1
|
Zhang J, Kong X, Zhou B, Li R, Yu Z, Zhu J, Xi Q, Li Y, Zhao Z, Zhang R. Metabolic reprogramming of drug resistance in pancreatic cancer: mechanisms and effects. Mol Aspects Med 2025; 103:101368. [PMID: 40398192 DOI: 10.1016/j.mam.2025.101368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 05/10/2025] [Accepted: 05/10/2025] [Indexed: 05/23/2025]
Abstract
Pancreatic cancer is a highly aggressive gastrointestinal malignancy, often termed the "king of cancers" due to its notoriously high mortality rate. Its clinical characteristics, including late diagnosis, low surgical resectability, high recurrence rates, significant chemoresistance, and poor prognosis have collectively driven the persistent rise in incidence and mortality. Despite ongoing advancements in therapeutic strategies, the management of pancreatic cancer, particularly at advanced stages, remains challenging. Chemotherapy remains the mainstay of current treatment. However, the prevalent problem of chemotherapy resistance poses a significant obstacle to effective treatment. Metabolic reprogramming, characterized by alterations in glucose metabolism, lipid biosynthesis, and amino acid utilization, supports the high energy demands and rapid proliferation of cancer cells. Emerging evidence suggests that these metabolic changes, possibly mediated by epigenetic mechanisms, also contribute to tumorigenesis and metastasis. These findings highlight the critical role of metabolic alterations in pancreatic cancer pathogenesis. This review explores the relationship between metabolic reprogramming and chemotherapy resistance, discussing underlying mechanisms and summarizing preclinical studies and drug development targeting metabolism. The aim is to provide a comprehensive perspective on potential therapeutic strategies for pancreatic cancer.
Collapse
Affiliation(s)
- Jinyi Zhang
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Xueqing Kong
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Boyan Zhou
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Rui Li
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhaoan Yu
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Jinrong Zhu
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Qing Xi
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yan Li
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Zichao Zhao
- Department of Emergency Medicine, Shaodong People's Hospital, Shaodong City, Hunan Province, China.
| | - Rongxin Zhang
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
2
|
Liu N, Wu T, Han G, Chen M. Exosome-mediated ferroptosis in the tumor microenvironment: from molecular mechanisms to clinical application. Cell Death Discov 2025; 11:221. [PMID: 40328736 PMCID: PMC12056189 DOI: 10.1038/s41420-025-02484-y] [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: 02/15/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Ferroptosis in the tumor microenvironment (TME) plays a crucial role in the development, metastasis, immune escape, and drug resistance of various types of cancer. A better understanding of ferroptosis in the TME could illuminate novel aspects of this process and promote the development of targeted therapies. Compelling evidence indicates that exosomes are key mediators in regulating the TME. In this respect, it is now understood that exosomes can deliver biologically functional molecules to recipient cells, influencing cancer progression by reprogramming the metabolism of cancer cells and their surrounding stromal cells through ferroptosis. In this review, we focus on the role of exosomes in the TME and describe how they contribute to tumor reprogramming, immunosuppression, and the formation of pre-metastatic niches through ferroptosis. In addition, we highlight exosome-mediated ferroptosis as a potential target for cancer therapy and discuss strategies employing exosomes in ferroptosis treatment. Finally, we outline the current applications and challenges of targeted exosome-mediated ferroptosis therapy in tumor immunotherapy and chemotherapy. Our aim is to advance research on the link between exosomes and ferroptosis in the TME, and we pose questions to guide future studies in this area.
Collapse
Affiliation(s)
- Na Liu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Tianqing Wu
- XJTLU Wisdom Lake Academy of Pharmacy, Suzhou, Jiangsu Province, China
| | - Guohu Han
- Department of Oncology, Jingjiang People's Hospital Affiliated with Yangzhou University, Jingjiang, China
| | - Minbin Chen
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| |
Collapse
|
3
|
Jin Y, Hu H, Tian Y, Xu H, Yu Q, Cheng L, Guo X, Wang Z, Huang X, Wang X, Wang G. The role of LncRNA-MANCR induced by HIF-1α drive the malignant progression of pancreatic cancer by targeting miRNA-494/SIRT1 signaling axis under hypoxic conditions. Cancer Gene Ther 2025:10.1038/s41417-025-00900-0. [PMID: 40195439 DOI: 10.1038/s41417-025-00900-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 03/08/2025] [Accepted: 03/25/2025] [Indexed: 04/09/2025]
Abstract
This study revealed the prospective biological role and fundamental mechanisms of hypoxia-induced lncRNA-MANCR (MANCR), which is notably upregulated in pancreatic cancer (PC). This work uncovered the potential biological function and underlying mechanisms of hypoxia-induced MANCR, which is significantly elevated in PC. Microarray assays confirmed MANCR expression in the tissues of patients with PC and patients with chronic pancreatitis (CP), which positively correlated with sirtuin-1 (SIRT1) mRNA levels. Chromatin immunoprecipitation and luciferase assays were employed to gauge binding within the hypoxia-inducible factor-1α (HIF-1α)/MANCR/miRNA-494/SIRT1 pathway. Additionally, the association between MANCR expression and the clinical outcomes of patients with PC was confirmed. MANCR is significantly upregulated in PC cells under hypoxic conditions, which is closely linked to poor prognosis in patients with PC. Depletion of MANCR repressed in vitro proliferation, migration, and invasion of PC cells and in vivo growth of PC xenograft tumours. We further demonstrated that MANCR is localised in the cytoplasm and competitively binds miR-494, which directly targets SIRT1. Mechanically, the overexpression of SIRT1 improved the stability of the HIF-1α protein through deacetylation, leading to enhanced HIF-1α assembly. Moreover, MANCR underwent transcriptional regulation by HIF-1α in a hypoxic setting. This modulation was ascribed to HIF-1α binding to hypoxia response elements present in the MANCR promoter sequence. Data revealed the potential possibility of feedback between MANCR and HIF-1α, which may be conducive to hypoxia-induced oncogenicity and PC tumorigenesis, thereby providing a suitable therapeutic target.
Collapse
Affiliation(s)
- Yan Jin
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
- Oncological and Laparoscopic Surgery Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hao Hu
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Yitong Tian
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Han Xu
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Qiao Yu
- Ultrasound medicine department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Long Cheng
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Oncological and Laparoscopic Surgery Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoyu Guo
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Oncological and Laparoscopic Surgery Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zongwei Wang
- School of Medicine, Stanford University, San Francisco, CA, USA
| | - Xiaoxu Huang
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Xiaoming Wang
- Department of Hepatobiliary Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Gang Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
- Oncological and Laparoscopic Surgery Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
4
|
Sun RQ, Ye YH, Xu Y, Wang B, Pan SY, Li N, Chen L, Pan JY, Hu ZQ, Fan J, Zhou ZJ, Zhou J, Song CL, Zhou SL. Integrated molecular characterization of sarcomatoid hepatocellular carcinoma. Clin Mol Hepatol 2025; 31:426-444. [PMID: 39657751 PMCID: PMC12016616 DOI: 10.3350/cmh.2024.0686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/15/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUNDS/AIMS Sarcomatoid hepatocellular carcinoma (HCC) is a rare histological subtype of HCC characterized by extremely poor prognosis; however, its molecular characterization has not been elucidated. METHODS In this study, we conducted an integrated multiomics study of whole-exome sequencing, RNA-seq, spatial transcriptome, and immunohistochemical analyses of 28 paired sarcomatoid tumor components and conventional HCC components from 10 patients with sarcomatoid HCC, in order to identify frequently altered genes, infer the tumor subclonal architectures, track the genomic evolution, and delineate the transcriptional characteristics of sarcomatoid HCCs. RESULTS Our results showed that the sarcomatoid HCCs had poor prognosis. The sarcomatoid tumor components and the conventional HCC components were derived from common ancestors, mostly accessing similar mutational processes. Clonal phylogenies demonstrated branched tumor evolution during sarcomatoid HCC development and progression. TP53 mutation commonly occurred at tumor initiation, whereas ARID2 mutation often occurred later. Transcriptome analyses revealed the epithelial-mesenchymal transition (EMT) and hypoxic phenotype in sarcomatoid tumor components, which were confirmed by immunohistochemical staining. Moreover, we identified ARID2 mutations in 70% (7/10) of patients with sarcomatoid HCC but only 1-5% of patients with non-sarcomatoid HCC. Biofunctional investigations revealed that inactivating mutation of ARID2 contributes to HCC growth and metastasis and induces EMT in a hypoxic microenvironment. CONCLUSION We offer a comprehensive description of the molecular basis for sarcomatoid HCC, and identify genomic alteration (ARID2 mutation) together with the tumor microenvironment (hypoxic microenvironment), that may contribute to the formation of the sarcomatoid tumor component through EMT, leading to sarcomatoid HCC development and progression.
Collapse
Affiliation(s)
- Rong-Qi Sun
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu-Hang Ye
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ye Xu
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bo Wang
- Department of General Surgery, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Si-Yuan Pan
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ning Li
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Long Chen
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing-Yue Pan
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhi-Qiang Hu
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zheng-Jun Zhou
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng-Li Song
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Shao-Lai Zhou
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Sun RQ, Zhou SL. Correspondence to editorial on "Integrated molecular characterization of sarcomatoid hepatocellular carcinoma". Clin Mol Hepatol 2025; 31:e192-e193. [PMID: 40007141 PMCID: PMC12016595 DOI: 10.3350/cmh.2025.0183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 02/22/2025] [Indexed: 02/27/2025] Open
Affiliation(s)
- Rong-Qi Sun
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shao-Lai Zhou
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Cao Z, Li J, Hu W, Xu J, Zhao F, Wang Y, Qin S, Liu M, Wang P, Duan J, Zhou W, Ding Z, Tang S, Ma X, Wang L. Near-Infrared Imaging Agent ABSi-148 Alleviates CA IX-Mediated Hypoxic Fibrosis in Inflammation-Cancer Transition. Adv Healthc Mater 2025; 14:e2404935. [PMID: 40099420 DOI: 10.1002/adhm.202404935] [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: 12/09/2024] [Revised: 03/07/2025] [Indexed: 03/19/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a formidable challenge due to its late diagnosis and intrinsic treatment resistance, exacerbates by its development from chronic inflammation to cancer transition (ICT). Here, this investigation aims to develop and evaluate ABSi-148, a novel near-infrared (NIR) agent targeting hypoxic carbonic anhydrase IX (CA IX), for its potential applications in ICT imaging and even PDAC treatment. ABSi-148 is synthesized from 4-(2-Aminoethyl) benzene sulfonamide (ABS), a sulfonamide derivative, conjugating with MHI-148 dye with merits of exceptional NIR-emitting traits, high biocompatibility, and deep tissue penetration imaging capability. It selectively accumulates in CoCl2-induced pancreatic stellate cells and pancreatic cancer cells via binding with transmembrane CA IX in vitro. Meanwhile, ABSi-148 effectively visualizes the early pancreatic lesion, and its long-term administration inhibits the progression of hypoxia-related fibrosis involved in pancreatic intraepithelial neoplasias (PanINs), and even PDAC progression in vivo. Besides, ABSi-148 monitors treatment efficacy and localizes hypoxic tumor regions, enhancing survival in tamoxifen combined with caerulein-induced KPC mice. Overall, ABSi-148 emerges as a theranostic NIR agent for precise diagnosis and targeted therapy in ICT of PDAC, promising to alleviate tumor progression and enhancing outcomes.
Collapse
Affiliation(s)
- Zhi Cao
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jingmin Li
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
- Internal medicine department, Guangzhou women and children's Medical Center, Guangzhou, 510623, China
| | - Weibin Hu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
- Department of Nuclear Medicine, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 518037, China
| | - Jian Xu
- Stroke center, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510799, China
| | - Fengyun Zhao
- Cancer Research Institute of Zhongshan City, Zhongshan City People's Hospital, Zhongshan, 528403, China
| | - Yishu Wang
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - Shuanglin Qin
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha, 410218, China
| | - Ming Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Ping Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510230, China
| | - Jingwei Duan
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - Wensheng Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Zhaowei Ding
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510230, China
| | - Shaohui Tang
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Xiaodong Ma
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - Lisheng Wang
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| |
Collapse
|
7
|
Feng H, Chen Z, Li J, Feng J, Yang F, Meng F, Yin H, Guo Y, Xu H, Liu Y, Liu R, Lou W, Liu L, Han X, Su H, Zhang L. Unveiling circulating targets in pancreatic cancer: Insights from proteogenomic evidence and clinical cohorts. iScience 2025; 28:111693. [PMID: 40060891 PMCID: PMC11889678 DOI: 10.1016/j.isci.2024.111693] [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: 03/02/2024] [Revised: 09/23/2024] [Accepted: 12/23/2024] [Indexed: 03/04/2025] Open
Abstract
Pancreatic cancer (PC), characterized by the absence of effective biomarkers and therapies, remains highly fatal. Data regarding the correlations between PC risk and individual plasma proteome known for minimally invasive biomarkers are scarce. Here, we analyzed 1,345 human plasma proteins using proteome-wide association studies, identifying 78 proteins significantly associated with PC risk. Of these, four proteins (ROR1, FN1, APOA5, and ABO) showed the most substantial causal link to PC, confirmed through Mendelian randomization and colocalization analyses. Data from two clinical cohorts further demonstrated that FN1 and ABO were notably overexpressed in both blood and tumor samples from PC patients, compared to healthy controls or para-tumor tissues. Additionally, elevated FN1 and ABO levels correlated with shorter median survival in patients. Multiple drugs targeting FN1 or ROR1 are available or in clinical trials. These findings suggest that plasma protein FN1 associated with PC holds potential as both prognostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Haokang Feng
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhixue Chen
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jianang Li
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jiale Feng
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Fei Yang
- Innovative Institute of Tumor Immunity and Medicine (ITIM), Hefei, Anhui, China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China
| | - Fansheng Meng
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hanlin Yin
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuquan Guo
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Huaxiang Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuxin Liu
- Department of Basic Medicine and Institute of Liver Diseases, Shan Xi Medical University, Taiyuan 030000, China
| | - Runjie Liu
- Department of Basic Medicine and Institute of Liver Diseases, Shan Xi Medical University, Taiyuan 030000, China
| | - Wenhui Lou
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- The Shanghai Geriatrics Medical Center, Zhongshan Hospital MinHang MeiLong Branch, Fudan University, Shanghai 201104, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xu Han
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hua Su
- Institutes of Biomedical Sciences, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lei Zhang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
8
|
Pérez-Moreno P, Muñoz JP, Retamal MA. Molecular Interplay Between Non-Coding RNAs and Connexins and Its Possible Role in Cancer. Int J Mol Sci 2025; 26:2538. [PMID: 40141179 PMCID: PMC11942031 DOI: 10.3390/ijms26062538] [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: 02/07/2025] [Revised: 03/07/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
Non-coding RNAs (ncRNAs) are sequences that do not encode for proteins and play key roles in different cellular processes, including cell proliferation and differentiation. On the other hand, connexins (Cxs) are transmembrane proteins that principally allow intercellular communication. In pathological conditions such as cancer, there is a deregulation in the expression and/or function of ncRNAs and Cxs, which in turn leads to an enhancement in the aggressive phenotype, such as a greater proliferative and invasive capacity. This suggests a plausible interplay between ncRNAs and Cxs. Based on that, this review aims to summarize the current knowledge regarding this relationship and to analyze how it may influence the development of aggressive traits in cancer cells and the clinicopathological features of cancer patients. Finally, we discuss the potential of ncRNAs and Cxs as promising clinical biomarkers for cancer diagnosis, prognosis, and therapeutic targeting.
Collapse
Affiliation(s)
- Pablo Pérez-Moreno
- Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7780272, Chile
| | - Juan P. Muñoz
- Laboratorio de Bioquímica, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile;
| | - Mauricio A. Retamal
- Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7780272, Chile
| |
Collapse
|
9
|
Wu J, Cheng Y, Qian K, Yang P, Zhou L, Xu M, Sheng D, Wang T, Li Y, Yang X, Wei Y, Zhang Q. siRNA-Encapsulated Biomimetic Liposomes Effectively Inhibit Tumor Cells' Hexosamine Biosynthesis Pathway for Attenuating Hyaluronan Barriers to Pancreatic Cancer Chemotherapy. ACS NANO 2025; 19:7928-7947. [PMID: 39978787 DOI: 10.1021/acsnano.4c14969] [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: 02/22/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses significant therapeutic challenges due to excessive hyaluronic acid (HA) accumulation, which impedes drug delivery. Here, we present a targeted approach to reduce HA production by specifically silencing glutamine-fructose-6-phosphate aminotransferase 1 (GFAT1), a key enzyme of the hexosamine biosynthesis pathway (HBP) in pancreatic cancer cells. An engineered liposomal system for siGFAT1 delivery, PMLip@siGFAT1, characterized by macrophage membrane camouflage, LFC131 peptide-mediated targeting, and calcium phosphate (CaP) as the core, was designed to ensure prolonged circulation, enhanced inflamed vascular endothelial penetration, and subsequent effective tumor cell uptake and endosomal escape. Consequently, PMLip@siGFAT1 markedly downregulated the HA level in the PDAC microenvironment, decompressing the tumor vasculature and weakening the stromal barrier, which in turn improved the permeability of chemotherapeutics. In combination with Doxil, PMLip@siGFAT1 demonstrated potent antitumor efficacy with minimal systemic toxicity. Importantly, unlike PEGPH20 (hyaluronidase), PMLip@siGFAT1 reduced tumor invasiveness, while preserving skeletal muscle integrity. These findings highlight that PMLip@siGFAT1 holds great potential to revitalize HA downregulation strategies in pancreatic cancer for enhanced drug delivery and efficacy.
Collapse
Affiliation(s)
- Jing Wu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yunlong Cheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
- Institute of Traditional Chinese Medicine, & Key Laboratory of TCM Drug Delivery, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi 710003, China
| | - Kang Qian
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Peng Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Lingling Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Dongyu Sheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Tianying Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yixian Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xiyu Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Qizhi Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
10
|
Leck LYW, Abd El-Aziz YS, McKelvey KJ, Park KC, Sahni S, Lane DJR, Skoda J, Jansson PJ. Cancer stem cells: Masters of all traits. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167549. [PMID: 39454969 DOI: 10.1016/j.bbadis.2024.167549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 10/01/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Cancer is a heterogeneous disease, which contributes to its rapid progression and therapeutic failure. Besides interpatient tumor heterogeneity, tumors within a single patient can present with a heterogeneous mix of genetically and phenotypically distinct subclones. These unique subclones can significantly impact the traits of cancer. With the plasticity that intratumoral heterogeneity provides, cancers can easily adapt to changes in their microenvironment and therapeutic exposure. Indeed, tumor cells dynamically shift between a more differentiated, rapidly proliferating state with limited tumorigenic potential and a cancer stem cell (CSC)-like state that resembles undifferentiated cellular precursors and is associated with high tumorigenicity. In this context, CSCs are functionally located at the apex of the tumor hierarchy, contributing to the initiation, maintenance, and progression of tumors, as they also represent the subpopulation of tumor cells most resistant to conventional anti-cancer therapies. Although the CSC model is well established, it is constantly evolving and being reshaped by advancing knowledge on the roles of CSCs in different cancer types. Here, we review the current evidence of how CSCs play a pivotal role in providing the many traits of aggressive tumors while simultaneously evading immunosurveillance and anti-cancer therapy in several cancer types. We discuss the key traits and characteristics of CSCs to provide updated insights into CSC biology and highlight its implications for therapeutic development and improved treatment of aggressive cancers.
Collapse
Affiliation(s)
- Lionel Y W Leck
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Yomna S Abd El-Aziz
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Oral Pathology Department, Faculty of Dentistry, Tanta University, Tanta, Egypt
| | - Kelly J McKelvey
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Kyung Chan Park
- Proteina Co., Ltd./Seoul National University, Seoul, South Korea
| | - Sumit Sahni
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Darius J R Lane
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
| | - Patric J Jansson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
11
|
Chen H, Wang X, Liu S, Tang Z, Xie F, Yin J, Sun P, Wang H. Circular RNA in Pancreatic Cancer: Biogenesis, Mechanism, Function and Clinical Application. Int J Med Sci 2025; 22:1612-1629. [PMID: 40093798 PMCID: PMC11905278 DOI: 10.7150/ijms.107773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Circular RNAs (circRNAs) are a class of novel RNA molecules featured by single-strand covalently closed circular structure, which not only are extensively found in eukaryotes and are highly conserved, but also conduct paramount roles in the occurrence and progression of pancreatic cancer (PC) through diverse mechanisms. As recent studies have demonstrated, circRNAs typically exhibit tissue-specific and cell specific expression patterns, with strong potential as biomarkers for disease diagnosis and prognosis. On the basis of their localization and specific interactions with DNA, RNA, and proteins, circRNAs are considered to possess specific biological functions by acting as microRNA (miRNA) sponges, RNA binding protein (RBP) sponges, transcriptional regulators, molecular scaffolds and translation templates. On that account, further addressing the technical difficulties in the detection and research of circRNAs and filling gaps in their biological knowledge will definitely push ahead this comparatively young research field and bring circRNAs to the forefront of clinical practice. Thus, this review systematically summarizes the biogenesis, function, molecular mechanisms, biomarkers and therapeutic targets of circRNAs in PC.
Collapse
Affiliation(s)
- Hang Chen
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China
| | - Xianxing Wang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China
| | - Shan Liu
- Department of Anesthesiology, Chongqing Seventh People's Hospital, Chongqing University of Technology, Chongqing, 400054, China
| | - Ziwei Tang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China
- Chongqing Medical University, Chongqing, 400016, China
| | - Fuming Xie
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China
| | - Jingyang Yin
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China
| | - Pijiang Sun
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China
| | - Huaizhi Wang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China
| |
Collapse
|
12
|
Abreu MM, Chocron AF, Smadja DM. From cold to hot: mechanisms of hyperthermia in modulating tumor immunology for enhanced immunotherapy. Front Immunol 2025; 16:1487296. [PMID: 40092992 PMCID: PMC11906415 DOI: 10.3389/fimmu.2025.1487296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
The emergence of immunotherapies has revolutionized cancer treatment by leveraging the immune system to target malignancies, offering new hope where traditional therapies often fall short. Within this context, hyperthermia (HT) has re-emerged as a promising adjunctive treatment, capable of enhancing the effectiveness of radiotherapy, chemotherapy, and immunotherapy. HT influences both the innate and adaptive immune systems, enhancing the activity of immune cells such as neutrophils, NK cells, and dendritic cells, while also modulating the tumor microenvironment (TME) to promote immunogenic cell death (ICD) and reduce immunosuppressive conditions. These effects contribute to the transformation of immunologically "cold" tumors into "hot" tumors, making them more susceptible to immune-mediated destruction. Furthermore, HT can amplify the efficacy of immune checkpoint inhibitors (ICIs) by improving immune cell infiltration, inducing damage-associated molecular pattern (DAMP) release, and enhancing antigen presentation. Preclinical and clinical studies support the combination of HT with ICIs, demonstrating improved outcomes in otherwise resistant tumors. However, the full therapeutic potential of the different technologies allowing to apply HT remains to be fully understood, and further research is needed to optimize treatment protocols, explore the differential impacts of local versus whole-body hyperthermia, and identify biomarkers for patient stratification. This review underscores the multifaceted role of HT in immunity and its potential to significantly enhance the efficacy of immunotherapy.
Collapse
Affiliation(s)
- M. Marc Abreu
- Medicine Department, BTT Medical Institute, Aventura, FL, United States
- BTT Engineering Department, BTT Medical Institute, Aventura, FL, United States
| | - Alberto F. Chocron
- Medicine Department, BTT Medical Institute, Aventura, FL, United States
- Research Service, Miami Veteran Administration Medical Center, Miami, FL, United States
| | - David M. Smadja
- Department of Hematology, AP-HP, Georges Pompidou European Hospital, Paris, France
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
13
|
Xu S, Xie X, He P, Zhu S, Li X, Chen Q, Ma X, Liang X. Nitric Oxide-Producing Multiple Functional Nanoparticle Remodeling Tumor Microenvironment for Synergistic Photodynamic Immunotherapy against Hypoxic Tumor. ACS NANO 2025; 19:6371-6387. [PMID: 39913864 DOI: 10.1021/acsnano.4c16329] [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: 02/19/2025]
Abstract
The treatment of pancreatic cancer faces significant challenges due to connective tissue hyperplasia and severe hypoxia. Unlike oxygen-dependent Type II photosensitizers, Type I photosensitizers can produce a substantial amount of reactive oxygen species, even under hypoxic conditions, making them more suitable for photodynamic therapy of pancreatic cancer. However, the dense extracellular matrix of pancreatic cancer limits the penetration efficiency of photosensitizers, and the presence of immunosuppressive cells in the tumor microenvironment reduces the therapeutic effect. To address these challenges, we designed the photoimmunotherapeutic M1@PAP nanoparticles composed of Type I photosensitizer and anti-PD-L1 siRNA (siPD-L1), which was encapsulated into M1 macrophage membrane vesicles. In this system, pyropheophorbide-a (PPA) was covalently conjugated to poly-l-arginine (Arg9). Notably, it was capable of generating sufficient superoxide anions under hypoxic conditions, thereby functioning as a Type I photosensitizer. Furthermore, Arg9 acted as a nitric oxide (NO) donor, enhancing the penetration efficiency of the nanophotosensitizer by inhibiting cancer-associated fibroblast (CAF) activation and decomposing the tumor extracellular matrix. Additionally, M1 macrophage membrane vesicles provided active targeting capabilities and reeducated immunosuppressed M2 macrophages. The reversal of immunosuppressive microenvironment further promoted the efficacy of anti-PD-L1 siRNA immunotherapy, showing great potential in synergistic photodynamic immunotherapy against hypoxic pancreatic tumor.
Collapse
Affiliation(s)
- Shuyu Xu
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Xinxin Xie
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Ping He
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Shiwei Zhu
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Xiaoda Li
- Peking University Health Science Center, Beijing 100191, China
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore
| | - Xiaotu Ma
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Xiaolong Liang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
14
|
Kanwal R, Esposito JE, Jawed B, Zakir SK, Pulcini R, Martinotti R, Botteghi M, Gaudio F, Martinotti S, Toniato E. Exploring the Role of Epithelial-Mesenchymal Transcriptional Factors Involved in Hematological Malignancy and Solid Tumors: A Systematic Review. Cancers (Basel) 2025; 17:529. [PMID: 39941895 PMCID: PMC11817253 DOI: 10.3390/cancers17030529] [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: 12/04/2024] [Revised: 01/13/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND The epithelial mesenchymal transition (EMT) is a biological process in which epithelial cells lose their polarity and adhesion characteristics, and adopt a mesenchymal phenotype. While the EMT naturally occurs during tissue fibrosis, wound healing, and embryonic development, it can be exploited by cancer cells and is strongly associated with cancer stem cell formation, tissue invasiveness, apoptosis, and therapy resistance. Transcription factors (TFs) such as SNAIL, ZEB, and TWIST play a pivotal role in driving the EMT. This systematic review aims to assess the impact of EMT-TFs on hematological malignancy and solid tumors. METHODS English-language literature published between 2010 and 2024 was systematically reviewed, utilizing databases such as PubMed and Google Scholar. RESULTS A total of 3250 studies were extracted. Of these, 92 publications meeting the inclusion criteria were analyzed to elucidate the role of EMT-TFs in cancer. The results demonstrated that the EMT-TFs play a critical role in both hematological and solid tumor development and progression. They promote invasive, migratory, and metastatic properties in these tumors, and contribute to therapeutic challenges by enhancing chemoresistance. A strong correlation between EMT-TFs and poor overall survival has been identified. CONCLUSIONS Our research concluded that EMT-TFs may serve as important predictive and prognostic factors, as well as potential therapeutic targets to mitigate cancer progression.
Collapse
Affiliation(s)
- Rimsha Kanwal
- Centre of Advanced Studies and Technology, Department of Innovative Technology in Medicine and Dentistry, G.d’ Annunzio University, 66100 Chieti, Italy; (R.K.); (J.E.E.); (B.J.); (S.K.Z.); (R.P.); (E.T.)
- Unit of Clinical Pathology and Microbiology, Miulli Generale Hospital, 70021 Acquaviva delle Fonti, Italy
| | - Jessica Elisabetta Esposito
- Centre of Advanced Studies and Technology, Department of Innovative Technology in Medicine and Dentistry, G.d’ Annunzio University, 66100 Chieti, Italy; (R.K.); (J.E.E.); (B.J.); (S.K.Z.); (R.P.); (E.T.)
| | - Bilal Jawed
- Centre of Advanced Studies and Technology, Department of Innovative Technology in Medicine and Dentistry, G.d’ Annunzio University, 66100 Chieti, Italy; (R.K.); (J.E.E.); (B.J.); (S.K.Z.); (R.P.); (E.T.)
- Unit of Clinical Pathology and Microbiology, Miulli Generale Hospital, 70021 Acquaviva delle Fonti, Italy
| | - Syed Khuram Zakir
- Centre of Advanced Studies and Technology, Department of Innovative Technology in Medicine and Dentistry, G.d’ Annunzio University, 66100 Chieti, Italy; (R.K.); (J.E.E.); (B.J.); (S.K.Z.); (R.P.); (E.T.)
- Unit of Clinical Pathology and Microbiology, Miulli Generale Hospital, 70021 Acquaviva delle Fonti, Italy
| | - Riccardo Pulcini
- Centre of Advanced Studies and Technology, Department of Innovative Technology in Medicine and Dentistry, G.d’ Annunzio University, 66100 Chieti, Italy; (R.K.); (J.E.E.); (B.J.); (S.K.Z.); (R.P.); (E.T.)
| | - Riccardo Martinotti
- Residency Program in Clinical Oncology, Faculty of Medicine, Umberto I University Hospital, University of Rome “La Sapienza”, 00185 Rome, Italy;
| | - Matteo Botteghi
- Experimental Pathology Research Group, Department of Clinical and Molecular Sciences, Universita Politecnica delle Marche, 60126 Ancona, Italy;
| | - Francesco Gaudio
- Unit of Haematology, Department of Medicine and Surgeon, F. Miulli University Hospital, LUM University, Casamassima, 70010 Bari, Italy
| | - Stefano Martinotti
- Unit of Clinical Pathology, Department of Medicine and Surgeon, F. Miulli University Hospital, LUM University, Casamassima, 70010 Bari, Italy
| | - Elena Toniato
- Centre of Advanced Studies and Technology, Department of Innovative Technology in Medicine and Dentistry, G.d’ Annunzio University, 66100 Chieti, Italy; (R.K.); (J.E.E.); (B.J.); (S.K.Z.); (R.P.); (E.T.)
| |
Collapse
|
15
|
Desai SA, Patel VP, Bhosle KP, Nagare SD, Thombare KC. The tumor microenvironment: shaping cancer progression and treatment response. J Chemother 2025; 37:15-44. [PMID: 38179655 DOI: 10.1080/1120009x.2023.2300224] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer progression and treatment response. It comprises a complex network of stromal cells, immune cells, extracellular matrix, and blood vessels, all of which interact with cancer cells and influence tumor behaviour. This review article provides an in-depth examination of the TME, focusing on stromal cells, blood vessels, signaling molecules, and ECM, along with commonly available therapeutic compounds that target these components. Moreover, we explore the TME as a novel strategy for discovering new anti-tumor drugs. The dynamic and adaptive nature of the TME offers opportunities for targeting specific cellular interactions and signaling pathways. We discuss emerging approaches, such as combination therapies that simultaneously target cancer cells and modulate the TME. Finally, we address the challenges and future prospects in targeting the TME. Overcoming drug resistance, improving drug delivery, and identifying new therapeutic targets within the TME are among the challenges discussed. We also highlight the potential of personalized medicine and the integration of emerging technologies, such as immunotherapy and nanotechnology, in TME-targeted therapies. This comprehensive review provides insights into the TME and its therapeutic implications. Understanding the TME's complexity and targeting its components offer promising avenues for the development of novel anti-tumor therapies and improved patient outcomes.
Collapse
Affiliation(s)
- Sharav A Desai
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| | - Vipul P Patel
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| | - Kunal P Bhosle
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| | - Sandip D Nagare
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| | - Kirti C Thombare
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| |
Collapse
|
16
|
Li Q, Yang Y, Lin X, Chu LT, Chen H, Chen L, Tang J, Zeng T. Regulation of pancreatic cancer cells by suppressing KIN17 through the PI3K/AKT/mTOR signaling pathway. Oncol Rep 2025; 53:31. [PMID: 39791213 PMCID: PMC11736091 DOI: 10.3892/or.2025.8864] [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: 09/06/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
Pancreatic cancer is an aggressive tumor, which is often associated with a poor clinical prognosis and resistance to conventional chemotherapy. Therefore, there is a need to identify new therapeutic markers for pancreatic cancer. Although KIN17 is a highly expressed DNA‑ and RNA‑binding protein in a number of types of human cancer, its role in pancreatic cancer development, especially in relation to progression, is currently unknown. The present study verified the upregulation of KIN17 in pancreatic cancer using The Cancer Genome Atlas and Gene Expression Omnibus databases (GSE15471, GSE71989 and GSE62165), and identified an association between the PI3K/Akt/mTOR pathway and patient prognosis using publicly available datasets (Gene Expression Profiling Interactive Analysis). Immunohistochemistry was performed to determine the association between KIN17 and the pathological features of clinical pancreatic cancer samples. Furthermore, knockdown of KIN17 was shown to inhibit the migration and invasion of pancreatic cancer cells, and to reverse epithelial‑mesenchymal transition in pancreatic cancer cells through downregulation of Vimentin and N‑cadherin, and upregulation of E‑cadherin. Through various cellular experiments, the role of KIIN17 was explored in PI3K/AKT/mTOR activity. KIN17 inhibition was shown to suppress the migration and invasion of pancreatic cancer cells through PI3K/AKT/mTOR‑mediated autophagy. Furthermore, combined with mTOR inhibition, dual inhibition could enhance autophagy, leading to anti‑migratory and anti‑invasion effects in pancreatic cancer. In conclusion, the present study indicated that KIN17 may have a role in carcinogenesis and could serve as a prognostic biomarker of pancreatic cancer, owing to its high expression. In addition, KIN17 may be considered a potential therapeutic target with its knockdown having an inhibitory effect on pancreatic cancer.
Collapse
Affiliation(s)
- Qiuyan Li
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Yuxia Yang
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Xiaocong Lin
- Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Lok Ting Chu
- Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Helian Chen
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Linsong Chen
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Jinjing Tang
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Tao Zeng
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| |
Collapse
|
17
|
Sun K, Zhang X, Shi J, Huang J, Wang S, Li X, Lin H, Zhao D, Ye M, Zhang S, Qiu L, Yang M, Liao C, He L, Lao M, Song J, Lu N, Ji Y, Yang H, Liu L, Liu X, Chen Y, Yao S, Xu Q, Lin J, Mao Y, Zhou J, Zhi X, Sun K, Lu X, Bai X, Liang T. Elevated protein lactylation promotes immunosuppressive microenvironment and therapeutic resistance in pancreatic ductal adenocarcinoma. J Clin Invest 2025; 135:e187024. [PMID: 39883522 PMCID: PMC11957693 DOI: 10.1172/jci187024] [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: 09/12/2024] [Accepted: 01/24/2025] [Indexed: 01/31/2025] Open
Abstract
Metabolic reprogramming shapes the tumor microenvironment (TME) and may lead to immunotherapy resistance in pancreatic ductal adenocarcinoma (PDAC). Elucidating the impact of pancreatic cancer cell metabolism in the TME is essential to therapeutic interventions. "Immune cold" PDAC is characterized by elevated lactate levels resulting from tumor cell metabolism, abundance of protumor macrophages, and reduced cytotoxic T cells in the TME. Analysis of fluorine-18 fluorodeoxyglucose (18F-FDG) uptake in patients showed that increased global protein lactylation in PDAC correlates with worse clinical outcomes in immunotherapy. Inhibition of lactate production in pancreatic tumors via glycolysis or mutant-KRAS inhibition reshaped the TME, thereby increasing their sensitivity to immune checkpoint blockade (ICB) therapy. In pancreatic tumor cells, lactate induces K63 lactylation of endosulfine α (ENSA-K63la), a crucial step that triggers STAT3/CCL2 signaling. Consequently, elevated CCL2 secreted by tumor cells facilitates tumor-associated macrophage (TAM) recruitment to the TME. High levels of lactate also drive transcriptional reprogramming in TAMs via ENSA-STAT3 signaling, promoting an immunosuppressive environment. Targeting ENSA-K63la or CCL2 enhances the efficacy of ICB therapy in murine and humanized pancreatic tumor models. In conclusion, elevated lactylation reshapes the TME and promotes immunotherapy resistance in PDAC. A therapeutic approach targeting ENSA-K63la or CCL2 has shown promise in sensitizing pancreatic cancer immunotherapy.
Collapse
Affiliation(s)
- Kang Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Xiaozhen Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Jiatao Shi
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Jinyan Huang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Sicheng Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Xiang Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Haixiang Lin
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Danyang Zhao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Mao Ye
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Sirui Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Li Qiu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Minqi Yang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Chuyang Liao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Lihong He
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Mengyi Lao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Jinyuan Song
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Na Lu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Yongtao Ji
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Hanshen Yang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Lingyue Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Xinyuan Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Yan Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Shicheng Yao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Qianhe Xu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Jieru Lin
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Yan Mao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Jingxing Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Xiao Zhi
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Ke Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Xiongbin Lu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine and
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
18
|
Sweeney A, Langley A, Xavierselvan M, Shethia RT, Solomon P, Arora A, Mallidi S. Vascular regional analysis unveils differential responses to anti-angiogenic therapy in pancreatic xenografts through macroscopic photoacoustic imaging. Theranostics 2025; 15:2649-2671. [PMID: 39990229 PMCID: PMC11840746 DOI: 10.7150/thno.99361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 12/17/2024] [Indexed: 02/25/2025] Open
Abstract
Background: Amongst the various imaging techniques that provide surrogate tumor radiographic indications to aid in planning, monitoring, and predicting outcomes of therapy, ultrasound-guided photoacoustic imaging (US-PAI) is a promising non-ionizing modality based on endogenous blood (hemoglobin) and blood oxygen saturation (StO₂) contrast. Adaptation of US-PAI to the clinical realm requires macroscopic system configurations for adequate depth visualization. Methods: Here we present a vascular regional analysis (VRA) methodology of obtaining areas of low and high vessel density regions within the tumor (LVD and HVD respectively) by frequency domain filtering of macroscopic PA images. In this work, we evaluated the various vascular and oxygenation profiles of different murine xenografts of pancreatic cancer (AsPC-1, MIA PaCa-2, and BxPC-3) that have varying levels of angiogenic potentials and investigated the effects of receptor tyrosine kinase inhibitor (sunitinib) on the tumor microvessel density and StO₂. Results: The administration of sunitinib resulted in transient deoxygenation and reduction in vessel density within 72 h in two (AsPC-1 and MIA PaCa-2) of the three tumor types. Utilizing VRA, the regional change in StO2 (∆StO2) revealed the preferential targeting of sunitinib in LVD regions in only the AsPC-1 tumors. We also identified the presence of vascular normalization (validated through immunohistochemistry) in the sunitinib treated AsPC-1 tumors at day 8 post-treatment where a significant increases in HVD ∆StO2 (~20%) were seen following the 72-hour time point, indicative of improved vessel flow and functionality. Treated AsPC-1 vasculature displayed increased maturity and functionality compared to non-treated tumors on day 8, while these same metrics showed no conclusive evidence of vascular normalization in MIA PaCa-2 or BxPC-3 tumors. Conclusion: Overall, VRA as a tool to monitor treatment response allowed us to identify time points of vascular remodeling, highlighting its ability to provide insights into the tumor microenvironment for sunitinib treatment and other anti-angiogenic therapies.
Collapse
Affiliation(s)
- Allison Sweeney
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Andrew Langley
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Marvin Xavierselvan
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Ronak T. Shethia
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Patrick Solomon
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Aayush Arora
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Srivalleesha Mallidi
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
19
|
Nan D, Yao W, Huang L, Liu R, Chen X, Xia W, Sheng H, Zhang H, Liang X, Lu Y. Glutamine and cancer: metabolism, immune microenvironment, and therapeutic targets. Cell Commun Signal 2025; 23:45. [PMID: 39856712 PMCID: PMC11760113 DOI: 10.1186/s12964-024-02018-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
Glutamine is the most abundant amino acid in human serum, and it can provide carbon and nitrogen for biosynthesis, which is crucial for proliferating cells. Moreover, it is widely known that glutamine metabolism is reprogrammed in cancer cells. Many cancer cells undergo metabolic reprogramming targeting glutamine, increasing its uptake to meet their rapid proliferation demands. An increasing amount of study is being done on the particular glutamine metabolic pathways in cancer cells.Further investigation into the function of glutamine in immune cells is warranted given the critical role these cells play in the fight against cancer. Immune cells use glutamine for a variety of biological purposes, including the growth, differentiation, and destruction of cancer cells. With the encouraging results of cancer immunotherapy in recent years, more investigation into the impact of glutamine metabolism on immune cell function in the cancer microenvironment could lead to the discovery of new targets and therapeutic approaches.Oral supplementation with glutamine also enhances the immune capabilities of cancer patients, improves the sensitivity to chemotherapy and radiotherapy, and improves prognosis. The unique metabolism of glutamine in cancer cells, its function in various immune cells, the impact of inhibitors of glutamine metabolism, and the therapeutic use of glutamine supplements are all covered in detail in this article.
Collapse
Affiliation(s)
- Ding Nan
- School of Clinical Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Department of Radiation Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Weiping Yao
- Department of Radiation Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Luanluan Huang
- Department of Radiation Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ruiqi Liu
- Department of Radiation Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaoyan Chen
- Department of Radiation Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wenjie Xia
- Department of Breast Surgery, General Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Hailong Sheng
- Department of Radiation Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Haibo Zhang
- School of Clinical Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China.
- Department of Radiation Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Xiaodong Liang
- School of Clinical Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China.
- Department of Radiation Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Yanwei Lu
- School of Clinical Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China.
- Department of Radiation Oncology, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
20
|
Chao YY, Lin RC, Su PJ, Wang CA, Tu TY, Hou YC, Tsai YT, Peng IC, Tsai SJ, Shan YS, Wang CY. Melanophilin-induced primary cilia promote pancreatic cancer metastasis. Cell Death Dis 2025; 16:22. [PMID: 39820281 PMCID: PMC11739566 DOI: 10.1038/s41419-025-07344-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 12/11/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant tumors because of its high metastatic ability. The glutamine (Gln)-deficient microenvironment contributes to PDAC metastasis; however, the underlying molecular mechanisms remain unclear. Here, we demonstrated that melanophilin (MLPH) promotes PDAC metastasis by inducing the regrowth of primary cilia. Using RNA sequencing, we found that MLPH was upregulated in Gln-deficient conditions. MLPH facilitated PDAC metastasis in vitro and in vivo. Clinically, high MLPH expression is positively correlated with metastasis and poor PDAC prognosis. MLPH localized to the centrosome and facilitated the regrowth of primary cilia. The primary ciliogenesis upregulated phospholipase C γ-1 (PLCG1) to promote PDAC metastasis. Interestingly, PLCG1 was localized to the primary cilia, and depletion of PLCG1 alleviated primary ciliogenesis, suggesting a feedforward role for PLCG1 in mediating primary ciliogenesis. Thus, our study revealed a novel function of the MLPH-primary cilia-PLCG1 axis in facilitating PDAC metastasis under Gln deficiency both in vitro and in vivo.
Collapse
Affiliation(s)
- Yu-Ying Chao
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ruei-Ci Lin
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ping-Jui Su
- Division of General Surgery, Department of Surgery, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Chu-An Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ting-Yuan Tu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Chin Hou
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Tzui Tsai
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - I-Chen Peng
- Department of Life Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Shaw-Jenq Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yan-Shen Shan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Surgery, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Yih Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
21
|
Tang HY, Cao YZ, Zhou YW, Ma YS, Jiang H, Zhang H, Jiang L, Yang QX, Tang XM, Yang C, Liu XY, Liu FX, Liu JB, Fu D, Wang YF, Yu H. The power and the promise of CAR-mediated cell immunotherapy for clinical application in pancreatic cancer. J Adv Res 2025; 67:253-267. [PMID: 38244773 PMCID: PMC11725162 DOI: 10.1016/j.jare.2024.01.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/24/2023] [Accepted: 01/11/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Pancreatic cancer, referred to as the "monarch of malignancies," is a neoplastic growth mostly arising from the epithelial cells of the pancreatic duct and acinar cells. This particular neoplasm has a highly unfavorable prognosis due to its marked malignancy, inconspicuous initial manifestation, challenging early detection, rapid advancement, and limited survival duration. Cellular immunotherapy is the ex vivo culture and expansion of immune effector cells, granting them the capacity to selectively target malignant cells using specialized techniques. Subsequently, these modified cells are reintroduced into the patient's organism with the purpose of eradicating tumor cells and providing therapeutic intervention for cancer. PRESENT SITUATION Presently, the primary cellular therapeutic modalities employed in the treatment of pancreatic cancer encompass CAR T-cell therapy, TCR T-cell therapy, NK-cell therapy, and CAR NK-cell therapy. AIM OF REVIEW This review provides a concise overview of the mechanisms and primary targets associated with various cell therapies. Additionally, we will explore the prospective outlook of cell therapy in the context of treating pancreatic cancer.
Collapse
Affiliation(s)
- Hao-Yu Tang
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China; Institute of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong 226631, Jiangsu, China; General Surgery, Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, Shanghai, China
| | - Yi-Zhi Cao
- General Surgery, Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, Shanghai, China
| | - Yi-Wei Zhou
- General Surgery, Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, Shanghai, China
| | - Yu-Shui Ma
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, Shanghai, China
| | - Hong Jiang
- Department of Thoracic Surgery, The 905th Hospital of Chinese People's Liberation Army Navy, Shanghai 200050, Shanghai, China
| | - Hui Zhang
- Institute of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong 226631, Jiangsu, China
| | - Lin Jiang
- Department of Anesthesiology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu 225300, China
| | - Qin-Xin Yang
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China
| | - Xiao-Mei Tang
- General Surgery, Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, Shanghai, China
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xin-Yun Liu
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China
| | - Fu-Xing Liu
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China
| | - Ji-Bin Liu
- Institute of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong 226631, Jiangsu, China.
| | - Da Fu
- Institute of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong 226631, Jiangsu, China; General Surgery, Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, Shanghai, China.
| | - Yun-Feng Wang
- Department of General Surgery, Pudong New Area People's Hospital, Shanghai 201299, China.
| | - Hong Yu
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China; Department of Pathology, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou 225300, Jiangsu, China.
| |
Collapse
|
22
|
Huang Y, Zhang R, Fan S, Shi M, Tang X, Wang X, Deng X. OSBPL10-CNBP axis mediates hypoxia-induced pancreatic cancer development. Biofactors 2025; 51:e2124. [PMID: 39329194 DOI: 10.1002/biof.2124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of malignancies with worst outcomes among digestive system tumors. Identification of novel biomarkers is of great significance for treatment researches and prognosis prediction of pancreatic cancer patients. Due to OSBPL10 known involvement in oncogenic activity in other tumors, we elucidated the mechanism underlying its contribution to pancreatic cancer progression. We employed data from the Gene Expression Omnibus database to detect the expression of OSBPL10 in normal and pancreatic cancer tissues. A series of assays were conducted to assess the impact of OSBPL10 on the proliferation and metastatic capacities of pancreatic cancer cells and the influence of OSBPL10 on macrophages were evaluated by Flow cytometry. In addition, Co-immunoprecipitation, mass spectrometry, and western blot assays were utilized to investigate the potential mechanisms of OSBPL10 activity. From our study, OSBPL10 is revealed to be upregulated in pancreatic cancer, with poor prognosis. The overexpression promotes malignant behaviors of pancreatic cancer cells and has an impact on tumor immune microenvironment by stimulating the transformation M1 macrophages into M2 macrophages. Mechanistically, hypoxia induces the expression of OSBPL10 through interaction between hypoxia-inducible factor 1-α and the promoter region of OSBPL10. Additionally, OSBPL10 directly bound to CNBP, mediating CNBP expression and ultimately regulating the proliferation and metastasis capacity of pancreatic cancer cells, as well as influencing macrophage polarization. The research emphasized the oncogenic role of OSBPL10 in pancreatic cancer, uncovering key mechanisms involving hypoxia, HIF-1α, and CNBP. The finding suggests that OSBPL10 is a novel biomarker in pancreatic cancer, making it a potential therapeutic target for intervention in this malignancy.
Collapse
Affiliation(s)
- Yishu Huang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Neoplasms Translational Medicine, Shanghai, China
| | - Ronghao Zhang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shuyang Fan
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
| | - Minmin Shi
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
| | - Xiaomei Tang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xinjing Wang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaxing Deng
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
23
|
Acimovic I, Gabrielová V, Martínková S, Eid M, Vlažný J, Moravčík P, Hlavsa J, Moráň L, Cakmakci RC, Staňo P, Procházka V, Kala Z, Trnka J, Vaňhara P. Ex-Vivo 3D Cellular Models of Pancreatic Ductal Adenocarcinoma: From Embryonic Development to Precision Oncology. Pancreas 2025; 54:e57-e71. [PMID: 39074056 DOI: 10.1097/mpa.0000000000002393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
ABSTRACT Pancreas is a vital gland of gastrointestinal system with exocrine and endocrine secretory functions, interweaved into essential metabolic circuitries of the human body. Pancreatic ductal adenocarcinoma (PDAC) represents one of the most lethal malignancies, with a 5-year survival rate of 11%. This poor prognosis is primarily attributed to the absence of early symptoms, rapid metastatic dissemination, and the limited efficacy of current therapeutic interventions. Despite recent advancements in understanding the etiopathogenesis and treatment of PDAC, there remains a pressing need for improved individualized models, identification of novel molecular targets, and development of unbiased predictors of disease progression. Here we aim to explore the concept of precision medicine utilizing 3-dimensional, patient-specific cellular models of pancreatic tumors and discuss their potential applications in uncovering novel druggable molecular targets and predicting clinical parameters for individual patients.
Collapse
Affiliation(s)
- Ivana Acimovic
- From the Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno
| | - Viktorie Gabrielová
- From the Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno
| | - Stanislava Martínková
- Department of Biochemistry, Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague
| | - Michal Eid
- Departments of Internal Medicine, Hematology and Oncology
| | | | - Petr Moravčík
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | - Jan Hlavsa
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | | | - Riza Can Cakmakci
- From the Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno
| | - Peter Staňo
- Departments of Internal Medicine, Hematology and Oncology
| | - Vladimír Procházka
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | - Zdeněk Kala
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | - Jan Trnka
- Department of Biochemistry, Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague
| | | |
Collapse
|
24
|
Wang Y, Peng L, Wang F. M6A-mediated molecular patterns and tumor microenvironment infiltration characterization in nasopharyngeal carcinoma. Cancer Biol Ther 2024; 25:2333590. [PMID: 38532632 DOI: 10.1080/15384047.2024.2333590] [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: 08/02/2023] [Accepted: 03/18/2024] [Indexed: 03/28/2024] Open
Abstract
N6-methyladenosine (m6A) is the most predominant RNA epigenetic regulation in eukaryotic cells. Numerous evidence revealed that m6A modification exerts a crucial role in the regulation of tumor microenvironment (TME) cell infiltration in several tumors. Nevertheless, the potential role and mechanism of m6A modification in nasopharyngeal carcinoma (NPC) remains unknown. mRNA expression data and clinical information from GSE102349, and GSE53819 datasets obtained from Gene Expression Omnibus (GEO) was used for differential gene expression and subsequent analysis. Consensus clustering was used to identify m6A-related molecular patterns of 88 NPC samples based on prognostic m6A regulators using Univariate Cox analysis. The TME cell-infiltrating characteristics of each m6A-related subclass were explored using single-sample gene set enrichment (ssGSEA) algorithm and CIBERSORT algotithm. DEGs between two m6A-related subclasses were screened using edgeR package. The prognostic signature and predicated nomogram were constructed based on the m6A-related DEGs. The cell infiltration and expression of prognostic signature in NPC was determined using immunohistochemistry (IHC) analysis. Chi-square test was used to analysis the significance of difference of the categorical variables. And survival analysis was performed using Kaplan-Meier plots and log-rank tests. The NPC samples were divided into two m6A-related subclasses. The TME cell-infiltrating characteristics analyses indicated that cluster 1 is characterized by immune-related and metabolism pathways activation, better response to anit-PD1 and anti-CTLA4 treatment and chemotherapy. And cluster 2 is characterized by stromal activation, low expression of HLA family and immune checkpoints, and a worse response to anti-PD1 and anti-CTLA4 treatment and chemotherapy. Furthermore, we identified 1558 DEGs between two m6A-related subclasses and constructed prognostic signatures to predicate the progression-free survival (PFS) for NPC patients. Compared to non-tumor samples, REEP2, TMSB15A, DSEL, and ID4 were upregulated in NPC samples. High expression of REEP2 and TMSB15A showed poor survival in NPC patients. The interaction between REEP2, TMSB15A, DSEL, ID4, and m6A regulators was detected. Our finding indicated that m6A modification plays an important role in the regulation of TME heterogeneity and complexity.
Collapse
Affiliation(s)
- Yong Wang
- Department of Radiotherapy, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Lisha Peng
- Department of Radiotherapy, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Feng Wang
- Department of Radiotherapy, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
25
|
Du W, Tang Z, Du A, Yang Q, Xu R. Bidirectional crosstalk between the epithelial-mesenchymal transition and immunotherapy: A bibliometric study. Hum Vaccin Immunother 2024; 20:2328403. [PMID: 38502119 PMCID: PMC10956627 DOI: 10.1080/21645515.2024.2328403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 03/06/2024] [Indexed: 03/20/2024] Open
Abstract
Immunotherapy has recently attracted considerable attention. However, currently, a thorough analysis of the trends associated with the epithelial-mesenchymal transition (EMT) and immunotherapy is lacking. In this study, we used bibliometric tools to provide a comprehensive overview of the progress in EMT-immunotherapy research. A total of 1,302 articles related to EMT and immunotherapy were retrieved from the Web of Science Core Collection (WOSCC). The analysis indicated that in terms of the volume of research, China was the most productive country (49.07%, 639), followed by the United States (16.89%, 220) and Italy (3.6%, 47). The United States was the most influential country according to the frequency of citations and citation burstiness. The results also suggested that Frontiers in Immunotherapy can be considered as the most influential journal with respect to the number of articles and impact factors. "Immune infiltration," "bioinformatics analysis," "traditional Chinese medicine," "gene signature," and "ferroptosis" were found to be emerging keywords in EMT-immunotherapy research. These findings point to potential new directions that can deepen our understanding of the mechanisms underlying the combined effects of immunotherapy and EMT and help develop strategies for improving immunotherapy.
Collapse
Affiliation(s)
- Wei Du
- Department of Pathology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan, China
| | - Zemin Tang
- Department of Pathology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan, China
| | - Ashuai Du
- Department of Infectious Diseases, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Qinglong Yang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
- Department of General Surgery, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Rong Xu
- Department of Pathology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan, China
| |
Collapse
|
26
|
Gu J, Xiao X, Zou C, Mao Y, Jin C, Fu D, Li R, Li H. Ubiquitin-specific protease 7 maintains c-Myc stability to support pancreatic cancer glycolysis and tumor growth. J Transl Med 2024; 22:1135. [PMID: 39707401 PMCID: PMC11662425 DOI: 10.1186/s12967-024-05962-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND The typical pathological feature of pancreatic ductal adenocarcinoma (PDAC) is a significant increase in stromal reaction, leading to a hypoxic and poorly vascularized tumor microenvironment. Tumor cells undergo metabolic reprogramming, such as the Warburg effect, yet the underlying mechanisms are not fully understood. METHODS Interference and overexpression experiments were conducted to analyze the in vivo and in vitro effects of USP7 on the growth and glycolysis of tumor cells. Small-molecule inhibitors of USP7 and transgenic mouse models of PDAC were employed to assess the consequences of targeting USP7 in PDAC. The molecular mechanism underlying USP7-induced c-Myc stabilization was determined by RNA sequencing, co-IP and western blot analyses. RESULTS USP7 is abnormally overexpressed in PDAC and predicts a poor prognosis. Hypoxia and extracellular matrix stiffness can induce USP7 expression in PDAC cells. Genetic silencing of USP7 inhibits the glycolytic phenotypes in PDAC cells, while its overexpression has the opposite effect, as demonstrated by glucose uptake, lactate production, and extracellular acidification rate. Importantly, USP7 promotes PDAC tumor growth in a glycolysis-dependent manner. The small-molecule inhibitor P5091 targeting USP7 effectively suppresses the Warburg effect and cell growth in PDAC. In a transgenic mouse model of PDAC, named KPC, P5091 effectively blocks tumor progression. Mechanistically, USP7 interacts with c-Myc, enhancing its stability and expression, which in turn upregulates expression of glycolysis-related genes. CONCLUSIONS This study sheds light on the molecular mechanisms underlying the Warburg effect in PDAC and unveils USP7 as a potential therapeutic target for improving PDAC treatment.
Collapse
Affiliation(s)
- Jichun Gu
- Department of Pancreatic surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xi Xiao
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Caifeng Zou
- Department of Pancreatic surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yishen Mao
- Department of Pancreatic surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Chen Jin
- Department of Pancreatic surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Deliang Fu
- Department of Pancreatic surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Rongkun Li
- Chest Oncology Department, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| | - Hengchao Li
- Department of Pancreatic surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
27
|
Shaik R, Malik MS, Basavaraju S, Qurban J, Al-Subhi FMM, Badampudi S, Peddapaka J, Shaik A, Abd-El-Aziz A, Moussa Z, Ahmed SA. Cellular and molecular aspects of drug resistance in cancers. Daru 2024; 33:4. [PMID: 39652186 PMCID: PMC11628481 DOI: 10.1007/s40199-024-00545-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 10/09/2024] [Indexed: 12/12/2024] Open
Abstract
OBJECTIVES Cancer drug resistance is a multifaceted phenomenon. The present review article aims to comprehensively analyze the cellular and molecular aspects of drug resistance in cancer and the strategies employed to overcome it. EVIDENCE ACQUISITION A systematic search of relevant literature was conducted using electronic databases such as PubMed, Scopus, and Web of Science using appropriate key words. Original research articles and secondary literature were taken into consideration in reviewing the development in the field. RESULTS AND CONCLUSIONS Cancer drug resistance is a pervasive challenge that causes many treatments to fail therapeutically. Despite notable advances in cancer treatment, resistance to traditional chemotherapeutic agents and novel targeted medications remains a formidable hurdle in cancer therapy leading to cancer relapse and mortality. Indeed, a majority of patients with metastatic cancer experience are compromised on treatment efficacy because of drug resistance. The multifaceted nature of drug resistance encompasses various factors, such as tumor heterogeneity, growth kinetics, immune system, microenvironment, physical barriers, and the emergence of undruggable cancer drivers. Additionally, alterations in drug influx/efflux transporters, DNA repair mechanisms, and apoptotic pathways further contribute to resistance, which may manifest as either innate or acquired traits, occurring prior to or following therapeutic intervention. Several strategies such as combination therapy, targeted therapy, development of P-gp inhibitors, PROTACs and epigenetic modulators are developed to overcome cancer drug resistance. The management of drug resistance is compounded by the patient and tumor heterogeneity coupled with cancer's ability to evade treatment. Gaining further insight into the mechanisms underlying medication resistance is imperative for the development of effective therapeutic interventions and improved patient outcomes.
Collapse
Affiliation(s)
- Rahaman Shaik
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - M Shaheer Malik
- Department of Chemistry, Faculty of Science, Umm Al-Qura University, Makkah, 21955, Saudi Arabia.
| | | | - Jihan Qurban
- Department of Chemistry, Faculty of Science, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Fatimah M M Al-Subhi
- Department of Environmental and Occupational Health, College of Public Health and Health Informatics, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Sathvika Badampudi
- Department of Pharmacology, St.Pauls College of Pharmacy, Turkayamjal, Hyderabad, India
| | - Jagruthi Peddapaka
- Department of Pharmaceutical Chemistry, St.Paul's College of Pharmacy, Turkayamjal, Hyderabad, India
| | - Azeeza Shaik
- Research&Development Department, KVB Asta Life sciences, Hyderabad, India
| | - Ahmad Abd-El-Aziz
- Qingdao Innovation and Development Center, Harbin Engineering University, Qingdao, 266400, China
| | - Ziad Moussa
- Department of Chemistry, College of Science, United Arab Emirates University, P.O. Box 15551, Al Ain, United Arab Emirates
| | - Saleh A Ahmed
- Department of Chemistry, Faculty of Science, Umm Al-Qura University, Makkah, 21955, Saudi Arabia.
- Department of Chemistry, Faculty of Science, Assiut University, Assiut, 71516, Egypt.
| |
Collapse
|
28
|
Mortoglou M, Lian M, Miralles F, Dart DA, Uysal-Onganer P. miR-210 Mediated Hypoxic Responses in Pancreatic Ductal Adenocarcinoma. ACS OMEGA 2024; 9:47872-47883. [PMID: 39651070 PMCID: PMC11618397 DOI: 10.1021/acsomega.4c08947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 12/11/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one among the most lethal malignancies due to its aggressive behavior and resistance to conventional therapies. Hypoxia significantly contributes to cancer progression and therapeutic resistance of PDAC. microRNAs (miRNAs/miRs) have emerged as critical regulators of various biological processes. miR-210 is known as the "hypoxamir" due to its prominent role in cellular responses to hypoxia. In this study, we investigated the multifaceted role of miR-210 in PDAC using miR-210 knockout (KO) cellular models to elucidate its functions under hypoxic conditions. Hypoxia-inducible factor-1α (HIF1-α), a key transcription factor activated in response to low oxygen levels, upregulates miR-210. miR-210 maintains cancer stem cell (CSC) phenotypes and promotes epithelial-mesenchymal transition (EMT), which is essential for tumor initiation, metastasis, and therapeutic resistance. Our findings demonstrate that miR-210 regulates the expression of CSC markers, such as CD24, CD44, and CD133, and EMT markers, including E-cadherin, Vimentin, and Snail. Specifically, depletion of miR-210 reversed EMT and CSC marker expression levels in hypoxic Panc-1 and MiaPaCa-2 PDAC cells. These regulatory actions facilitate a more invasive and treatment-resistant PDAC phenotype. Understanding the regulatory network involving miR-210 under hypoxic conditions may reveal new therapeutic targets for combating PDAC and improving patient outcomes. Our data suggest that miR-210 is a critical regulator of HIF1-α expression, EMT, and the stemness of PDAC cells in hypoxic environments.
Collapse
Affiliation(s)
- Maria Mortoglou
- Cancer
Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, U.K.
| | - Mutian Lian
- Cancer
Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, U.K.
| | - Francesc Miralles
- School
of Health and Medical Sciences, City St
George’s, University of London, Cranmer Terrace, London SW17 0RE, U.K.
| | - D. Alwyn Dart
- UCL
Cancer Institute, University College London, Paul O’Gorman Building, 72
Huntley Street, London WC1E 6DD, U.K.
| | - Pinar Uysal-Onganer
- Cancer
Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, U.K.
| |
Collapse
|
29
|
Nair ST, Abhi C, Kamalasanan K, Pavithran K, Unni AR, Sithara MS, Sarma M, Mangalanandan TS. Pathophysiology-Driven Approaches for Overcoming Nanomedicine Resistance in Pancreatic Cancer. Mol Pharm 2024; 21:5960-5988. [PMID: 39561094 DOI: 10.1021/acs.molpharmaceut.4c00801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Tumor heterogeneity poses a significant challenge in cancer therapy. To address this, we analyze pharmacotherapeutic challenges by categorizing them into static and dynamic barriers, reframing these challenges to improve drug delivery, efficacy, and the development of controlled-release nanomedicines (CRNMs). This pathophysiology-driven approach facilitates the design of novel therapeutics tailored to overcome obstacles in pancreatic ductal adenocarcinoma (PDAC) using nanotechnology. Advanced biomaterials in nanodrug delivery systems offer innovative solutions by combining controlled release, stimuli sensitivity, and smart design strategies. CRNMs are engineered to modulate spatiotemporal signaling and control drug release in PDAC, where resistance to conventional therapies is particularly high. This review explores pharmacokinetic considerations for nanomedicine design, RNA interference (RNAi) for stromal modulation, and the development of targeted nanomedicine strategies. Additionally, we highlight the limitations of current animal models in capturing the complexities of PDAC and discuss notable clinical failures, such as PEGylated hyaluronidase (Phase III HALO 109-301 trial) and evofosfamide (TH-302) with gemcitabine (MAESTRO trial), underscoring the need for improved models and treatment strategies. By targeting pathways like Notch and Hedgehog and incorporating stimuli-sensitive and pathway-modulating agents, CRNMs offer a promising avenue to enhance drug penetration and efficacy, reshaping the paradigm of pancreatic cancer treatment.
Collapse
Affiliation(s)
- Sreejith Thrivikraman Nair
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - C Abhi
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - Kaladhar Kamalasanan
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - K Pavithran
- Department of Medical Oncology and Hematology, School of Medicine, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - Ashok R Unni
- Department of Veterinary Medicine, Central Animal Facility, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - M S Sithara
- Department of Veterinary Medicine, Central Animal Facility, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - Manjit Sarma
- Department of Nuclear Medicine, Amrita School of Medicine, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - T S Mangalanandan
- Department of Endocrinology, Amrita Institute of Medical Sciences and Research Centre, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| |
Collapse
|
30
|
Xu Y, Zhu K, Wu J, Zheng S, Zhong R, Zhou W, Cao Y, Liu J, Wang H. HBOC alleviated tumour hypoxia during radiotherapy more intensely in large solid tumours than regular ones. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:1-14. [PMID: 37994792 DOI: 10.1080/21691401.2023.2276768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/07/2023] [Indexed: 11/24/2023]
Abstract
Radiotherapy (RT) is a highly valuable method in cancer therapy, but its therapeutic efficacy is limited by its side effects and tumour radiation resistance. The resistance is mainly induced by hypoxia in the tumour microenvironment (TME). As a nano-oxygen carrier, Haemoglobin-based oxygen carriers (HBOCs) administration is a promising strategy to alleviate tumour hypoxia which may remodel TME to ameliorate radiation resistance and enable RT more effective. In this study, we administered fractionated RT combined with HBOC to treat Miapaca-2 cell and Hela cell xenografts on nude mice. The study found that HBOC relieved hypoxic environment and down-regulate expression of hypoxia-inducible factor-1α (Hif-1α) both in regular (100 mm3) and large (360/400 mm3) tumours. The proliferation and metastasis of tumour tissue also decreased after HBOC application. Nevertheless, in vivo RT combined with HBOC performed more effectively to suppress tumour growth in large tumours than in regular tumours. This is due to more severe hypoxic regions exist in the large solid tumours compared to the regular counterparts, and HBOC administration may be more effective in alleviating hypoxia in large tumours. Thus, HBOC sensitization therapy is more suitable for large solid tumours.
Collapse
Affiliation(s)
- Yingcan Xu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Peking Union Medical College, Chengdu, China
| | - Kehui Zhu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Peking Union Medical College, Chengdu, China
| | - Jiakang Wu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Peking Union Medical College, Chengdu, China
| | - Shifan Zheng
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Peking Union Medical College, Chengdu, China
| | - Rui Zhong
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Peking Union Medical College, Chengdu, China
| | - Wentao Zhou
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Peking Union Medical College, Chengdu, China
| | - Ye Cao
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Peking Union Medical College, Chengdu, China
| | - Jiaxin Liu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Peking Union Medical College, Chengdu, China
| | - Hong Wang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Peking Union Medical College, Chengdu, China
| |
Collapse
|
31
|
Li DM, Cao XY, Jiang J. Hypoxia-related bioinformatic signatures associated with prognosis and tumor microenvironment of pancreatic cancer: Current status, concerns, and future perspectives. World J Gastroenterol 2024; 30:4689-4696. [PMID: 39610772 PMCID: PMC11580612 DOI: 10.3748/wjg.v30.i44.4689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/25/2024] [Accepted: 11/04/2024] [Indexed: 11/12/2024] Open
Abstract
Pancreatic cancer (PC), a highly lethal tumor with nearly identical incidence and mortality rates, has become the sixth leading cause of cancer-related deaths. Hypoxia is an important malignant factor in PC, as it regulates angiogenesis, metabolic reprogramming, tumor progression, and metastasis. Disrupting the hypoxic microenvironment can enhance the efficacy of antitumor therapy and improve the prognosis of patients with PC. With the advent of bioinformatics, hypoxia-related PC models have emerged in recent years. They provide a reference for estimating the prognosis and immune microenvironment of patients with PC and identify potential biomarkers for targeting hypoxic microenvironment. However, these findings based on bioinformatic analysis may not be completely reliable without further experimental evidence and clinical cohort validation. The application of these models and biomarkers in clinical practice to predict survival time and develop anti hypoxic therapeutic strategies for patients with PC remains in its infancy. In this editorial, we review the current status of hypoxia-related prognostic models in PC, analyze their similarities and differences, discuss several existing challenges, and provide potential solutions and directions for further studies. This editorial will facilitate the optimization, validation, and determination of the molecular mechanisms of related models.
Collapse
Affiliation(s)
- Dong-Ming Li
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Xue-Yuan Cao
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Jing Jiang
- Department of Clinical Epidemiology, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|
32
|
Xu E, Huang Z, Zhu K, Hu J, Ma X, Wang Y, Zhu J, Zhang C. PDGFRB promotes dedifferentiation and pulmonary metastasis through rearrangement of cytoskeleton under hypoxic microenvironment in osteosarcoma. Cell Signal 2024; 125:111501. [PMID: 39505287 DOI: 10.1016/j.cellsig.2024.111501] [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: 04/08/2024] [Revised: 10/15/2024] [Accepted: 10/30/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Osteosarcoma (OS) cells commonly suffer from hypoxia and dedifferentiation, resulting in poor prognosis. We plan to identify the role of hypoxia on dedifferentiation and the associated cellular signaling. METHODS We performed sphere formation assays and determined spheroid cells as dedifferentiated cells by detecting stem cell-like markers. RNAi assay was used to explore the relationship between hypoxia inducible factor 1 subunit alpha (HIF1A) and platelet derived growth factor receptor beta (PDGFRB). We obtained PDGFRB knockdown and overexpression cells through lentiviral infection experiments and detected the expression of PDGFRB, p-PDGFRB, focal adhesion kinase (FAK), p-FAK, phosphorylated myosin light chain 2 (p-MLC2), and ras homolog family member A (RhoA) in each group. The effects of PDGFRB on cytoskeleton rearrangement and cell adhesion were explored by immunocytochemistry. Wound-healing experiments, transwell assays, and animal trials were employed to investigate the effect of PDGFRB on OS cell metastasis both in vitro and in vivo. RESULTS Dedifferentiated OS cells were found to exhibit high expression of HIF1A and PDGFRB, and HIF1A upregulated PDGFRB, subsequently activated RhoA, and increased the phosphorylation of MLC2. PDGFRB also enhanced the phosphorylation of FAK. The OS cell morphology and vinculin distribution were altered by PDGFRB. PDGFRB promoted cell dedifferentiation and had a significant impact on the migration and invasion abilities of OS cells in vitro. In addition, PDGFRB increased pulmonary metastasis of OS cells in vivo. CONCLUSION Our results demonstrated that HIF1A up-regulated PDGFRB under hypoxic conditions, and PDGFRB regulated the actin cytoskeleton, a process likely linked to the activation of RhoA and the phosphorylation of, thereby promoting OS dedifferentiation and pulmonary metastasis.
Collapse
Affiliation(s)
- Enjie Xu
- Department of Orthopedic Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, PR China; Institute of Bone Tumor Affiliated to Tongji University School of Medicine, Shanghai 200072, PR China
| | - Zhen Huang
- Department of Orthopedic Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, PR China; Institute of Bone Tumor Affiliated to Tongji University School of Medicine, Shanghai 200072, PR China
| | - Kunpeng Zhu
- Department of Orthopedic Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, PR China; Institute of Bone Tumor Affiliated to Tongji University School of Medicine, Shanghai 200072, PR China
| | - Jianping Hu
- Department of Orthopedic Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, PR China; Institute of Bone Tumor Affiliated to Tongji University School of Medicine, Shanghai 200072, PR China
| | - Xiaolong Ma
- Department of Orthopedic Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, PR China; Institute of Bone Tumor Affiliated to Tongji University School of Medicine, Shanghai 200072, PR China
| | - Yongjie Wang
- Department of Orthopedic Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, PR China; Institute of Bone Tumor Affiliated to Tongji University School of Medicine, Shanghai 200072, PR China
| | - Jiazhuang Zhu
- Department of Orthopedic Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, PR China; Institute of Bone Tumor Affiliated to Tongji University School of Medicine, Shanghai 200072, PR China
| | - Chunlin Zhang
- Department of Orthopedic Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, PR China; Institute of Bone Tumor Affiliated to Tongji University School of Medicine, Shanghai 200072, PR China.
| |
Collapse
|
33
|
Kennedy PR, Arvindam US, Phung SK, Ettestad B, Feng X, Li Y, Kile QM, Hinderlie P, Khaw M, Huang RS, Kaufman M, Puchalska P, Russell A, Butler J, Abbott L, McClure P, Luo X, Lu QT, Blazar BR, Crawford PA, Lim J, Miller JS, Felices M. Metabolic programs drive function of therapeutic NK cells in hypoxic tumor environments. SCIENCE ADVANCES 2024; 10:eadn1849. [PMID: 39475618 PMCID: PMC11524192 DOI: 10.1126/sciadv.adn1849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 09/24/2024] [Indexed: 11/02/2024]
Abstract
Limited oxygen (hypoxia) in solid tumors poses a challenge to successful immunotherapy with natural killer (NK) cells. NK cells have impaired cytotoxicity when cultured in hypoxia (1% oxygen) but not physiologic (>5%) or atmospheric oxygen (20%). We found that changes to cytotoxicity were regulated at the transcriptional level and accompanied by metabolic dysregulation. Dosing with interleukin-15 (IL-15) enhanced NK cell cytotoxicity in hypoxia, but preactivation with feeder cells bearing IL-21 and 4-1BBL was even better. Preactivation resulted in less perturbed metabolism in hypoxia; greater resistance to oxidative stress; and no hypoxia-induced loss of transcription factors (T-bet and Eomes), activating receptors, adhesion molecules (CD2), and cytotoxic proteins (TRAIL and FasL). There remained a deficit in CD122/IL-2Rβ when exposed to hypoxia, which affected IL-15 signaling. However, tri-specific killer engager molecules that deliver IL-15 in the context of anti-CD16/FcγRIII were able to bypass this deficit, enhancing cytotoxicity of both fresh and preactivated NK cells in hypoxia.
Collapse
Affiliation(s)
- Philippa R. Kennedy
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Upasana Sunil Arvindam
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Shee Kwan Phung
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Brianna Ettestad
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | | | - Yunmin Li
- Xcell Biosciences, San Francisco, CA, USA
| | - Quinlan M. Kile
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Peter Hinderlie
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Melissa Khaw
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Rih-Sheng Huang
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Marissa Kaufman
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Patrycja Puchalska
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Amanda Russell
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Jonah Butler
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Lucas Abbott
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Paul McClure
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Xianghua Luo
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | | | - Bruce R. Blazar
- Division of Pediatric Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Peter A. Crawford
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - James Lim
- Xcell Biosciences, San Francisco, CA, USA
| | - Jeffrey S. Miller
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Martin Felices
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
34
|
Yang L, Qiao S, Zhang G, Lu A, Li F. Inflammatory Processes: Key Mediators of Oncogenesis and Progression in Pancreatic Ductal Adenocarcinoma (PDAC). Int J Mol Sci 2024; 25:10991. [PMID: 39456771 PMCID: PMC11506938 DOI: 10.3390/ijms252010991] [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: 09/25/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Associations between inflammation and cancer were first discovered approximately 160 years ago by Rudolf Virchow, who observed that tumors were infiltrated with inflammatory cells, and defined inflammation as a pathological condition. Inflammation has now emerged as one of the key mediators in oncogenesis and tumor progression, including pancreatic ductal adenocarcinoma (PDAC). However, the role of inflammatory processes in cancers is complicated and controversial, and the detailed regulatory mechanisms are still unclear. This review elucidates the dynamic interplay between inflammation and immune regulation, microenvironment alteration, metabolic reprogramming, and microbiome risk factors in PDAC, committing to exploring a deeper understanding of the role of crucial inflammatory pathways and molecules for providing insights into therapeutic strategies.
Collapse
Affiliation(s)
- Liu Yang
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (L.Y.); (S.Q.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China;
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Shuangying Qiao
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (L.Y.); (S.Q.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China;
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ge Zhang
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China;
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Aiping Lu
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (L.Y.); (S.Q.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China;
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Fangfei Li
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (L.Y.); (S.Q.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China;
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
35
|
Zhu Q, Zeng S, Yang J, Zhuo J, Wang P, Wen S, Fang C. Plectin-1-targeted recognition for enhancing comprehensive therapy in pancreatic ductal adenocarcinoma. NANOSCALE 2024; 16:18584-18596. [PMID: 39291372 DOI: 10.1039/d4nr01587h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses a formidable challenge due to its aggressive nature and poor prognosis. Gemcitabine (Gem), a primary therapeutic option, functions by inhibiting DNA synthesis and promoting apoptosis, thereby impeding the progression of PDAC. However, Gem is hindered by suboptimal pharmacokinetics and efficacy. In response to these challenges, we have developed a nanoparticle (NP) designed for specific recognition of plectin-1 in PDAC cell membranes. The NPs encapsulate Gem while demonstrating pH-responsive drug release characteristics in the acidic tumor microenvironment. This targeted approach enhances local drug delivery while alleviating concerns about systemic toxicity. Furthermore, the NPs are enriched with indocyanine green (ICG), renowned for its strong photothermal effects, thereby further enhancing therapeutic outcomes. This study presents an innovative therapeutic strategy for PDAC based on a plectin-1-targeted recognition delivery approach. The approach is applied to enhance chemotherapy, combined with photothermal therapy (PTT), inducing apoptosis in PDAC cell lines and improving the pharmacokinetics of Gem. In conclusion, the delivery strategy based on plectin-1-targeted recognition shows promising preclinical prospects for enhancing therapeutic efficacy in PDAC, offering valuable insights for future clinical applications.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Silue Zeng
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Junying Yang
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Jiaming Zhuo
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Peifeng Wang
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Sai Wen
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Chihua Fang
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| |
Collapse
|
36
|
Doctor A, Laube M, Meister S, Kiss OC, Kopka K, Hauser S, Pietzsch J. Combined PET Radiotracer Approach Reveals Insights into Stromal Cell-Induced Metabolic Changes in Pancreatic Cancer In Vitro and In Vivo. Cancers (Basel) 2024; 16:3393. [PMID: 39410013 PMCID: PMC11475921 DOI: 10.3390/cancers16193393] [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: 08/09/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objective Pancreatic stellate cells (PSCs) in pancreatic adenocarcinoma (PDAC) are producing extracellular matrix, which promotes the formation of a dense fibrotic microenvironment. This makes PDAC a highly heterogeneous tumor-stroma-driven entity, associated with reduced perfusion, limited oxygen supply, high interstitial fluid pressure, and limited bioavailability of therapeutic agents. Methods In this study, spheroid and tumor xenograft models of human PSCs and PanC-1 cells were characterized radiopharmacologically using a combined positron emission tomography (PET) radiotracer approach. [18F]FDG, [18F]FMISO, and [18F]FAPI-74 were employed to monitor metabolic activity, hypoxic metabolic state, and functional expression of fibroblast activation protein alpha (FAPα), a marker of activated PSCs. Results In vitro, PanC-1 and multi-cellular tumor spheroids demonstrated comparable glucose uptake and hypoxia, whereas FAPα expression was significantly higher in PSC spheroids. In vivo, glucose uptake as well as the transition to hypoxia were comparable in PanC-1 and multi-cellular xenograft models. In mice injected with PSCs, FAPα expression decreased over a period of four weeks post-injection, which was attributed to the successive death of PSCs. In contrast, FAPα expression increased in both PanC-1 and multi-cellular xenograft models over time due to invasion of mouse fibroblasts. Conclusion The presented models are suitable for subsequently characterizing stromal cell-induced metabolic changes in tumors using noninvasive molecular imaging techniques.
Collapse
Affiliation(s)
- Alina Doctor
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany; (A.D.); (M.L.); (S.M.); (K.K.); (S.H.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
| | - Markus Laube
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany; (A.D.); (M.L.); (S.M.); (K.K.); (S.H.)
| | - Sebastian Meister
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany; (A.D.); (M.L.); (S.M.); (K.K.); (S.H.)
| | - Oliver C. Kiss
- Department of Targetry, Target Chemistry and Radiopharmacy, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany;
| | - Klaus Kopka
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany; (A.D.); (M.L.); (S.M.); (K.K.); (S.H.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
- National Center for Tumor Diseases (NCT) Dresden, Partner Site Dresden, University Cancer Center (UCC), Fetscherstraße 74, 01307 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany; (A.D.); (M.L.); (S.M.); (K.K.); (S.H.)
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany; (A.D.); (M.L.); (S.M.); (K.K.); (S.H.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
| |
Collapse
|
37
|
Tao J, Gu Y, Zhang Z, Weng G, Liu Y, Ren J, Shi Y, Qiu J, Wang Y, Su D, Wang R, Fu Y, Liu T, Ye L, Luo W, Chen H, Yang G, Cao Z, Huang H, Xiao J, Ren B, You L, Zhang T, Zhao Y. CALB2 drives pancreatic cancer metastasis through inflammatory reprogramming of the tumor microenvironment. J Exp Clin Cancer Res 2024; 43:277. [PMID: 39358777 PMCID: PMC11448066 DOI: 10.1186/s13046-024-03201-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Early dissemination to distant organs accounts for the dismal prognosis of patients with pancreatic ductal adenocarcinoma (PDAC). Chronic, dysregulated, persistent and unresolved inflammation provides a preferred tumor microenvironment (TME) for tumorigenesis, development, and metastasis. A better understanding of the key regulators that maintain inflammatory TME and the development of predictive biomarkers to identify patients who are most likely to benefit from specific inflammatory-targeted therapies is crucial for advancing personalized cancer treatment. METHODS This study identified cell-specific expression of CALB2 in human PDAC through single-cell RNA sequencing analysis and assessed its clinicopathological correlations in tissue microarray using multi-color immunofluorescence. Co-culture systems containing cancer-associated fibroblasts (CAFs) and patient-derived organoids (PDOs) in vitro and in vivo were employed to elucidate the effects of CALB2-activated CAFs on PDAC malignancy. Furthermore, CUT&RUN assays, luciferase reporter assays, RNA sequencing, and gain- or loss-of-function assays were used to unravel the molecular mechanisms of CALB2-mediated inflammatory reprogramming and metastasis. Additionally, immunocompetent KPC organoid allograft models were constructed to evaluate CALB2-induced immunosuppression and PDAC metastasis, as well as the efficacy of inflammation-targeted therapy. RESULTS CALB2 was highly expressed both in CAFs and cancer cells and correlated with an unfavorable prognosis and immunosuppressive TME in PDAC patients. CALB2 collaborated with hypoxia to activate an inflammatory fibroblast phenotype, which promoted PDAC cell migration and PDO growth in vitro and in vivo. In turn, CALB2-activated CAFs upregulated CALB2 expression in cancer cells through IL6-STAT3 signaling-mediated direct transcription. In cancer cells, CALB2 further activated Ca2+-CXCL14 inflammatory axis to facilitate PDAC metastatic outgrowth and immunosuppression. Genetic or pharmaceutical inhibition of CXCL14 significantly suppressed CALB2-mediated metastatic colonization of PDAC cells in vivo and extended mouse survival. CONCLUSIONS These findings identify CALB2 as a key regulator of inflammatory reprogramming to promote PDAC metastatic progression. Combination therapy with αCXCL14 monoclonal antibody and gemcitabine emerges as a promising strategy to suppress distant metastasis and improve survival outcomes in PDAC with CALB2 overexpression.
Collapse
Affiliation(s)
- Jinxin Tao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yani Gu
- Institute of Clinical Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Translational Medicine Center, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Zeyu Zhang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Guihu Weng
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yueze Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jie Ren
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yanan Shi
- Biomedical Engineering Facility of National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jiangdong Qiu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yuanyang Wang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Dan Su
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ruobing Wang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Department of General Surgery, China‑Japan Friendship Hospital, Beijing, 100029, China
| | - Yifan Fu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Tao Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Liyuan Ye
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Wenhao Luo
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hao Chen
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Gang Yang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhe Cao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hua Huang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jianchun Xiao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Bo Ren
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Lei You
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Taiping Zhang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Yupei Zhao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
38
|
Li X, Hou W, Xiao C, Yang H, Zhao C, Cao D. Panoramic tumor microenvironment in pancreatic ductal adenocarcinoma. Cell Oncol (Dordr) 2024; 47:1561-1578. [PMID: 39008192 DOI: 10.1007/s13402-024-00970-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is notorious for its resistance to various treatment modalities. The genetic heterogeneity of PDAC, coupled with the presence of a desmoplastic stroma within the tumor microenvironment (TME), contributes to an unfavorable prognosis. The mechanisms and consequences of interactions among different cell types, along with spatial variations influencing cellular function, potentially play a role in the pathogenesis of PDAC. Understanding the diverse compositions of the TME and elucidating the functions of microscopic neighborhoods may contribute to understanding the immune microenvironment status in pancreatic cancer. As we delve into the spatial biology of the microscopic neighborhoods within the TME, aiding in deciphering the factors that orchestrate this intricate ecosystem. This overview delineates the fundamental constituents and the structural arrangement of the PDAC microenvironment, highlighting their impact on cancer cell biology.
Collapse
Affiliation(s)
- Xiaoying Li
- Department of Abdominal Oncology, Division of Abdominal Tumor Multimodality Treatment, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610017, People's Republic of China
| | - Wanting Hou
- Department of Abdominal Oncology, Division of Abdominal Tumor Multimodality Treatment, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610017, People's Republic of China
| | - Chaoxin Xiao
- State Key Laboratory of Biotherapy and Cancer Center, West China HospitaL, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, Sichuan, 610017, People's Republic of China
| | - Heqi Yang
- Department of Abdominal Oncology, Division of Abdominal Tumor Multimodality Treatment, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610017, People's Republic of China
| | - Chengjian Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China HospitaL, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, Sichuan, 610017, People's Republic of China
| | - Dan Cao
- Department of Abdominal Oncology, Division of Abdominal Tumor Multimodality Treatment, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610017, People's Republic of China.
| |
Collapse
|
39
|
Rauth S, Malafa M, Ponnusamy MP, Batra SK. Emerging Trends in Gastrointestinal Cancer Targeted Therapies: Harnessing Tumor Microenvironment, Immune Factors, and Metabolomics Insights. Gastroenterology 2024; 167:867-884. [PMID: 38759843 PMCID: PMC11793124 DOI: 10.1053/j.gastro.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/23/2024] [Accepted: 05/01/2024] [Indexed: 05/19/2024]
Abstract
Gastrointestinal (GI) cancers are the leading cause of new cancer cases and cancer-related deaths worldwide. The treatment strategies for patients with GI tumors have focused on oncogenic molecular profiles associated with tumor cells. Recent evidence has demonstrated that the tumor cell functions are modulated by its microenvironment, compromising fibroblasts, extracellular matrices, microbiome, immune cells, and the enteric nervous system. Along with the tumor microenvironment components, alterations in key metabolic pathways have emerged as a hallmark of tumor cells. From these perspectives, this review will highlight the functions of different cellular components of the GI tumor microenvironment and their implications for treatment. Furthermore, we discuss the major metabolic reprogramming in GI tumor cells and how understanding metabolic rewiring could lead to new therapeutic strategies. Finally, we briefly summarize the targeted agents currently being studied in GI cancers. Understanding the complex interplay between tumor cell-intrinsic and -extrinsic factors during tumor progression is critical for developing new therapeutic strategies.
Collapse
Affiliation(s)
- Sanchita Rauth
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska
| | - Mokenge Malafa
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center at Omaha, Omaha, Nebraska.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center at Omaha, Omaha, Nebraska.
| |
Collapse
|
40
|
Helen H, Gunawan MC, Halim P, Dinata MR, Ahmed A, Dalimunthe A, Marianne M, Ribeiro RIMDA, Hasibuan PAZ, Nurkolis F, Hey-Hawkins E, Park MN, Harahap U, Kim SH, Kim B, Syahputra RA. Flavonoids as modulators of miRNA expression in pancreatic cancer: Pathways, Mechanisms, And Therapeutic Potential. Biomed Pharmacother 2024; 179:117347. [PMID: 39241569 DOI: 10.1016/j.biopha.2024.117347] [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: 06/14/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/09/2024] Open
Abstract
Pancreatic cancer (PC) is a complex malignancy, distinguished by its aggressive characteristics and unfavorable prognosis. Recent developments in understanding the molecular foundations of this disease have brought attention to the noteworthy involvement of microRNAs (miRNAs) in disease development, advancement, and treatment resistance. The anticancer capabilities of flavonoids, which are a wide range of phytochemicals present in fruits and vegetables, have attracted considerable interest because of their ability to regulate miRNA expression. This review provides the effects of flavonoids on miRNA expression in PC, explains the underlying processes, and explores the possible therapeutic benefits of flavonoid-based therapies. Flavonoids inhibit PC cell proliferation, induce apoptosis, and enhance chemosensitivity via the modulation of miRNAs involved in carcinogenesis. Additionally, this review emphasizes the significance of certain miRNAs as targets of flavonoid action. These miRNAs have a role in regulating important signaling pathways such as the phosphoinositide-3-kinase-protein kinase B/Protein kinase B (Akt), mitogen activated protein kinase (MAPK), Janus kinase/signal transducers and activators of transcription (JAK/STAT), and Wnt/β-catenin pathways. This review aims to consolidate current knowledge on the interaction between flavonoids and miRNAs in PC, providing a comprehensive analysis of how flavonoid-mediated modulation of miRNA expression could influence cancer progression and therapy. It highlights the use of flavonoid nanoformulations to enhance stability, increase absorption, and maximize anti-PC activity, improving patient outcomes. The review calls for further research to optimize the use of flavonoid nanoformulations in clinical trials, leading to innovative treatment strategies and more effective approaches for PC.
Collapse
Affiliation(s)
- Helen Helen
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Mega Carensia Gunawan
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Princella Halim
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Muhammad Riza Dinata
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Amer Ahmed
- Department of Bioscience, Biotechnology and Environment, University of Bari, Bari, Italy
| | - Aminah Dalimunthe
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Marianne Marianne
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Rosy Iara Maciel De Azambuja Ribeiro
- Experimental Pathology Laboratory, Federal University of São João del Rei (UFSJ), 400, Sebastião Gonçalves Coelho, Chanadour, Divinópolis 35501-296, MG, Brazil
| | | | - Fahrul Nurkolis
- Biological Sciences, Faculty of Sciences and Technology, UIN Sunan Kalijaga, Yogyakarta, Indonesia
| | - Evamarie Hey-Hawkins
- Leipzig University, Faculty of Chemistry and Mineralogy, Centre for Biotechnology and Biomedicine (BBZ), Institute of Bioanalytical Chemistry, Deutscher Platz 5, 04103 Leipzig, Germany
| | - Moon Nyeo Park
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea; College of Korean Medicine, Kyung Hee University, Hoegidong, Dongdaemungu, Seoul, 05253, Republic of Korea
| | - Urip Harahap
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Sung-Hoon Kim
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Bonglee Kim
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea; College of Korean Medicine, Kyung Hee University, Hoegidong, Dongdaemungu, Seoul, 05253, Republic of Korea
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia.
| |
Collapse
|
41
|
Yang F, Jiang N, Li XY, Qi XS, Tian ZB, Guo YJ. Construction and validation of a pancreatic cancer prognostic model based on genes related to the hypoxic tumor microenvironment. World J Gastroenterol 2024; 30:4057-4070. [PMID: 39351249 PMCID: PMC11439118 DOI: 10.3748/wjg.v30.i36.4057] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/22/2024] [Accepted: 09/05/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Pancreatic cancer is one of the most lethal malignancies, characterized by poor prognosis and low survival rates. Traditional prognostic factors for pancreatic cancer offer inadequate predictive accuracy, often failing to capture the complexity of the disease. The hypoxic tumor microenvironment has been recognized as a significant factor influencing cancer progression and resistance to treatment. This study aims to develop a prognostic model based on key hypoxia-related molecules to enhance prediction accuracy for patient outcomes and to guide more effective treatment strategies in pancreatic cancer. AIM To develop and validate a prognostic model for predicting outcomes in patients with pancreatic cancer using key hypoxia-related molecules. METHODS This pancreatic cancer prognostic model was developed based on the expression levels of the hypoxia-associated genes CAPN2, PLAU, and CCNA2. The results were validated in an independent dataset. This study also examined the correlations between the model risk score and various clinical features, components of the immune microenvironment, chemotherapeutic drug sensitivity, and metabolism-related pathways. Real-time quantitative PCR verification was conducted to confirm the differential expression of the target genes in hypoxic and normal pancreatic cancer cell lines. RESULTS The prognostic model demonstrated significant predictive value, with the risk score showing a strong correlation with clinical features: It was significantly associated with tumor grade (G) (b P < 0.01), moderately associated with tumor stage (T) (a P < 0.05), and significantly correlated with residual tumor (R) status (b P < 0.01). There was also a significant negative correlation between the risk score and the half-maximal inhibitory concentration of some chemotherapeutic drugs. Furthermore, the risk score was linked to the enrichment of metabolism-related pathways in pancreatic cancer. CONCLUSION The prognostic model based on hypoxia-related genes effectively predicts pancreatic cancer outcomes with improved accuracy over traditional factors and can guide treatment selection based on risk assessment.
Collapse
Affiliation(s)
- Fan Yang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Na Jiang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Xiao-Yu Li
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Xing-Si Qi
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Zi-Bin Tian
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Ying-Jie Guo
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| |
Collapse
|
42
|
Hendley AM, Ashe S, Urano A, Ng M, Phu TA, Peng XL, Luan C, Finger AM, Jang GH, Kerper NR, Berrios DI, Jin D, Lee J, Riahi IR, Gbenedio OM, Chung C, Roose JP, Yeh JJ, Gallinger S, Biankin AV, O'Kane GM, Ntranos V, Chang DK, Dawson DW, Kim GE, Weaver VM, Raffai RL, Hebrok M. nSMase2-mediated exosome secretion shapes the tumor microenvironment to immunologically support pancreatic cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614610. [PMID: 39399775 PMCID: PMC11468832 DOI: 10.1101/2024.09.23.614610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
The pleiotropic roles of nSMase2-generated ceramide include regulation of intracellular ceramide signaling and exosome biogenesis. We investigated the effects of eliminating nSMase2 on early and advanced PDA, including its influence on the microenvironment. Employing the KPC mouse model of pancreatic cancer, we demonstrate that pancreatic epithelial nSMase2 ablation reduces neoplasia and promotes a PDA subtype switch from aggressive basal-like to classical. nSMase2 elimination prolongs survival of KPC mice, hinders vasculature development, and fosters a robust immune response. nSMase2 loss leads to recruitment of cytotoxic T cells, N1-like neutrophils, and abundant infiltration of anti-tumorigenic macrophages in the pancreatic preneoplastic microenvironment. Mechanistically, we demonstrate that nSMase2-expressing PDA cell small extracellular vesicles (sEVs) reduce survival of KPC mice; PDA cell sEVs generated independently of nSMase2 prolong survival of KPC mice and reprogram macrophages to a proinflammatory phenotype. Collectively, our study highlights previously unappreciated opposing roles for exosomes, based on biogenesis pathway, during PDA progression. Graphical abstract
Collapse
|
43
|
Li MM, Zhang Y, Sun F, Huai MX, Zhang FY, Pan JX, Qu CY, Shen F, Li ZH, Xu LM. Photodynamic Therapy Using RGD-Functionalized Quantum Dots Elicit a Potent Immune Response in a Syngeneic Mouse Model of Pancreatic Cancer. Int J Nanomedicine 2024; 19:9487-9502. [PMID: 39290860 PMCID: PMC11406538 DOI: 10.2147/ijn.s479123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024] Open
Abstract
Purpose Photodynamic therapy (PDT) induces anti-tumor immune responses by triggering immunogenic cell death in tumor cells. Previously, we demonstrated that novel QDs-RGD nanoparticles exhibited high efficiency as photosensitizers in the treatment of pancreatic cancer. However, the underlying mechanism of the anti-tumor immune effects induced by the photosensitizer remains unknown. This study assessed the anticancer immune effect of QDs-RGD, as well as the conventional photosensitizer chlorine derivative, YLG-1, for comparison, against pancreatic cancer in support of superior therapeutic efficacy. Methods The pancreatic cancer cell line, Panc02, was used for in vitro studies. C57BL/6 mice bearing pancreatic cancer cell-derived xenografts were generated for in vivo studies to assess the anti-tumor effects of QDs-RGD-PDT and YLG-1-PDT. The immunostimulatory ability of both photosensitizers was examined by measuring the expression of damage-associated molecular patterns (DAMP), such as calreticulin (CRT), assessing dendritic cell (DC) maturation, and analyzing cytokine expression. The specific immunity of QDs-RGD and YLG-1-PDT on distant tumor were determined by combining PDT with anti-CTLA-4 antibody. Results QDs-RGD-PDT and YLG-1-PDT significantly inhibited pancreatic cancer cell growth in a dose- and time-dependent manner. While both photosensitizers significantly promoted CRT release, DC maturation, and interferon γ (IFN-γ) and tumor necrosis factor α (TNF-α) expression, QDs-RGD exerted a stronger immunostimulatory effect than YLG-1. Combination treatment with QDs-RGD and CTLA-4 blockade was able to significantly inhibit the growth of distant tumors. Conclusion QDs-RGD is a novel and effective PDT strategy for treating pancreatic tumors by inducing anti-tumor immune responses.
Collapse
Affiliation(s)
- Ming-Ming Li
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Yi Zhang
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Fang Sun
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Man-Xiu Huai
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Fei-Yu Zhang
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Jia-Xing Pan
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Chun-Ying Qu
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Feng Shen
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Zheng-Hong Li
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Lei-Ming Xu
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| |
Collapse
|
44
|
Ogunleye AO, Gayen N, Rauth S, Marimuthu S, Nimmakayala RK, Alsafwani ZW, Cox JL, Batra SK, Ponnusamy MP. PAF1/HIF1α axis rewires the glycolytic metabolism to fuel aggressiveness of pancreatic cancer. Cancer Metab 2024; 12:26. [PMID: 39242538 PMCID: PMC11380429 DOI: 10.1186/s40170-024-00354-2] [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: 02/09/2024] [Accepted: 08/19/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND PAF1/PD2 deregulation contributes to tumorigenesis, drug resistance, and cancer stem cell maintenance in Pancreatic Cancer (PC). Recent studies demonstrate that metabolic reprogramming plays a role in PC progression, but the mechanism is poorly understood. Here, we focused on examining the role of PAF1/PD2 in the metabolic rewiring of PC. METHODS Cell lines were transfected with shRNAs to knockdown PAF1/PD2. Metabolic genes regulated by PAF1/PD2 were identified by qPCR/western blot, and metabolic assays were performed. Immunoprecipitations/ChIP were performed to identify PAF1/PD2 protein partners and confirm PAF1/HIF1α sub-complex binding to LDHA. RESULTS PAF1 and LDHA showed progressively increased expression in human pancreatic tumor sections. Aerobic glycolysis genes were downregulated in PAF1-depleted PC cells. Metabolic assays indicated a decreased lactate production and glucose uptake in knockdown cells. Furthermore, PAF1/PD2 depletion showed a reduced glycolytic rate and increased oxidative phosphorylation by ECAR and OCR analysis. Interestingly, we identified that HIF1α interacts and co-localizes with PAF1, specifically in PC cells. We also observed that the PAF1/PD2-HIF1α complex binds to the LDHA promoter to regulate its expression, reprogramming the metabolism to utilize the aerobic glycolysis pathway preferentially. CONCLUSION Overall, the results indicate that PAF1/PD2 rewires PC metabolism by interacting with HIF1α to regulate the expression of LDHA.
Collapse
Affiliation(s)
- Ayoola O Ogunleye
- Department of Biochemistry and Molecular Biology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, NE, 985870, USA
| | - Neelanjana Gayen
- Department of Biochemistry and Molecular Biology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, NE, 985870, USA
| | - Sanchita Rauth
- Department of Biochemistry and Molecular Biology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, NE, 985870, USA
| | - Saravanakumar Marimuthu
- Department of Biochemistry and Molecular Biology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, NE, 985870, USA
| | - Rama Krishna Nimmakayala
- Department of Biochemistry and Molecular Biology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, NE, 985870, USA
| | - Zahraa W Alsafwani
- Department of Biochemistry and Molecular Biology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, NE, 985870, USA
| | - Jesse L Cox
- Department of Pathology and Microbiology, University of Nebraska Medical Center at Omaha, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, NE, 985870, USA.
- Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center at Omaha, Omaha, NE, USA.
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, NE, 985870, USA.
- Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center at Omaha, Omaha, NE, USA.
| |
Collapse
|
45
|
Do CTP, Prochnau JY, Dominguez A, Wang P, Rao MK. The Road Ahead in Pancreatic Cancer: Emerging Trends and Therapeutic Prospects. Biomedicines 2024; 12:1979. [PMID: 39335494 PMCID: PMC11428787 DOI: 10.3390/biomedicines12091979] [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: 08/01/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
This review explores the challenges and emerging trends in pancreatic cancer therapy. In particular, we focus on the tumor microenvironment and the potential of immunotherapy for pancreatic cancer. Pancreatic ductal adenocarcinoma, characterized by its dense stromal architecture, presents unique challenges for effective treatment. Recent advancements have emphasized the role of the tumor microenvironment in therapeutic resistance and disease progression. We discuss novel strategies targeting the desmoplastic barrier and immunosuppressive cells to enhance immune cell infiltration and activation. Recent clinical trials, particularly those involving novel immunotherapeutic agents and tumor vaccines, are examined to understand their efficacy and limitations. Our analysis reveals that combining immunotherapy with chemotherapy, radiation therapy, or drugs targeting epigenetic processes shows promise, improving overall survival rates and response to treatment. For instance, trials utilizing checkpoint inhibitors in combination with standard chemotherapies have extended disease-free survival by up to 6 months compared to chemotherapy alone. Importantly, vaccines targeting specific tumor neoantigens have shown the potential to increase patient survival. However, these approaches also face significant challenges, including overcoming the immunosuppressive tumor microenvironment and enhancing the delivery and efficacy of therapeutic agents. By providing an overview of both the promising results and the obstacles encountered, this review aims to highlight ongoing efforts to refine immunotherapy approaches for better patient outcomes.
Collapse
Affiliation(s)
- Chris T P Do
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jack Y Prochnau
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Angel Dominguez
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Pei Wang
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Manjeet K Rao
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
46
|
Swain S, Narayan RK, Mishra PR. Unraveling the interplay: exploring signaling pathways in pancreatic cancer in the context of pancreatic embryogenesis. Front Cell Dev Biol 2024; 12:1461278. [PMID: 39239563 PMCID: PMC11374643 DOI: 10.3389/fcell.2024.1461278] [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: 07/08/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024] Open
Abstract
Pancreatic cancer continues to be a deadly disease because of its delayed diagnosis and aggressive tumor biology. Oncogenes and risk factors are being reported to influence the signaling pathways involved in pancreatic embryogenesis leading to pancreatic cancer genesis. Although studies using rodent models have yielded insightful information, the scarcity of human pancreatic tissue has made it difficult to comprehend how the human pancreas develops. Transcription factors like IPF1/PDX1, HLXB9, PBX1, MEIS, Islet-1, and signaling pathways, including Hedgehog, TGF-β, and Notch, are directing pancreatic organogenesis. Any derangements in the above pathways may lead to pancreatic cancer. TP53: and CDKN2A are tumor suppressor genes, and the mutations in TP53 and somatic loss of CDKN2A are the drivers of pancreatic cancer. This review clarifies the complex signaling mechanism involved in pancreatic cancer, the same signaling pathways in pancreas development, the current therapeutic approach targeting signaling molecules, and the mechanism of action of risk factors in promoting pancreatic cancer.
Collapse
|
47
|
Zhou W, Deng X, Liu L, Yuan Y, Meng X, Ma J. PELI1 overexpression contributes to pancreatic cancer progression through upregulating ubiquitination-mediated INPP5J degradation. Cell Signal 2024; 120:111194. [PMID: 38685520 DOI: 10.1016/j.cellsig.2024.111194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 04/12/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
Inositol Polyphosphate-5-Phosphatase J (INPP5J), a 5-phosphatase, has been identified as a tumor suppressor in several types of cancer. However, its role in pancreatic cancer (PC) is unknown. We found that the INPP5J expression was markedly lower in PC tissues (n = 50) compared to paired adjacent non-tumor tissues, and the lower INPP5J expression was relevant to a worse prognosis of PC patients. We thus proposed that INPP5J might inhibit PC progression and conducted gain-of- and loss-of-function experiments to test our hypothesis. Our results showed that overexpression of INPP5J inhibited cell proliferation, invasion, migration, and xenografted tumor of PC cells. INPP5J silencing showed the opposite effect. Pellino E3 Ubiquitin Protein Ligase 1 (PELI1) is one of the ubiquitin ligases known to promote ubiquitination of its downstream targets. We found that PELI1 could interact with INPP5J and promote the ubiquitination and degradation of INPP5J. PELI1 overexpression enhanced malignant behaviors of PC cells. However, INPP5J overexpression restored the alterations caused by PELI1 overexpression. In conclusion, the results suggest that the decreased INPP5J expression, caused by PELI1 through ubiquitination, may promote PC progression. The PELI1-INPP5J axis represents a potential therapeutic targetable node for PC.
Collapse
Affiliation(s)
- Wenyang Zhou
- Department of Pathology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xin Deng
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Liling Liu
- Department of Pathology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yifeng Yuan
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiangpeng Meng
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Jia Ma
- Department of Gastroenterology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
48
|
Molon AC, Heguedusch D, Nunes FD, Cecatto RB, Dos Santos Franco AL, de Oliveira Rodini Pegoraro C, Rodrigues MFSD. A 5-ALA mediated photodynamic therapy increases natural killer cytotoxicity against oral squamous cell carcinoma cell lines. JOURNAL OF BIOPHOTONICS 2024:e202400176. [PMID: 39023037 DOI: 10.1002/jbio.202400176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/03/2024] [Accepted: 06/25/2024] [Indexed: 07/20/2024]
Abstract
Oral squamous cell carcinoma (OSCC) constitutes over 90% of oral cancers, known for its aggressiveness and poor prognosis. Photodynamic therapy (PDT) has emerged as a promising adjuvant therapy and is linked to immunogenic cell death, activating innate and adaptive anti-tumor responses. Natural Killer (NK) cells, key players in malignant cell elimination, have not been extensively studied in PDT. This study evaluates whether PDT increases OSCC cell lines' susceptibility to NK cell cytotoxicity. PDT, using 5-aminolevulinic acid (5-ALA) and LED irradiation, was applied to Ca1 and Luc4 cell lines. Results showed a dose-dependent viability decrease post-PDT. Gene expression analysis revealed upregulation of NK cell-activating ligands (ULBP1-4, MICA/B) and decreased MHC class I expression in Ca1, suggesting increased NK cell susceptibility. Enhanced NK cell cytotoxicity was confirmed in Ca1 but not in Luc4 cells. These findings indicate that PDT may enhance NK cell-mediated cytotoxicity in OSCC, offering potential for improved treatment strategies.
Collapse
Affiliation(s)
- Angela Cristina Molon
- Post Graduate Program in Biophotonics Applied to Health Sciences, Nove de Julho University, São Paulo, Brazil
| | - Daniele Heguedusch
- Department of Stomatology, Discipline of Oral and Maxillofacial Pathology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Fabio Daumas Nunes
- Department of Stomatology, Discipline of Oral and Maxillofacial Pathology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Rebeca Boltes Cecatto
- Post Graduate Program in Biophotonics Applied to Health Sciences, Nove de Julho University, São Paulo, Brazil
| | | | | | | |
Collapse
|
49
|
Pan L, Qin Z, Zhou Q, Zheng P, Li H, Zhou X, Qin Y. Unlocking the Therapeutic Potential: Harnessing miR-125a-5p To Enhance Autophagy and Apoptosis in Pancreatic Cancer through Targeting STAT3. J Cancer 2024; 15:4955-4968. [PMID: 39132168 PMCID: PMC11310883 DOI: 10.7150/jca.97102] [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: 04/08/2024] [Accepted: 06/26/2024] [Indexed: 08/13/2024] Open
Abstract
Objectives: miR-125a-5p's role in various cancers has been recognized, yet its specific function in pancreatic cancer (PCa) demands further exploration. This study aimed to reveal the potential function of miR-125a-5p in PCa. Methods: With publicly available databases, we explored the expression pattern and prognostic relevance of miR-125a-5p and STAT3 in PCa. We measured miR-125a-5p levels in PCa tissues, plasma and cell lines using RT-qPCR. To assess functional effects, PANC-1 cells were transfected with miR-125a-5p mimics and inhibitors, as well as siRNA-STAT3 and STAT3 vectors. Cell proliferation was estimated using Cell Counting Kit-8, while autophagy and apoptosis were examined by transmission electron microscopy and TUNEL assay, respectively. Western blot analysis was also performed to detect proteins associated with autophagy and apoptosis. The regulatory relationship of miR-125a-5p on STAT3 was verified using a dual luciferase reporter assay. The influence of miR-125a-5p on tumor development was evaluated in xenograft models. Results: Decreased expression of miR-125a-5p was found in PCa samples, and low expression of miR-125a-5p was associated with a poorer prognosis in PCa patients. Functional assays indicated miR-125a-5p suppressed cell growth while enhancing apoptosis and autophagy in PCa cells. STAT3 represents a specific target of miR-125a-5p, inhibiting STAT3 reversed the inhibitory effect of overexposed miR-125a-5p. Additionally, miR-125a-5p significantly restrained tumor development in mice. Conclusions: miR-125a-5p functions as a tumor suppressor in PCa by targeting STAT3, thereby inducing autophagy and apoptosis. Its regulatory role underscores its potential as a valuable biomarker for PCa diagnosis and therapy, warranting further clinical investigation.
Collapse
Affiliation(s)
- Lujuan Pan
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong Province, China
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Province, China
- Key Laboratory of Tumor Molecular Pathology of Baise, Baise, Guangxi Province, China
| | - Zongshuai Qin
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong Province, China
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Province, China
| | - Qinghong Zhou
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Province, China
| | - Pin Zheng
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Province, China
| | - Hua Li
- Department of General Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Province, China
| | - Xihan Zhou
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Province, China
| | - Yueqiu Qin
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong Province, China
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Province, China
| |
Collapse
|
50
|
Saadh MJ, Mustafa MA, Malathi H, Ahluwalia G, Kaur S, Al-Dulaimi MAAH, Alubiady MHS, Zain Al-Abdeen SH, Shakier HG, Ali MS, Ahmad I, Abosaoda MK. Targeting the pancreatic tumor microenvironment by plant-derived products and their nanoformulations. Med Oncol 2024; 41:201. [PMID: 39001987 DOI: 10.1007/s12032-024-02443-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 06/27/2024] [Indexed: 07/15/2024]
Abstract
Pancreatic cancer remains a significant health issue with limited treatment options. The tumor stroma, a complex environment made up of different cells and proteins, plays a crucial role in tumor growth and chemoresistance. Targeting tumor stroma, consisting of diverse non-tumor cells such as fibroblasts, extracellular matrix (ECM), immune cells, and also pre-vascular cells is encouraging for remodeling solid cancers, such as pancreatic cancer. Remodeling the stroma of pancreas tumors can be suggested as a strategy for reducing resistance to chemo/immunotherapy. Several studies have shown that phytochemicals from plants can affect the tumor environment and have anti-cancer properties. By targeting key pathways involved in stromal activation, phytochemicals may disrupt communication between the tumor and stroma and make tumor cells more sensitive to different treatments. Additionally, phytochemicals have immunomodulatory and anti-angiogenic properties, all of which contribute to their potential in treating pancreatic cancer. This review will provide a detailed look at how phytochemicals impact the tumor stroma and their effects on pancreatic tumor growth, spread, and response to treatment. It will also explore the potential of combining phytochemicals with other treatment options like chemotherapy, immunotherapy, and radiation.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Mohammed Ahmed Mustafa
- Department of Medical Laboratory Technology, University of Imam Jaafar AL-Sadiq, Baghdad, Iraq
| | - H Malathi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Gunveen Ahluwalia
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, 303012, Rajasthan, India
| | - Sumeet Kaur
- Department of Applied Sciences, Chandigarh Engineering Colleges, Chandigarh Group of Colleges, Jhanjeri, 140307, Mohali, Punjab, India
| | | | | | | | | | | | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| | - Munther Kadhim Abosaoda
- College of Pharmacy, The Islamic University, Najaf, Iraq
- College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|