AOPC- KRAS WT ] months; P =0.008) but not KRAS -mutant tumors ( P =0.084). Conclusion In this large, real-world multi-omic characterization of age-stratified molecular differences in PDAC, YOPC is associated with a distinct molecular landscape that has prognostic and therapeutic implications.
Collapse
|
21
|
Liu H, Li Z, Han X, Li Z, Zhao Y, Liu F, Zhu Z, Lv Y, Liu Z, Zhang N. The prognostic impact of tumor-infiltrating B lymphocytes in patients with solid malignancies: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2023; 181:103893. [PMID: 36481308 DOI: 10.1016/j.critrevonc.2022.103893] [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: 08/11/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
This study reviewed the prognostic effect of tumor-infiltrating B lymphocytes (TIBLs) on solid malignancies, to determine the potential role of TIBLs in predicting cancer patient's prognosis and their response to immunotherapy. A total of 45 original papers involving 11,099 individual patients were included in this meta-analysis covering 7 kinds of cancer. The pooled results suggested that high levels of TIBLs were correlated with favorable OS in lung, esophageal, gastric, colorectal, liver, and breast cancer; improved RFS in lung cancer; and improved DFS in gastrointestinal neoplasms. Additionally, TIBLs were significantly correlated with negative lymphatic invasion in gastric cancer, small tumor size in hepatocellular carcinoma, and negative distant metastasis in colorectal cancer. Additionally, TIBLs were reported as a discriminative feature of patients treated with immunotherapy with improved survival. We concluded that TIBLs play a favorable prognostic role among the common solid malignancie, providing theoretical evidence for further prognosis prediction for solid tumors.
Collapse
Affiliation(s)
- Hao Liu
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhuoqun Li
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xuan Han
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Zhujun Li
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yan Zhao
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Fenghua Liu
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Ziyu Zhu
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yi Lv
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Zhijun Liu
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| | - Nana Zhang
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
22
|
Kanno H, Hisaka T, Fujiyoshi K, Akiba J, Hashimoto K, Fujita F, Akagi Y. Prognostic Significance of the Histopathological Growth Pattern and Tumor-Infiltrating Lymphocytes in Stratifying Survival After Hepatectomy for Colorectal Liver Metastases. Ann Surg Oncol 2022; 30:3139-3147. [PMID: 36520232 DOI: 10.1245/s10434-022-12905-2] [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: 09/20/2022] [Accepted: 11/16/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND A histopathological growth pattern (HGP) occurs at the interface between tumor cells and the surrounding liver parenchyma. Desmoplastic HGP (dHGP) is associated with a favorable prognosis and shows denser infiltration of lymphocytes than other HGPs. Tumor-infiltrating lymphocytes (TILs) exert antitumor immunity, nonetheless, their prognostic significance in patients with dHGP is unknown. This study aimed to identify the prognostic significance of HGP and TILs in colorectal liver metastasis (CRLM). METHODS The study analyzed 140 patients who underwent hepatectomy for CRLM. Depending on the type of HGP and TIL, the patients were categorized into four groups (dHGP/high TIL, dHGP/low TIL, non-dHGP/high TIL, and non-dHGP/low TIL) for a comparison of their recurrence-free survival (RFS) and overall survival (OS). Uni- and multivariate analyses were performed using a Cox proportional hazards model. RESULTS The RFS and OS curves differed significantly between the groups. The multivariate analysis showed that a combination of HGP and TIL could stratify the recurrence and survival outcomes. CONCLUSION This study indicated that a combination of HGP and TIL can stratify the risk of survival after hepatectomy in patients with CRLM.
Collapse
Affiliation(s)
- Hiroki Kanno
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan.
| | - Toru Hisaka
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Kenji Fujiyoshi
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Jun Akiba
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Japan
| | - Kazuaki Hashimoto
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Fumihiko Fujita
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Yoshito Akagi
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
23
|
Bhatia R, Bhyravbhatla N, Kisling A, Li X, Batra SK, Kumar S. Cytokines chattering in pancreatic ductal adenocarcinoma tumor microenvironment. Semin Cancer Biol 2022; 86:499-510. [PMID: 35346801 PMCID: PMC9510605 DOI: 10.1016/j.semcancer.2022.03.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) tumor microenvironment (TME) consists of multiple cell types interspersed by dense fibrous stroma. These cells communicate through low molecular weight signaling molecules called cytokines. The cytokines, through their receptors, facilitate PDAC initiation, progression, metastasis, and distant colonization of malignant cells. These signaling mediators secreted from tumor-associated macrophages, and cancer-associated fibroblasts in conjunction with oncogenic Kras mutation initiate acinar to ductal metaplasia (ADM), resulting in the appearance of early preneoplastic lesions. Further, M1- and M2-polarized macrophages provide proinflammatory conditions and promote deposition of extracellular matrix, whereas myofibroblasts and T-lymphocytes, such as Th17 and T-regulatory cells, create a fibroinflammatory and immunosuppressive environment with a significantly reduced cytotoxic T-cell population. During PDAC progression, cytokines regulate the expression of various oncogenic regulators such as NFκB, c-myc, growth factor receptors, and mucins resulting in the formation of high-grade PanIN lesions, epithelial to mesenchymal transition, invasion, and extravasation of malignant cells, and metastasis. During metastasis, PDAC cells colonize at the premetastatic niche created in the liver, and lung, an organotropic function primarily executed by cytokines in circulation or loaded in the exosomes from the primary tumor cells. The indispensable contribution of these cytokines at every stage of PDAC tumorigenesis makes them exciting candidates in combination with immune-, chemo- and targeted radiation therapy.
Collapse
Affiliation(s)
- Rakesh Bhatia
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Namita Bhyravbhatla
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Andrew Kisling
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xiaoqi Li
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Fred and Pamela Buffett Cancer Center, Omaha, NE, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Fred and Pamela Buffett Cancer Center, Omaha, NE, USA.
| |
Collapse
|
24
|
Kazemi MH, Sadri M, Najafi A, Rahimi A, Baghernejadan Z, Khorramdelazad H, Falak R. Tumor-infiltrating lymphocytes for treatment of solid tumors: It takes two to tango? Front Immunol 2022; 13:1018962. [PMID: 36389779 PMCID: PMC9651159 DOI: 10.3389/fimmu.2022.1018962] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/14/2022] [Indexed: 07/30/2023] Open
Abstract
Tumor-infiltrating lymphocytes (TILs), frontline soldiers of the adaptive immune system, are recruited into the tumor site to fight against tumors. However, their small number and reduced activity limit their ability to overcome the tumor. Enhancement of TILs number and activity against tumors has been of interest for a long time. A lack of knowledge about the tumor microenvironment (TME) has limited success in primary TIL therapies. Although the advent of engineered T cells has revolutionized the immunotherapy methods of hematologic cancers, the heterogeneity of solid tumors warrants the application of TILs with a wide range of specificity. Recent advances in understanding TME, immune exhaustion, and immune checkpoints have paved the way for TIL therapy regimens. Nowadays, TIL therapy has regained attention as a safe personalized immunotherapy, and currently, several clinical trials are evaluating the efficacy of TIL therapy in patients who have failed conventional immunotherapies. Gaining favorable outcomes following TIL therapy of patients with metastatic melanoma, cervical cancer, ovarian cancer, and breast cancer has raised hope in patients with refractory solid tumors, too. Nevertheless, TIL therapy procedures face several challenges, such as high cost, timely expansion, and technical challenges in selecting and activating the cells. Herein, we reviewed the recent advances in the TIL therapy of solid tumors and discussed the challenges and perspectives.
Collapse
Affiliation(s)
- Mohammad Hossein Kazemi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Sadri
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Najafi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Rahimi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Baghernejadan
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Gough MJ, Crittenden MR. The paradox of radiation and T cells in tumors. Neoplasia 2022; 31:100808. [PMID: 35691060 PMCID: PMC9194456 DOI: 10.1016/j.neo.2022.100808] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/05/2022] [Accepted: 05/13/2022] [Indexed: 10/27/2022]
Abstract
In this review we consider what appears to be a paradox in immunotherapies based around radiation therapy. The paradox is based on three main points. 1. That T cells are needed for radiation's efficacy; 2. That tumor-specific T cells are enriched in the field of treatment; and 3. That radiation kills T cells in the treatment field. We discuss evidence of the effect of radiation on T cells in the field given their ongoing movement in and out of tissues and the tumor, and how the movement of T cells impacts the treated primary tumor and untreated distant metastases. Given this evidence, we revisit the paradox to understand how the extraordinary efficacy of radiation and immunity in preclinical models is dependent on this radiation sensitive cell.
Collapse
Affiliation(s)
- Michael J Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St., Portland, OR 97213, USA.
| | - Marka R Crittenden
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St., Portland, OR 97213, USA; The Oregon Clinic, Portland, OR, 97213, USA
| |
Collapse
|
26
|
Li Q, Yu J, Zhang H, Meng Y, Liu YF, Jiang H, Zhu M, Li N, Zhou J, Liu F, Fang X, Li J, Feng X, Lu J, Shao C, Bian Y. Prediction of Tumor-Infiltrating CD20 + B-Cells in Patients with Pancreatic Ductal Adenocarcinoma Using a Multilayer Perceptron Network Classifier Based on Non-contrast MRI. Acad Radiol 2022; 29:e167-e177. [PMID: 34922828 DOI: 10.1016/j.acra.2021.11.013] [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: 08/19/2021] [Revised: 11/13/2021] [Accepted: 11/17/2021] [Indexed: 11/01/2022]
Abstract
RATIONALE AND OBJECTIVES Conventional chemotherapy has limited benefit in pancreatic ductal adenocarcinoma (PDAC), necessitating identification of novel therapeutic targets. Radiomics may enable non-invasive prediction of CD20 expression, a hypothesized therapeutic target in PDAC. To develop a machine learning classifier based on noncontrast magnetic resonance imaging for predicting CD20 expression in PDAC. MATERIALS AND METHODS Retrospective study was conducted on preoperative noncontrast magnetic resonance imaging of 156 patients with pathologically confirmed PDAC from January 2017 to April 2018. For each patient, 1409 radiomics features were selected using minimum absolute contraction and selective operator logistic regression algorithms. CD20 expression was quantified using immunohistochemistry. A multilayer perceptron network classifier was developed using the training and validation set. RESULTS A log-rank test showed that the CD20-high group (22.37 months, 95% CI: 19.10-25.65) had significantly longer survival than the CD20-low group (14.9 months, 95% CI: 10.96-18.84). The predictive model showed good differentiation in training (area under the curve [AUC], 0.79) and validation (AUC, 0.79) sets. Sensitivity, specificity, accuracy, positive predictive value, and negative predictive value were 73.21%, 75.47%, 0.74, 0.76, and 0.73, respectively, for the training set and 69.23%, 80.95%, 0.74, 0.82, and 0.68, respectively, for the validation set. CONCLUSION Multilayer perceptron classifier based on noncontrast magnetic resonance imaging scanning can predict the level of CD20 expression in PDAC patients.
Collapse
Affiliation(s)
- Qi Li
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Jieyu Yu
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Hao Zhang
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yinghao Meng
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yan Fang Liu
- Department of Pathology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hui Jiang
- Department of Pathology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Mengmeng Zhu
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Na Li
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Jian Zhou
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Fang Liu
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Xu Fang
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Jing Li
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Xiaochen Feng
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Jianping Lu
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Chengwei Shao
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yun Bian
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China.
| |
Collapse
|
27
|
Shen F, Liu C, Zhang W, He S, Wang F, Wang J, Li Q, Zhou F. Serum levels of IL-6 and CRP can predict the efficacy of mFOLFIRINOX in patients with advanced pancreatic cancer. Front Oncol 2022; 12:964115. [PMID: 35965580 PMCID: PMC9372918 DOI: 10.3389/fonc.2022.964115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/05/2022] [Indexed: 12/24/2022] Open
Abstract
Objectives There is an urgent need for biomarkers that predict the survival outcome of patients diagnosed with metastatic pancreatic cancer, undergoing systemic chemotherapy. This study aimed to identify biomarkers associated with the survival of mPC patients treated with modified FOLFIRINOX (mFOLFIRINOX) as first-line chemotherapy. Methods This was a retrospective study of 30 patients with mPC who received mFOLFIRINOX between October 2018 and March 2021. Data on carcinoembryonic antigen (CEA), cancer antigen (CA)199, interleukin (IL)-6, C-reactive protein (CRP), neutrophils, platelets, lymphocytes, and albumin were collected and dichotomized using the upper or lower limit, as appropriate. These markers were examined for their association with progression-free survival (PFS). A receiver operating characteristic (ROC) curve analysis was used to explore a suitable model to predict mFOLFIRINOX effectiveness. Results IL-6 and CRP levels were associated with poor progression (P = 0.004 and P = <0.001, respectively) of mPC. The high IL-6 level was an independent poor prognostic factor for PFS (HR=4.66, 95%CI: 1.32-16.37, P=0.016) in the multivariable analysis. Patients with high IL-6 levels had a shorter PFS than those with low IL-6 levels (median PFS: 257 vs. 150 days, P=0.020). An increase in IL-6 and CRP levels during chemotherapy positively correlated with disease progression (P = <0.001 for both). The model combining IL-6 with CRP levels helped predict the outcomes of mPC patients treated with mFOLFIRINOX (AUC: 0.811, 95%CI: 0.639-0.983, P=0.003). Conclusions The serum levels of IL-6 and CRP might be considered as valuable biomarkers in predicting the outcomes of patients with mPC who received the mFOLFIRINOX regimen.
Collapse
Affiliation(s)
- Feifei Shen
- Department of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chuan Liu
- Department of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weiguo Zhang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Sijia He
- Department of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fan Wang
- Department of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jingjue Wang
- Department of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qi Li
- Department of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fei Zhou
- Department of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- *Correspondence: Fei Zhou,
| |
Collapse
|
28
|
Gao Q, Wang S, Li F, Lian J, Cheng S, Yue D, Zhang Z, Liu S, Ren F, Zhang D, Wang S, Wang L, Zhang Y. High Mobility Group Protein B1 Decreases Surface Localization of PD-1 to Augment T-cell Activation. Cancer Immunol Res 2022; 10:844-855. [PMID: 35580259 DOI: 10.1158/2326-6066.cir-21-0652] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 01/13/2022] [Accepted: 05/12/2022] [Indexed: 11/16/2022]
Abstract
High-mobility group protein B1 (HMGB1) is a danger signaling molecule that has been found to trigger an effective antitumor immune response. However, the mechanisms underlying its antitumor effects are not fully understood. Here, we found that HMGB1 release induced by chemotherapy in patients with non-small cell lung cancer was negatively correlated with PD-1 expression on CD8+ T cells. In vitro analysis indicated that treatment with HMGB1 led to a significant decrease in the level of expression of PD-1 on CD8+ T cells. Further analysis demonstrated that HMGB1 reduced PD-1 expression by inducing dynamin-mediated internalization of the protein, leading to early endocytosis in the cytoplasm, and subsequently degradation in the lysosomes. In a xenograft model, HER2-targeted chimeric antigen receptor (CAR) T cells had enhanced function in the presence of HMGB1. These data identify a role for HMGB1 as a negative regulator of PD-1 signaling in lung cancer and the observed antitumor effect of HMGB1 on CAR T cells may provide a theoretical foundation for a new immunotherapy combination.
Collapse
Affiliation(s)
- Qun Gao
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Shumin Wang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Feng Li
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Jingyao Lian
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Shaoyan Cheng
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Dongli Yue
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Zhen Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Shasha Liu
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Feifei Ren
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Daiqun Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Shengdian Wang
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
| | - Liping Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, Henan, P.R. China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, P.R. China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, P.R. China
| |
Collapse
|
29
|
Reactive Oxygen Species Bridge the Gap between Chronic Inflammation and Tumor Development. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2606928. [PMID: 35799889 PMCID: PMC9256443 DOI: 10.1155/2022/2606928] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/10/2022] [Indexed: 02/07/2023]
Abstract
According to numerous animal studies, adverse environmental stimuli, including physical, chemical, and biological factors, can cause low-grade chronic inflammation and subsequent tumor development. Human epidemiological evidence has confirmed the close relationship between chronic inflammation and tumorigenesis. However, the mechanisms driving the development of persistent inflammation toward tumorigenesis remain unclear. In this study, we assess the potential role of reactive oxygen species (ROS) and associated mechanisms in modulating inflammation-induced tumorigenesis. Recent reports have emphasized the cross-talk between oxidative stress and inflammation in many pathological processes. Exposure to carcinogenic environmental hazards may lead to oxidative damage, which further stimulates the infiltration of various types of inflammatory cells. In turn, increased cytokine and chemokine release from inflammatory cells promotes ROS production in chronic lesions, even in the absence of hazardous stimuli. Moreover, ROS not only cause DNA damage but also participate in cell proliferation, differentiation, and apoptosis by modulating several transcription factors and signaling pathways. We summarize how changes in the redox state can trigger the development of chronic inflammatory lesions into tumors. Generally, cancer cells require an appropriate inflammatory microenvironment to support their growth, spread, and metastasis, and ROS may provide the necessary catalyst for inflammation-driven cancer. In conclusion, ROS bridge the gap between chronic inflammation and tumor development; therefore, targeting ROS and inflammation represents a new avenue for the prevention and treatment of cancer.
Collapse
|
30
|
Zhang JJ, Shao C, Yin YX, Sun Q, Li YN, Zha YW, Li MY, Hu BL. Hypoxia-Related Signature Is a Prognostic Biomarker of Pancreatic Cancer. DISEASE MARKERS 2022; 2022:6449997. [PMID: 35789607 PMCID: PMC9250441 DOI: 10.1155/2022/6449997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/20/2022] [Accepted: 06/04/2022] [Indexed: 11/20/2022]
Abstract
Background Hypoxia plays a significant role in the pathogenesis of pancreatic cancer, but the effect of hypoxia-related genes in pancreatic cancer remains to be elucidated. This study aimed to identify hypoxia-related genes related to pancreatic cancer and construct a prognostic signature. Methods Pancreatic cancer datasets were retrieved from TCGA database. Cox regression analyses were used to identify hypoxia-related genes and construct a prognostic signature. Datasets from International Cancer Genome Consortium and GEO databases were used as validated cohorts. The CIBERSORT method was applied to estimate the fractions of immune cell types. DNA methylation and protein levels of the genes in pancreatic cancer were examined. Results Three hypoxia-related genes (TES, LDHA, and ANXA2) were identified as associated with patient survival and selected to construct a prognostic signature. Patients were divided into high- and low-risk groups based on the signature. Those in the high-risk group showed worse survival than those in the low-risk group. The signature was shown to be involved in the HIF-1 signaling pathway. The time-dependent ROC analyses of three independent validated cohorts further revealed that this signature had a better prognostic value in the prediction of the survival of pancreatic cancer patients. Immune cells analysis for three datasets demonstrated that high-risk signature was significantly associated with macrophages and T cells. DNA methylation and protein levels of the three genes validated their aberrant expression in pancreatic cancer. Conclusions Our research provided a novel and reliable prognostic signature that composes of three hypoxia-related genes to estimate the prognosis of pancreatic cancer.
Collapse
Affiliation(s)
- Jing-jing Zhang
- Cancer Institute of Zhongshan City People's Hospital, Zhongshan, 528403 Guangdong, China
| | - Chao Shao
- Cancer Institute of Zhongshan City People's Hospital, Zhongshan, 528403 Guangdong, China
| | - Yi-xin Yin
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, 530021 Guangxi, China
| | - Qiang Sun
- Department of Hepatobiliary Surgery, Zhongshan City People's Hospital, Zhongshan, 528403 Guangdong, China
| | - Ya-ni Li
- Cancer Institute of Zhongshan City People's Hospital, Zhongshan, 528403 Guangdong, China
| | - Ya-wen Zha
- Cancer Institute of Zhongshan City People's Hospital, Zhongshan, 528403 Guangdong, China
| | - Min-ying Li
- Cancer Institute of Zhongshan City People's Hospital, Zhongshan, 528403 Guangdong, China
| | - Bang-li Hu
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, 530021 Guangxi, China
| |
Collapse
|
31
|
Zhang Z, Zhao W, Li Y, Li Y, Cheng H, Zheng L, Sun X, Liu H, Shao R. YOD1 serves as a potential prognostic biomarker for pancreatic cancer. Cancer Cell Int 2022; 22:203. [PMID: 35642058 PMCID: PMC9158148 DOI: 10.1186/s12935-022-02616-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 05/12/2022] [Indexed: 11/10/2022] Open
Abstract
Background Ubiquitination is a basic post-translational modification of intracellular proteins and can be reversed enzymatically by DUBs (deubiquitinating enzymes). More than 90 DUBs have been identified. Among them, the deubiquitinating enzyme YOD1, a member of the ovarian tumor domain protease (OTUs) subfamily, is involved in the regulation of endoplasmic reticulum (ER)-related degradation pathways. In fact, it is reported that YOD1 is an important proliferation and metastasis-inducing gene, which can stimulate the characteristics of cancer stem cells and maintain circulating tumor cells (CTC). However, the expression level, prognostic effect and biological functional mechanism of YOD1 in pancreatic cancer are still unclear. Results In the GEO and TCGA databases, YOD1 mRNA expression is significantly up regulated in a variety of human pancreatic cancer tissues. Survival analysis showed that the up regulation of YOD1 can predict poor prognosis of pancreatic cancer. Cox analysis showed that high YOD1 expression is an independent prognostic factor of pancreatic cancer. ROC analysis shows that YOD1 has significant diagnostic value. The immunohistochemistry (IHC) results showed that the protein expression level of YOD1 in pancreatic cancer tissue was higher than that in neighboring non-pancreatic cancer tissues (P < 0.001). In addition, we found that YOD1 expression is negatively correlated with the infiltration level of CD8 + T cells, macrophages, neutrophils and dendritic cells (DC) in pancreatic cancer. The expression of YOD1 has a strong correlation with the different immune marker sets in PAAD. Co-expression network and functional enrichment analysis indicate that YOD1 may participate in the development of pancreatic cancer through cell adhesion molecules, p53, Hippo, TGF-β and other pathways. The experimental results of EDU, Transwell, Immunohistochemistry (IHC), Western blot and Flow Cytometry indicate that YOD1 is highly expressed in pancreatic cancer cells and pancreatic cancer tissues, and its overexpression can promote the proliferation and metastasis of pancreatic cancer cells and affect the immune microenvironment. Conclusion Our results indicate that YOD1 may be a useful biomarker for the prognosis of human pancreatic cancer, and it may also be a potential molecular target for the diagnosis and treatment of pancreatic cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02616-9.
Collapse
Affiliation(s)
- Zhishuo Zhang
- Department of Organ Transplantation and Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.,School of Pharmacy, Department of Pharmacology, China Medical University, Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, Liaoning, China
| | - Wenxia Zhao
- NHC Key Laboratory of Antibiotic Bioengineering, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Tiantan Xili, Beijing, 100050, China.,School of Pharmacy, Department of Pharmacology, China Medical University, Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, Liaoning, China
| | - Yiming Li
- NHC Key Laboratory of Antibiotic Bioengineering, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Tiantan Xili, Beijing, 100050, China.,School of Pharmacy, Department of Pharmacology, China Medical University, Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, Liaoning, China
| | - Yang Li
- NHC Key Laboratory of Antibiotic Bioengineering, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Tiantan Xili, Beijing, 100050, China
| | - Hanzeng Cheng
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050, China.,School of Pharmacy, Department of Pharmacology, China Medical University, Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, Liaoning, China
| | - Liyun Zheng
- NHC Key Laboratory of Antibiotic Bioengineering, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Tiantan Xili, Beijing, 100050, China
| | - Xiaoyu Sun
- School of Pharmacy, Department of Pharmacology, China Medical University, Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, Liaoning, China
| | - Hao Liu
- Department of Organ Transplantation and Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| | - Rongguang Shao
- NHC Key Laboratory of Antibiotic Bioengineering, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Tiantan Xili, Beijing, 100050, China.
| |
Collapse
|
32
|
Agostini A, Orlacchio A, Carbone C, Guerriero I. Understanding Tricky Cellular and Molecular Interactions in Pancreatic Tumor Microenvironment: New Food for Thought. Front Immunol 2022; 13:876291. [PMID: 35711414 PMCID: PMC9193393 DOI: 10.3389/fimmu.2022.876291] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/29/2022] [Indexed: 12/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents 90% of all pancreatic cancer cases and shows a high mortality rate among all solid tumors. PDAC is often associated with poor prognosis, due to the late diagnosis that leads to metastasis development, and limited efficacy of available treatments. The tumor microenvironment (TME) represents a reliable source of novel targets for therapy, and even if many of the biological interactions among stromal, immune, and cancer cells that populate the TME have been studied, much more needs to be clarified. The great limitation in the efficacy of current standard chemoterapy is due to both the dense fibrotic inaccessible TME barrier surrounding cancer cells and the immunological evolution from a tumor-suppressor to an immunosuppressive environment. Nevertheless, combinatorial therapies may prove more effective at overcoming resistance mechanisms and achieving tumor cell killing. To achieve this result, a deeper understanding of the pathological mechanisms driving tumor progression and immune escape is required in order to design rationale-based therapeutic strategies. This review aims to summarize the present knowledge about cellular interactions in the TME, with much attention on immunosuppressive functioning and a specific focus on extracellular matrix (ECM) contribution.
Collapse
Affiliation(s)
- Antonio Agostini
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Medical Oncology, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Arturo Orlacchio
- NYU Grossman School of Medicine, NYU Langone Health, New York, NY, United States
| | - Carmine Carbone
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ilaria Guerriero
- Biogem, Biology and Molecular Genetics Institute, Ariano Irpino, Italy
| |
Collapse
|
33
|
Jiang Y, Lin L, Lv H, Zhang H, Jiang L, Ma F, Wang Q, Ma X, Yu S. Immune cell infiltration and immunotherapy in hepatocellular carcinoma. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:7178-7200. [PMID: 35730302 DOI: 10.3934/mbe.2022339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Hepatocellular carcinoma is a highly malignant tumor and patients yield limited benefits from the existing treatments. The application of immune checkpoint inhibitors is promising but the results described in the literature are not favorable. It is therefore urgent to systematically analyze the immune microenvironment of HCC and screen the population best suited for the application of immune checkpoint inhibitors to provide a basis for clinical treatment. In this study, we collected The Cancer Genome Atlas Liver Hepatocellular Carcinoma (TCGA-LIHC)-related data sets to evaluate the immune microenvironment and immune cell infiltration (ICI) in HCC. Three independent ICI subtypes showing significant differences in survival were identified. Further, TCGA-LIHC immunophenoscore (IPS) was used to identify the differentially expressed genes between high- and low-IPS in HCC, so as to identify the immune gene subtypes in HCC tumors. The ICI score model for HCC was constructed, whereby we divided HCC samples into high- and low-score groups based on the median ICI score. The differences between these groups in genomic mutation load and immunotherapy benefit in HCC were examined in detail to provide theoretical support for accurate immunotherapy strategy in HCC. Finally, four genes were screened, which could accurately predict the subtype based on the tumor immune infiltration score. The findings may provide a basis and simplify the process for screening clinical drugs suitable for relevant subgroups.
Collapse
Affiliation(s)
- Yu Jiang
- Institute of Molecular Medicine, Medical College of Eastern Liaoning University, Dandong 118000, China
| | - Lijuan Lin
- Institute of Molecular Medicine, Medical College of Eastern Liaoning University, Dandong 118000, China
| | - Huiming Lv
- Institute of Molecular Medicine, Medical College of Eastern Liaoning University, Dandong 118000, China
| | - He Zhang
- Institute of Molecular Medicine, Medical College of Eastern Liaoning University, Dandong 118000, China
| | - Lili Jiang
- Institute of Molecular Medicine, Medical College of Eastern Liaoning University, Dandong 118000, China
| | - Fenfen Ma
- Institute of Molecular Medicine, Medical College of Eastern Liaoning University, Dandong 118000, China
| | - Qiuyue Wang
- Institute of Molecular Medicine, Medical College of Eastern Liaoning University, Dandong 118000, China
| | - Xue Ma
- Institute of Molecular Medicine, Medical College of Eastern Liaoning University, Dandong 118000, China
| | - Shengjin Yu
- Institute of Molecular Medicine, Medical College of Eastern Liaoning University, Dandong 118000, China
| |
Collapse
|
34
|
Machine Learning for Computed Tomography Radiomics: Prediction of Tumor-Infiltrating Lymphocytes in Patients With Pancreatic Ductal Adenocarcinoma. Pancreas 2022; 51:549-558. [PMID: 35877153 DOI: 10.1097/mpa.0000000000002069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES The aims of the study were to develop and validate a machine learning classifier for preoperative prediction of tumor-infiltrating lymphocytes (TILs) in patients with pancreatic ductal adenocarcinoma (PDAC). METHODS In this retrospective study of 183 PDAC patients who underwent multidetector computed tomography and surgical resection, CD4 + , CD8 + , and CD20 + expression was evaluated using immunohistochemistry, and TIL scores were calculated using the Cox regression model. The patients were divided into TIL-low and TIL-high groups. An extreme gradient boosting (XGBoost) classifier was developed using a training set consisting of 136 consecutive patients, and the model was validated in 47 consecutive patients. The discriminative ability, calibration, and clinical utility of the XGBoost classifier were evaluated. RESULTS The prediction model showed good discrimination in the training (area under the curve, 0.93; 95% confidence interval, 0.89-0.97) and validation (area under the curve, 0.79; 95% confidence interval, 0.65-0.92) sets with good calibration. The sensitivity, specificity, accuracy, positive predictive value, and negative predictive value for the training set were 0.93, 0.85, 0.90, 0.89, and 0.91, respectively, while those for the validation set were 0.63, 0.91, 0.77, 0.88, and 0.70, respectively. CONCLUSIONS The XGBoost-based model could predict PDAC TILs and may facilitate clinical decision making for immune therapy.
Collapse
|
35
|
Li A, Ye B, Lin F, Wang Y, Miao X, Jiang Y. A Novel Immunogenomic Signature to Predict Prognosis and Reveal Immune Infiltration Characteristics in Pancreatic Ductal Adenocarcinoma. PRECISION CLINICAL MEDICINE 2022; 5:pbac010. [PMID: 35694712 PMCID: PMC9172649 DOI: 10.1093/pcmedi/pbac010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 12/09/2022] Open
Abstract
Background The immune response in the tumor microenvironment (TME) plays a crucial role in cancer progression and recurrence. We aimed to develop an immune-related gene (IRG) signature to improve prognostic predictive power and reveal the immune infiltration characteristics of pancreatic ductal adenocarcinoma (PDAC). Methods The Cancer Genome Atlas (TCGA) PDAC was used to construct a prognostic model as a training cohort. The International Cancer Genome Consortium (ICGC) and the Gene Expression Omnibus (GEO) databases were set as validation datasets. Prognostic genes were screened by using univariate Cox regression. Then, a novel optimal prognostic model was developed by using least absolute shrinkage and selection operator (LASSO) Cox regression. Cell type identification by estimating the relative subsets of RNA transcripts (CIBERSORT) and estimation of stromal and immune cells in malignant tumors using expression data (ESTIMATE) algorithms were used to characterize tumor immune infiltrating patterns. The tumor immune dysfunction and exclusion (TIDE) algorithm was used to predict immunotherapy responsiveness. Results A prognostic signature based on five IRGs (MET, ERAP2, IL20RB, EREG, and SHC2) was constructed in TCGA-PDAC and comprehensively validated in ICGC and GEO cohorts. Multivariate Cox regression analysis demonstrated that this signature had an independent prognostic value. The area under the curve (AUC) values of the receiver operating characteristic (ROC) curve at 1, 3, and 5 years of survival were 0.724, 0.702, and 0.776, respectively. We further demonstrated that our signature has better prognostic performance than recently published ones and is superior to traditional clinical factors such as grade and tumor node metastasis classification (TNM) stage in predicting survival. Moreover, we found higher abundance of CD8+ T cells and lower M2-like macrophages in the low-risk group of TCGA-PDAC, and predicted a higher proportion of immunotherapeutic responders in the low-risk group. Conclusions We constructed an optimal prognostic model which had independent prognostic value and was comprehensively validated in external PDAC databases. Additionally, this five-genes signature could predict immune infiltration characteristics. Moreover, the signature helped stratify PDAC patients who might be more responsive to immunotherapy.
Collapse
Affiliation(s)
- Ang Li
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Bicheng Ye
- Medical College of Yangzhou Polytechnic College, Yangzhou, China
| | - Fangnan Lin
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Yilin Wang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Xiaye Miao
- School of Clinical Medicine, Yangzhou University, Yangzhou, China
| | - Yanfang Jiang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
36
|
Bian Y, Liu C, Li Q, Meng Y, Liu F, Zhang H, Fang X, Li J, Yu J, Feng X, Ma C, Zhao Z, Wang L, Xu J, Shao C, Lu J. Preoperative Radiomics Approach to Evaluating Tumor-Infiltrating CD8 + T Cells in Patients With Pancreatic Ductal Adenocarcinoma Using Noncontrast Magnetic Resonance Imaging. J Magn Reson Imaging 2022; 55:803-814. [PMID: 34355834 DOI: 10.1002/jmri.27871] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/20/2021] [Accepted: 07/20/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND CD8+ T cell in pancreatic ductal adenocarcinoma (PDAC) is closely related to the prognosis and treatment response of patients. Accurate preoperative CD8+ T-cell expression can better identify the population benefitting from immunotherapy. PURPOSE To develop and validate a machine learning classifier based on noncontrast magnetic resonance imaging (MRI) for the preoperative prediction of CD8+ T-cell expression in patients with PDAC. STUDY TYPE Retrospective cohort study. POPULATION Overall, 114 patients with PDAC undergoing MR scan and surgical resection; 97 and 47 patients in the training and validation cohorts. FIELD STRENGTH/SEQUENCE/3 T: Breath-hold single-shot fast-spin echo T2-weighted sequence and noncontrast T1-weighted fat-suppressed sequences. ASSESSMENT CD8+ T-cell expression was quantified using immunohistochemistry. For each patient, 2232 radiomics features were extracted from noncontrast T1- and T2-weighted images and reduced using the Wilcoxon rank-sum test and least absolute shrinkage and selection operator method. Linear discriminative analysis was used to construct radiomics and mixed models. Model performance was determined by its discriminative ability, calibration, and clinical utility. STATISTICAL TESTS Kaplan-Meier estimates, Student's t-test, the Kruskal-Wallis H test, and the chi-square test, receiver operating characteristic curve, and decision curve analysis. RESULTS A log-rank test showed that the survival duration in the CD8-high group (25.51 months) was significantly longer than that in the CD8-low group (22.92 months). The mixed model included all MRI characteristics and 13 selected radiomics features, and the area under the curve (AUC) was 0.89 (95% confidence interval [CI], 0.77-0.92) and 0.69 (95% CI, 0.53-0.82) in the training and validation cohorts. The radiomics model included 13 radiomics features, which showed good discrimination in the training cohort (AUC, 0.85; 95% CI, 0.77-0.92) and the validation cohort (AUC, 0.76; 95% CI, 0.61-0.87). DATA CONCLUSIONS This study developed a noncontrast MRI-based radiomics model that can preoperatively determine CD8+ T-cell expression in patients with PDAC and potentially immunotherapy planning. EVIDENCE LEVEL 5 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Yun Bian
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Cong Liu
- Jiangsu Key Laboratory of Big Data Analysis Technique, School of Automation, Nanjing University of Information Science and Technology, Nanjing, China
| | - Qi Li
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Yinghao Meng
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Fang Liu
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Hao Zhang
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Xu Fang
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Jing Li
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Jieyu Yu
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Xiaochen Feng
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Chao Ma
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Zengrui Zhao
- Institute for AI in Medicine, School of Automation, Nanjing University of Information Science and Technology, Nanjing, 210044, China
| | - Li Wang
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Jun Xu
- Jiangsu Key Laboratory of Big Data Analysis Technique, School of Automation, Nanjing University of Information Science and Technology, Nanjing, China
| | - Chengwei Shao
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Jianping Lu
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| |
Collapse
|
37
|
Muller M, Haghnejad V, Schaefer M, Gauchotte G, Caron B, Peyrin-Biroulet L, Bronowicki JP, Neuzillet C, Lopez A. The Immune Landscape of Human Pancreatic Ductal Carcinoma: Key Players, Clinical Implications, and Challenges. Cancers (Basel) 2022; 14:cancers14040995. [PMID: 35205742 PMCID: PMC8870260 DOI: 10.3390/cancers14040995] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 02/04/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and deadliest cancer worldwide with an overall survival rate, all stages combined, of still <10% at 5 years. The poor prognosis is attributed to challenges in early detection, a low opportunity for radical resection, limited response to chemotherapy, radiotherapy, and resistance to immune therapy. Moreover, pancreatic tumoral cells are surrounded by an abundant desmoplastic stroma, which is responsible for creating a mechanical barrier, preventing appropriate vascularization and leading to poor immune cell infiltration. Accumulated evidence suggests that PDAC is impaired with multiple “immune defects”, including a lack of high-quality effector cells (CD4, CD8 T cells, dendritic cells), barriers to effector cell infiltration due to that desmoplastic reaction, and a dominance of immune cells such as regulatory T cells, myeloid-derived suppressor cells, and M2 macrophages, resulting in an immunosuppressive tumor microenvironment (TME). Although recent studies have brought new insights into PDAC immune TME, its understanding remains not fully elucidated. Further studies are required for a better understanding of human PDAC immune TME, which might help to develop potent new therapeutic strategies by correcting these immune defects with the hope to unlock the resistance to (immune) therapy. In this review, we describe the main effector immune cells and immunosuppressive actors involved in human PDAC TME, as well as their implications as potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Marie Muller
- Department of Gastroenterology, Nancy University Hospital, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France; (V.H.); (M.S.); (B.C.); (L.P.-B.); (J.-P.B.); (A.L.)
- Correspondence:
| | - Vincent Haghnejad
- Department of Gastroenterology, Nancy University Hospital, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France; (V.H.); (M.S.); (B.C.); (L.P.-B.); (J.-P.B.); (A.L.)
| | - Marion Schaefer
- Department of Gastroenterology, Nancy University Hospital, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France; (V.H.); (M.S.); (B.C.); (L.P.-B.); (J.-P.B.); (A.L.)
| | - Guillaume Gauchotte
- Department of Pathology, Nancy University Hospital, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France;
- INSERM U1256, NGERE, Faculty of Medicine, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France
| | - Bénédicte Caron
- Department of Gastroenterology, Nancy University Hospital, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France; (V.H.); (M.S.); (B.C.); (L.P.-B.); (J.-P.B.); (A.L.)
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, Nancy University Hospital, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France; (V.H.); (M.S.); (B.C.); (L.P.-B.); (J.-P.B.); (A.L.)
- INSERM U1256, NGERE, Faculty of Medicine, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France
| | - Jean-Pierre Bronowicki
- Department of Gastroenterology, Nancy University Hospital, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France; (V.H.); (M.S.); (B.C.); (L.P.-B.); (J.-P.B.); (A.L.)
- INSERM U1256, NGERE, Faculty of Medicine, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France
| | - Cindy Neuzillet
- Medical Oncology Department, Curie Institute, Versailles Saint-Quentin University (UVQ), Paris Saclay University, 92064 Saint-Cloud, France;
| | - Anthony Lopez
- Department of Gastroenterology, Nancy University Hospital, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France; (V.H.); (M.S.); (B.C.); (L.P.-B.); (J.-P.B.); (A.L.)
| |
Collapse
|
38
|
Ma L, Cho W, Nelson ER. Our evolving understanding of how 27-hydroxycholesterol influences cancer. Biochem Pharmacol 2022; 196:114621. [PMID: 34043965 PMCID: PMC8611110 DOI: 10.1016/j.bcp.2021.114621] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 02/09/2023]
Abstract
Cholesterol has been implicated in the pathophysiology and progression of several cancers now, although the mechanisms by which it influences cancer biology are just emerging. Two likely contributing mechanisms are the ability for cholesterol to directly regulate signaling molecules within the membrane, and certain metabolites acting as signaling molecules. One such metabolite is the oxysterol 27-hydroxycholesterol (27HC), which is a primary metabolite of cholesterol synthesized by the enzyme Cytochrome P450 27A1 (CYP27A1). Physiologically, 27HC is involved in the regulation of cholesterol homeostasis and contributes to cholesterol efflux through liver X receptor (LXR) and inhibition of de novo cholesterol synthesis through the insulin-induced proteins (INSIGs). 27HC is also a selective modulator of the estrogen receptors. An increasing number of studies have identified its importance in cancer progression of various origins, especially in breast cancer. In this review, we discuss the physiological roles of 27HC targeting these two nuclear receptors and the subsequent contribution to cancer progression. We describe how 27HC promotes tumor growth directly through cancer-intrinsic factors, and indirectly through its immunomodulatory roles which lead to decreased immune surveillance and increased tumor invasion. This review underscores the importance of the cholesterol metabolic pathway in cancer progression and the potential therapeutic utility of targeting this metabolic pathway.
Collapse
Affiliation(s)
- Liqian Ma
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL
| | - Wonhwa Cho
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL
| | - Erik R. Nelson
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL,Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL,Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL,University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL,Carl R. Woese Institute for Genomic Biology, Anticancer Discovery from Pets to People Theme, University of Illinois Urbana-Champaign, Urbana, IL,To whom correspondence and reprint requests should be addressed: Erik R. Nelson. University of Illinois at Urbana-Champaign. 407 S Goodwin Ave (MC-114), Urbana, IL, 61801. Phone: 217-244-5477. Fax: 217-333-1133.
| |
Collapse
|
39
|
Immune Score Predicts Outcomes of Gastric Cancer Patients Treated with Adjuvant Chemoradiotherapy. JOURNAL OF ONCOLOGY 2022; 2021:9344124. [PMID: 34987582 PMCID: PMC8723845 DOI: 10.1155/2021/9344124] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/01/2021] [Indexed: 02/06/2023]
Abstract
Background Substantial evidence has demonstrated that tumor-infiltrating lymphocytes (TILs) are correlated with patient prognosis. The TIL-based immune score (IS) affects prognosis in various cancers, but its prognostic impact in gastric cancer (GC) patients treated with adjuvant chemoradiotherapy remains unclear. Methods A total of 101 GC patients who received chemoradiotherapy after gastrectomy were retrospectively analyzed in this study. Immunohistochemistry staining for CD3+ and CD8+ T-cell counts in both tumor center (CT) and invasive margin (IM) regions was built into the IS. Patients were then divided into three groups based on their differential IS levels. The correlation between IS and clinical parameters was analyzed. The prognostic impact of IS and clinical parameters was evaluated using Kaplan-Meier analysis and Cox proportional hazard regression analysis. Receiver operating characteristic (ROC) curves were plotted to compare the area under the curve (AUC) of IS with other clinical parameters. Nomograms for disease-free survival (DFS) and overall survival (OS) prediction were constructed based on the identified parameters. Results Finally, 20 (19.8%), 57 (56.4%), and 24 (23.8%) GC patients were identified with low, intermediate, and high IS levels, respectively. GC patients with higher IS levels exhibited better DFS (p < 0.001) and OS (p < 0.001). IS was an independent prognostic factor for both DFS (p < 0.001) and OS (p < 0.001) in multivariate analysis. IS presented a better predictive ability than the traditional pathological tumor-node-metastasis (pTNM) staging system (AUC: 0.801 vs. 0.677 and 0.800 vs. 0.660, respectively) with respect to both DFS and OS. The C-index of the nomograms for DFS and OS prediction was 0.737 and 0.774, respectively. Conclusions IS is a strong predictive factor for both DFS and OS in GC patients treated with adjuvant chemoradiotherapy, which may complement the traditional pTNM staging system.
Collapse
|
40
|
Tang P, Qu W, Wu D, Chen S, Liu M, Chen W, Ai Q, Tang H, Zhou H. Identifying and Validating an Acidosis-Related Signature Associated with Prognosis and Tumor Immune Infiltration Characteristics in Pancreatic Carcinoma. J Immunol Res 2021; 2021:3821055. [PMID: 34993253 PMCID: PMC8727107 DOI: 10.1155/2021/3821055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/29/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Acidosis in the tumor microenvironment (TME) is involved in tumor immune dysfunction and tumor progression. We attempted to develop an acidosis-related index (ARI) signature to improve the prognostic prediction of pancreatic carcinoma (PC). METHODS Differential gene expression analyses of two public datasets (GSE152345 and GSE62452) from the Gene Expression Omnibus database were performed to identify the acidosis-related genes. The Cancer Genome Atlas-pancreatic carcinoma (TCGA-PAAD) cohort in the TCGA database was set as the discovery dataset. Univariate Cox regression and the Kaplan-Meier method were applied to screen for prognostic genes. The least absolute shrinkage and selection operator (LASSO) Cox regression was used to establish the optimal model. The tumor immune infiltrating pattern was characterized by the single-sample gene set enrichment analysis (ssGSEA) method, and the prediction of immunotherapy responsiveness was conducted using the tumor immune dysfunction and exclusion (TIDE) algorithm. RESULTS We identified 133 acidosis-related genes, of which 37 were identified as prognostic genes by univariate Cox analysis in combination with the Kaplan-Meier method (p values of both methods < 0.05). An acidosis-related signature involving seven genes (ARNTL2, DKK1, CEP55, CTSV, MYEOV, DSG2, and GBP2) was developed in TCGA-PAAD and further validated in GSE62452. Patients in the acidosis-related high-risk group consistently showed poorer survival outcomes than those in the low-risk group. The 5-year AUCs (areas under the curve) for survival prediction were 0.738 for TCGA-PAAD and 0.889 for GSE62452, suggesting excellent performance. The low-risk group in TCGA-PAAD showed a higher abundance of CD8+ T cells and activated natural killer cells and was predicted to possess an elevated proportion of immunotherapeutic responders compared with the high-risk counterpart. CONCLUSIONS We developed a reliable acidosis-related signature that showed excellent performance in prognostic prediction and correlated with tumor immune infiltration, providing a new direction for prognostic evaluation and immunotherapy management in PC.
Collapse
Affiliation(s)
- Pingfei Tang
- Department of Digestive Diseases, Zhuzhou Central Hospital, The Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China
| | - Weiming Qu
- Department of Digestive Diseases, Zhuzhou Central Hospital, The Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China
| | - Dajun Wu
- Department of Digestive Diseases, Zhuzhou Central Hospital, The Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China
| | - Shihua Chen
- Department of Digestive Diseases, Zhuzhou Central Hospital, The Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China
| | - Minji Liu
- Department of Digestive Diseases, Zhuzhou Central Hospital, The Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China
| | - Weishun Chen
- Department of Digestive Diseases, Zhuzhou Central Hospital, The Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China
| | - Qiongjia Ai
- Department of Digestive Diseases, Zhuzhou Central Hospital, The Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China
| | - Haijuan Tang
- Department of Digestive Diseases, Zhuzhou Central Hospital, The Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China
| | - Hongbing Zhou
- Department of Digestive Diseases, Zhuzhou Central Hospital, The Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China
| |
Collapse
|
41
|
Zhu WJ, Hu ZF, Yuan Z. Progress in research of tumor infiltrating lymphocytes in pancreatic cancer. Shijie Huaren Xiaohua Zazhi 2021; 29:1207-1214. [DOI: 10.11569/wcjd.v29.i21.1207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The 5-year survival rate of pancreatic cancer is less than 5%, and the only available treatments, surgery, chemotherapy, and chemoradiation, have shown limited effectiveness. Therefore, alternative treatment strategies are urgently needed. In recent years, tumor infiltrating lymphocyte (TIL) therapy has shown promising successes in the treatment of some types of solid tumors because of its diverse TCR clonality, superior tumor-homing ability, and low off-target toxicity. The significant association between a high TIL density in pancreatic cancer tissue and a good clinical outcome and success of pancreatic cancer-specific TIL expansion ex vivo potentiates the rationality of the TIL therapy in pancreatic cancer. However, there are still many challenges ahead, such as neoantigen screening, rapid cell expansion, and low cytotoxicity. This article reviews the recent advances and limitations of TIL therapy in pancreatic cancer and discusses its future directions.
Collapse
Affiliation(s)
- Wen-Jun Zhu
- CAR-T (Shanghai) Cell Biotechnology Co., Ltd, Shanghai 200433, China
| | - Zhan-Fei Hu
- CAR-T (Shanghai) Cell Biotechnology Co., Ltd, Shanghai 200433, China
| | - Zhou Yuan
- The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai 200233, China
| |
Collapse
|
42
|
Wang C, Zainal NS, Chai SJ, Dickie J, Gan CP, Zulaziz N, Lye BKW, Sutavani RV, Ottensmeier CH, King EV, Abraham MT, Ismail SMB, Lau SH, Kallarakkal TG, Mun KS, Zain RB, Abdul Rahman ZA, Thomas GJ, Cheong SC, Savelyeva N, Lim KP. DNA Vaccines Targeting Novel Cancer-Associated Antigens Frequently Expressed in Head and Neck Cancer Enhance the Efficacy of Checkpoint Inhibitor. Front Immunol 2021; 12:763086. [PMID: 34733290 PMCID: PMC8559892 DOI: 10.3389/fimmu.2021.763086] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 09/27/2021] [Indexed: 12/24/2022] Open
Abstract
HPV-independent head and neck squamous cell carcinoma (HNSCC) is a common cancer globally. The overall response rate to anti-PD1 checkpoint inhibitors (CPIs) in HNSCC is ~16%. One major factor influencing the effectiveness of CPI is the level of tumor infiltrating T cells (TILs). Converting TILlow tumors to TILhigh tumors is thus critical to improve clinical outcome. Here we describe a novel DNA vaccines to facilitate the T-cell infiltration and control tumor growth. We evaluated the expression of target antigens and their respective immunogenicity in HNSCC patients. The efficacy of DNA vaccines targeting two novel antigens were evaluated with or without CPI using a syngeneic model. Most HNSCC patients (43/44) co-expressed MAGED4B and FJX1 and their respective tetramer-specific T cells were in the range of 0.06-0.12%. In a preclinical model, antigen-specific T cells were induced by DNA vaccines and increased T cell infiltration into the tumor, but not MDSC or regulatory T cells. The vaccines inhibited tumor growth and improved the outcome alone and upon combination with anti-PD1 and resulted in tumor clearance in approximately 75% of mice. Pre-existence of MAGED4B and FJX1-reactive T cells in HNSCC patients suggests that these widely expressed antigens are highly immunogenic and could be further expanded by vaccination. The DNA vaccines targeting these antigens induced robust T cell responses and with the anti-PD1 antibody conferring excellent tumor control. This opens up an opportunity for combination immunotherapy that might benefit a wider population of HNSCC patients in an antigen-specific manner.
Collapse
Affiliation(s)
- Chuan Wang
- Head and Neck Cancer Center, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Nur Syafinaz Zainal
- Cancer Immunology and Immunotherapy Unit, Cancer Research Malaysia, Selangor, Malaysia
| | - San Jiun Chai
- Cancer Immunology and Immunotherapy Unit, Cancer Research Malaysia, Selangor, Malaysia
| | - James Dickie
- Cancer Sciences, University of Southampton, Southampton, United Kingdom
| | - Chai Phei Gan
- Cancer Immunology and Immunotherapy Unit, Cancer Research Malaysia, Selangor, Malaysia
| | - Natasha Zulaziz
- Cancer Immunology and Immunotherapy Unit, Cancer Research Malaysia, Selangor, Malaysia
| | - Bryan Kit Weng Lye
- Cancer Immunology and Immunotherapy Unit, Cancer Research Malaysia, Selangor, Malaysia
| | - Ruhcha V Sutavani
- Cancer Sciences, University of Southampton, Southampton, United Kingdom
| | - Christian H Ottensmeier
- Head and Neck Cancer Center, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Emma V King
- Cancer Sciences, University of Southampton, Southampton, United Kingdom
| | - Mannil Thomas Abraham
- Ministry of Health Malaysia, Department of Oral & Maxillofacial Surgery, Tengku Ampuan Rahimah Hospital, Klang, Malaysia
| | - Siti Mazlipah Binti Ismail
- Department of Oral and Maxillofacial Clinical Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Shin Hin Lau
- Stomatology Unit, Cancer Research Centre, Institute for Medical Research, Kuala Lumpur, Malaysia
| | - Thomas George Kallarakkal
- Department of Oral and Maxillofacial Clinical Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia.,Oral Cancer Research and Coordinating Centre, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Kein Seong Mun
- Department of Pathology, Faculty of Medicine, University of Malaya, Selangor, Malaysia
| | - Rosnah Binti Zain
- Oral Cancer Research and Coordinating Centre, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia.,Faculty of Dentistry, Malaysian Allied Health Sciences Academy (MAHSA) University, Selangor, Malaysia
| | - Zainal Ariff Abdul Rahman
- Department of Oral and Maxillofacial Clinical Sciences, Faculty of Dentistry, University Teknologi Majlis Amanah Rakyat (MARA), Selangor, Malaysia
| | - Gareth J Thomas
- Cancer Sciences, University of Southampton, Southampton, United Kingdom
| | - Sok Ching Cheong
- Cancer Immunology and Immunotherapy Unit, Cancer Research Malaysia, Selangor, Malaysia
| | - Natalia Savelyeva
- Head and Neck Cancer Center, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Kue Peng Lim
- Cancer Immunology and Immunotherapy Unit, Cancer Research Malaysia, Selangor, Malaysia
| |
Collapse
|
43
|
Yang X, Guo Y, Chen C, Shao B, Zhao L, Zhou Q, Liu J, Wang G, Yuan W, Sun Z. Interaction between intestinal microbiota and tumour immunity in the tumour microenvironment. Immunology 2021; 164:476-493. [PMID: 34322877 PMCID: PMC8517597 DOI: 10.1111/imm.13397] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/18/2021] [Accepted: 07/20/2021] [Indexed: 11/27/2022] Open
Abstract
In recent years, an increasing number of studies have reported that intestinal microbiota have an important effect on tumour immunity by affecting the tumour microenvironment (TME). The intestinal microbiota are closely associated with various immune cells, such as T lymphocytes, natural killer cells (NK cells) and macrophages. Some bacteria, such as Akkermansia muciniphila (A. muciniphila) and Lactobacillus reuteri (L. reuteri), have been shown to improve the effect of tumour immunity. Furthermore, microbial imbalance, such as the increased abundance of Fusobacterium nucleatum (F. nucleatum) and Helicobacter hepaticus (H. hepaticus), generally causes tumour formation and progression. In addition, some microbiota also play important roles in tumour immunotherapy, especially PD-L1-related therapies. Therefore, what is the relationship between these processes and how do they affect each other? In this review, we summarize the interactions and corresponding mechanisms among the intestinal microbiota, immune system and TME to facilitate the research and development of new targeted drugs and provide new approaches to tumour therapy.
Collapse
Affiliation(s)
- Xiuxiu Yang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- School of MedicineZhengzhou UniversityZhengzhouChina
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Yaxin Guo
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
- Department of Basic MedicalAcademy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Academy of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Chen Chen
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
- School of Life SciencesZhengzhou UniversityZhengzhouChina
| | - Bo Shao
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Luyang Zhao
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
- Department of Basic MedicalAcademy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Academy of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Quanbo Zhou
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jinbo Liu
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Guixian Wang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Weitang Yuan
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhenqiang Sun
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| |
Collapse
|
44
|
Yin H, Pu N, Chen Q, Zhang J, Zhao G, Xu X, Wang D, Kuang T, Jin D, Lou W, Wu W. Gut-derived lipopolysaccharide remodels tumoral microenvironment and synergizes with PD-L1 checkpoint blockade via TLR4/MyD88/AKT/NF-κB pathway in pancreatic cancer. Cell Death Dis 2021; 12:1033. [PMID: 34718325 PMCID: PMC8557215 DOI: 10.1038/s41419-021-04293-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 09/05/2021] [Accepted: 09/24/2021] [Indexed: 02/07/2023]
Abstract
Lipopolysaccharide (LPS) as an important inflammatory mediator activates the innate/adaptive immune system. The existence of LPS in pancreatic ductal adenocarcinoma (PDAC) has been reported, however, its biological function in PDAC remains unclear. Here, we demonstrated that circulating and tumoral LPS was significantly increased by intestinal leakage in the orthotopic murine PDAC model, and LPS administration promoted T cell infiltration but exhaustion paradoxically in the subcutaneous murine PDAC model. By bioinformatic analysis, Toll-like receptor 4 (TLR4), LPS receptor, was further found to enrich in immune tolerance signaling in PDAC tissues. Then, a significant positive correlation was found between TLR4 and programmed death ligand-1 (PD-L1) in clinical PDAC tissues, as well as serum LPS and tumoral PD-L1. Meanwhile, LPS stimulation in vitro and in vivo obviously upregulated tumor PD-L1 expression, and effectively promoted cancer cells resistance to T cell cytotoxicity. Mechanistically, the activation of TLR4/MyD88/AKT/NF-κB cascade was found to participate in LPS mediated PD-L1 transcription via binding to its promoter regions, which was enhanced by crosstalk between NF-κB and AKT pathways. Finally, PD-L1 blockade could significantly reverse LPS-induced immune escape, and synergized with LPS treatment. Taken together, LPS can remodel tumor microenvironment, and synergize with PD-L1 blockade to suppress tumor growth, which may be a promising comprehensive strategy for PDAC.
Collapse
MESH Headings
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Aged
- Animals
- B7-H1 Antigen/genetics
- B7-H1 Antigen/metabolism
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Disease Models, Animal
- Female
- Gastrointestinal Tract/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Immune Checkpoint Inhibitors/pharmacology
- Immune Evasion/drug effects
- Immune Tolerance/drug effects
- Lipopolysaccharides
- Lymphocytes, Tumor-Infiltrating/drug effects
- Male
- Mice, Inbred BALB C
- Models, Biological
- Myeloid Differentiation Factor 88/metabolism
- NF-kappa B/metabolism
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Proto-Oncogene Proteins c-akt/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction
- Toll-Like Receptor 4/metabolism
- Transcription, Genetic/drug effects
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
- Mice
Collapse
Affiliation(s)
- Hanlin Yin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ning Pu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Qiangda Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jicheng Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Guochao Zhao
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xuefeng Xu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Dansong Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Tiantao Kuang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Dayong Jin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenhui Lou
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Wenchuan Wu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
45
|
Bian Y, Liu YF, Jiang H, Meng Y, Liu F, Cao K, Zhang H, Fang X, Li J, Yu J, Feng X, Li Q, Wang L, Lu J, Shao C. Machine learning for MRI radiomics: a study predicting tumor-infiltrating lymphocytes in patients with pancreatic ductal adenocarcinoma. Abdom Radiol (NY) 2021; 46:4800-4816. [PMID: 34189612 DOI: 10.1007/s00261-021-03159-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To develop and validate a machine learning classifier based on magnetic resonance imaging (MRI), for the preoperative prediction of tumor-infiltrating lymphocytes (TILs) in patients with pancreatic ductal adenocarcinoma (PDAC). MATERIALS AND METHODS In this retrospective study, 156 patients with PDAC underwent MR scan and surgical resection. The expression of CD4, CD8 and CD20 was detected and quantified using immunohistochemistry, and TILs score was achieved by Cox regression model. All patients were divided into TILs score-low and TILs score-high groups. The least absolute shrinkage and selection operator method and the extreme gradient boosting (XGBoost) were used to select the features and to construct a prediction model. The performance of the models was assessed using the training cohort (116 patients) and the validation cohort (40 patients), and decision curve analysis (DCA) was applied for clinical use. RESULTS The XGBoost prediction model showed good discrimination in the training (AUC 0.86; 95% CI 0.79-0.93) and validation sets (AUC 0.79; 95% CI 0.64-0.93). The sensitivity, specificity, and accuracy for the training set were 86.67%, 75.00%, and 0.81, respectively, whereas those for the validation set were 84.21%, 66.67%, and 0.75, respectively. Decision curve analysis indicated the clinical usefulness of the XGBoost classifier. CONCLUSION The model constructed by XGBoost could predict PDAC TILs and may aid clinical decision making for immune therapy.
Collapse
Affiliation(s)
- Yun Bian
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, 200434, China
| | - Yan Fang Liu
- Department of Pathology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Hui Jiang
- Department of Pathology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Yinghao Meng
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, 200434, China
| | - Fang Liu
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, 200434, China
| | - Kai Cao
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, 200434, China
| | - Hao Zhang
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, 200434, China
| | - Xu Fang
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, 200434, China
| | - Jing Li
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, 200434, China
| | - Jieyu Yu
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, 200434, China
| | - Xiaochen Feng
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, 200434, China
| | - Qi Li
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, 200434, China
| | - Li Wang
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, 200434, China
| | - Jianping Lu
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, 200434, China
| | - Chengwei Shao
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, 200434, China.
- Department of Radiology, Changhai Hospital, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
46
|
Ware MB, El-Rayes BF, Lesinski GB. Mirage or long-awaited oasis: reinvigorating T-cell responses in pancreatic cancer. J Immunother Cancer 2021; 8:jitc-2020-001100. [PMID: 32843336 PMCID: PMC7449491 DOI: 10.1136/jitc-2020-001100] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is plagued by a dismal 5-year survival rate, early onset of metastasis and limited efficacy of systemic therapies. This scenario highlights the need to fervently pursue novel therapeutic strategies to treat this disease. Recent research has uncovered complicated dynamics within the tumor microenvironment (TME) of PDAC. An abundant stroma provides a framework for interactions between cancer-associated fibroblasts, suppressive myeloid cells and regulatory lymphocytes, which together create an inhospitable environment for adaptive immune responses. This accounts for the poor infiltration and exhausted phenotypes of effector T cells within pancreatic tumors. Innovative studies in genetically engineered mouse models have established that with appropriate pharmacological modulation of suppressive elements in the TME, T cells can be prompted to regress pancreatic tumors. In light of this knowledge, innovative combinatorial strategies involving immunotherapy and targeted therapies working in concert are rapidly emerging. This review will highlight recent advances in the field related to immune suppression in PDAC, emerging preclinical data and rationale for ongoing immunotherapy clinical trials. In particular, we draw attention to foundational findings involving T-cell activity in PDAC and encourage development of novel therapeutics to improve T-cell responses in this challenging disease.
Collapse
Affiliation(s)
- Michael Brandon Ware
- Hematology and Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Bassel F El-Rayes
- Hematology and Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Gregory B Lesinski
- Hematology and Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| |
Collapse
|
47
|
Jung BK, Ko HY, Kang H, Hong J, Ahn HM, Na Y, Kim H, Kim JS, Yun CO. Relaxin-expressing oncolytic adenovirus induces remodeling of physical and immunological aspects of cold tumor to potentiate PD-1 blockade. J Immunother Cancer 2021; 8:jitc-2020-000763. [PMID: 32753544 PMCID: PMC7406118 DOI: 10.1136/jitc-2020-000763] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2020] [Indexed: 02/07/2023] Open
Abstract
Background Currently, several antibody (Ab)-based therapies have shown excellent therapeutic effects in the clinic. Nonetheless, Ab penetration into tumor tissues is limited due to abnormal vasculature, tumor interstitial pressure, and excessive extracellular matrix (ECM) accumulation, thus demanding novel strategies to overcome these barriers. Methods The intratumoral distribution of therapeutic Abs were detected by fluorescence microscopy or positron emission tomography in both human gastric xenograft and syngeneic pancreatic hamster tumor models. The antitumor efficacy by combination of oncolytic adenovirus (Ad), which coexpresses relaxin (RLX), interleukin (IL)-12, and granulocyte macrophage colony-stimulating factor (GM-CSF) (oAd/IL12/GM-RLX) and antibody against the programmed cell death protein 1 (αPD-1) was examined in hamster subcutaneous and orthotopic pancreatic tumor models. The immunological aspects of these combination therapy regimen were assessed by flow cytometry or immunohistochemistry in subcutaneous hamster tumor models. Results Relaxin-expressing oncolytic Ad effectively degraded tumor ECM and enhanced the tumor penetration of trastuzumab in comparison with trastuzumab monotherapy. Based on these results, an oAd/IL12/GM-RLX was used to enhance the potency of immune checkpoint blockade. The combination of the oAd/IL12/GM-RLX and αPD-1 promoted a concomitant degradation of the tumor ECM and amelioration of the immunosuppressive tumor niches, ultimately enhanced intratumoral infiltration of both αPD-1 and activated T cells. Of note, the combination therapy was able to elicit a potent and durable antitumor immune response against cold tumors that were refractory to immune checkpoint inhibitor monotherapy. Conclusions Our findings are the first to demonstrate that expression of four genes (IL-12p35, IL-12p40, GM-CSF, and RLX) mediated by a single oncolytic Ad vector can promote remodeling of both physical and immunological aspects of the tumor niches to overcome the major limitations of Ab-based therapies that have emerged in recent clinical trials.
Collapse
Affiliation(s)
- Bo-Kyeong Jung
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Korea (the Republic of)
| | - Hae Young Ko
- Division of RI Application, Korea Institute of Radiological and Medical Sciences, Seoul, Korea (the Republic of)
| | - Hyunji Kang
- Division of RI Application, Korea Institute of Radiological and Medical Sciences, Seoul, Korea (the Republic of)
| | - JinWoo Hong
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Korea (the Republic of).,Department of Research and Development, GeneMedicine Co., Ltd, Seoul, Korea (the Republic of)
| | - Hyo Min Ahn
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Korea (the Republic of).,Department of Research and Development, GeneMedicine Co., Ltd, Seoul, Korea (the Republic of)
| | - Youjin Na
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Korea (the Republic of)
| | - Hyeongi Kim
- Division of RI Application, Korea Institute of Radiological and Medical Sciences, Seoul, Korea (the Republic of)
| | - Jin Su Kim
- Division of RI Application, Korea Institute of Radiological and Medical Sciences, Seoul, Korea (the Republic of) .,Radiological and Medico-Oncological Sciences, University of science and technology (UST), Seoul, Korea (the Republic of)
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Korea (the Republic of) .,Department of Research and Development, GeneMedicine Co., Ltd, Seoul, Korea (the Republic of).,Institute of Nano Science and Technology (INST), Hanyang University, Seoul, Korea (the Republic of)
| |
Collapse
|
48
|
Liu SL, Bian LJ, Liu ZX, Chen QY, Sun XS, Sun R, Luo DH, Li XY, Xiao BB, Yan JJ, Lu ZJ, Yan SM, Yuan L, Tang LQ, Li JM, Mai HQ. Development and validation of the immune signature to predict distant metastasis in patients with nasopharyngeal carcinoma. J Immunother Cancer 2021; 8:jitc-2019-000205. [PMID: 32303611 PMCID: PMC7204817 DOI: 10.1136/jitc-2019-000205] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The tumor immune microenvironment has clinicopathological significance in predicting prognosis and therapeutic efficacy. We aimed to develop an immune signature to predict distant metastasis in patients with nasopharyngeal carcinoma (NPC). METHODS Using multiplexed quantitative fluorescence, we detected 17 immune biomarkers in a primary screening cohort of 54 NPC tissues presenting with/without distant metastasis following radical therapy. The LASSO (least absolute shrinkage and selection operator) logistic regression model used statistically significant survival markers in the training cohort (n=194) to build an immune signature. The prognostic and predictive accuracy of it was validated in an external independent group of 304 patients. RESULTS Eight statistically significant markers were identified in the screening cohort. The immune signature consisting of four immune markers (PD-L1+ CD163+, CXCR5, CD117) in intratumor was adopted to classify patients into high and low risk in the training cohort and it showed a high level of reproducibility between different batches of samples (r=0.988 for intratumor; p<0.0001). High-risk patients had shorter distant metastasis-free survival (HR 5.608, 95% CI 2.619 to 12.006; p<0.0001) and progression-free survival (HR 2.798, 95% CI 1.498 to 5.266; p=0·001). The C-indexes which reflected the predictive capacity in training and validation cohort were 0.703 and 0.636, respectively. Low-risk patients benefited from induction chemotherapy plus concurrent chemoradiotherapy (IC+CCRT) (HR 0.355, 95% CI 0.147 to 0.857; p=0·021), while high-risk patients did not (HR 1.329, 95% CI 0.543 to 3.253; p=0·533). To predict the individual risk of distant metastasis, nomograms with the integration of both immune signature and clinicopathological risk factors were developed. CONCLUSIONS The immune signature provided a reliable estimate of distant metastasis risk in patients with NPC and might be applied to identify the cohort which benefit from IC+CCRT.
Collapse
Affiliation(s)
- Sai-Lan Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong Province, People's Republic of China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Li-Juan Bian
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong Province, People's Republic of China
| | - Ze-Xian Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong Province, People's Republic of China
| | - Qiu-Yan Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong Province, People's Republic of China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Xue-Song Sun
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong Province, People's Republic of China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Rui Sun
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong Province, People's Republic of China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Dong-Hua Luo
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong Province, People's Republic of China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Xiao-Yun Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong Province, People's Republic of China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Bei-Bei Xiao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong Province, People's Republic of China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Jin-Jie Yan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong Province, People's Republic of China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Zi-Jian Lu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong Province, People's Republic of China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Shu-Mei Yan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Li Yuan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong Province, People's Republic of China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Lin-Quan Tang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong Province, People's Republic of China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Jian-Ming Li
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong Province, People's Republic of China
| | - Hai-Qiang Mai
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong Province, People's Republic of China .,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| |
Collapse
|
49
|
Mao M, Ling H, Lin Y, Chen Y, Xu B, Zheng R. Construction and Validation of an Immune-Based Prognostic Model for Pancreatic Adenocarcinoma Based on Public Databases. Front Genet 2021; 12:702102. [PMID: 34335699 PMCID: PMC8318842 DOI: 10.3389/fgene.2021.702102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/21/2021] [Indexed: 12/19/2022] Open
Abstract
Background Pancreatic adenocarcinoma (PAAD) is a highly lethal and aggressive tumor with poor prognoses. The predictive capability of immune-related genes (IRGs) in PAAD has yet to be explored. We aimed to explore prognostic-related immune genes and develop a prediction model for indicating prognosis in PAAD. Methods The messenger (m)RNA expression profiles acquired from public databases were comprehensively integrated and differentially expressed genes were identified. Univariate analysis was utilized to identify IRGs that related to overall survival. Whereafter, a multigene signature in the Cancer Genome Atlas cohort was established based on the least absolute shrinkage and selection operator (LASSO) Cox regression analysis. Moreover, a transcription factors regulatory network was constructed to reveal potential molecular processes in PAAD. PAAD datasets downloaded from the Gene Expression Omnibus database were applied for the validations. Finally, correlation analysis between the prognostic model and immunocyte infiltration was investigated. Results Totally, 446 differentially expressed immune-related genes were screened in PAAD tissues and normal tissues, of which 43 IRGs were significantly related to the overall survival of PAAD patients. An immune-based prognostic model was developed, which contained eight IRGs. Univariate and multivariate Cox regression revealed that the risk score model was an independent prognostic indicator in PAAD (HR > 1, P < 0.001). Besides, the sensitivity of the model was evaluated by the receiver operating characteristic curve analysis. Finally, immunocyte infiltration analysis revealed that the eight-gene signature possibly played a pivotal role in the status of the PAAD immune microenvironment. Conclusion A novel prognostic model based on immune genes may serve to characterize the immune microenvironment and provide a basis for PAAD immunotherapy.
Collapse
Affiliation(s)
- Miaobin Mao
- The Graduate School, Fujian Medical University, Fuzhou, China.,Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, China.,Union Clinical Medicine College, Fujian Medical University, Fuzhou, China
| | - Hongjian Ling
- The Graduate School, Fujian Medical University, Fuzhou, China.,Union Clinical Medicine College, Fujian Medical University, Fuzhou, China
| | - Yuping Lin
- The Graduate School, Fujian Medical University, Fuzhou, China.,Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, China.,Union Clinical Medicine College, Fujian Medical University, Fuzhou, China
| | - Yanling Chen
- Department of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Benhua Xu
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, China.,Union Clinical Medicine College, Fujian Medical University, Fuzhou, China.,College of Medical Technology and Engineering, Fujian Medical University, Fuzhou, China.,School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| | - Rong Zheng
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, China.,Union Clinical Medicine College, Fujian Medical University, Fuzhou, China.,College of Medical Technology and Engineering, Fujian Medical University, Fuzhou, China.,School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| |
Collapse
|
50
|
Ding G, Guo M, Yang Y, Sun C, Wu S, Liu X, Wang J, Jiang H, Liu Y, Zheng J. Large-Section Histopathology Can Better Indicate the Immune Microenvironment and Predict the Prognosis of Pancreatic Ductal Adenocarcinoma Than Small-Section Histopathology. Front Oncol 2021; 11:694933. [PMID: 34367978 PMCID: PMC8340684 DOI: 10.3389/fonc.2021.694933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/28/2021] [Indexed: 11/25/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor and is insensitive to radiotherapy and chemotherapy, as it is highly correlated with its complex tumor microenvironment (TME). A comprehensive description of PDAC's immune microenvironment at the pathological level has not been reported, thus limiting its treatment. Previous studies have shown that large-section histopathology (LSH) can reveal the complete structure and margin of the tumor on a single slice and effectively reflect intratumoral heterogeneity. LSH, as opposed to classic small-section histopathology (SSH), can also be used to explore the infiltration state of immune cells in different regions. In the current study, EnVision immunohistochemical staining was used to explore the panoramic distribution of CD4-, CD8-, CD15-, CD20-, and CD56 (surface markers of helper T cells, cytotoxic T cells, neutrophils, B cells, and NK cells, respectively)-positive cells in 102 pairs of paraffin wax-embedded PDAC samples (LSH vs SSH) for the first time. These indicators were then analyzed, and correlations of clinicopathological characteristics with clinical prognoses were analyzed. The findings of this study show that LSH can effectively indicate more immune cells than SSH. Upregulated CD4, CD8, CD20, and CD56 or downregulated CD15 was correlated with a good prognosis in PDAC patients. However, analysis of SSH showed that only upregulated CD4 and CD8 can be used as indicators of a good prognosis. Multivariate Cox regression analysis showed that 7 variables, namely, pTNM stage (P=0.002), PDL1 expression (P=0.001), CDX2 expression (P=0.008), DPC4 expression (P=0.004), CD4 expression in LSH (P<0.001), CD8 expression in LSH (P=0.010) and CD15 expression in LSH (P=0.031), were significantly correlated with the prognosis of PDAC patients. The findings of this study indicate that LSH is an effective tool for a panoramic assessment of the immune microenvironment in pancreatic cancer patients.
Collapse
Affiliation(s)
- Guiling Ding
- Department of Pathology, Shanghai General Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
- Department of Pathology, Changhai Hospital Affiliated to Navy Medical University (Second Military Medical University), Shanghai, China
| | - Meng Guo
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yelin Yang
- Department of Pathology, Changhai Hospital Affiliated to Navy Medical University (Second Military Medical University), Shanghai, China
| | - Chen Sun
- Department of Pathology, Changhai Hospital Affiliated to Navy Medical University (Second Military Medical University), Shanghai, China
| | - Shengyong Wu
- Department of Health Statistics, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xingchen Liu
- Department of Pathology, Changhai Hospital Affiliated to Navy Medical University (Second Military Medical University), Shanghai, China
| | - Jin Wang
- Department of Pathology, Changhai Hospital Affiliated to Navy Medical University (Second Military Medical University), Shanghai, China
| | - Hui Jiang
- Department of Pathology, Changhai Hospital Affiliated to Navy Medical University (Second Military Medical University), Shanghai, China
| | - Yanfang Liu
- Department of Pathology, Changhai Hospital Affiliated to Navy Medical University (Second Military Medical University), Shanghai, China
| | - Jianming Zheng
- Department of Pathology, Changhai Hospital Affiliated to Navy Medical University (Second Military Medical University), Shanghai, China
| |
Collapse
|