1
|
Dreyer SB, Beer P, Hingorani SR, Biankin AV. Improving outcomes of patients with pancreatic cancer. Nat Rev Clin Oncol 2025; 22:439-456. [PMID: 40329051 DOI: 10.1038/s41571-025-01019-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2025] [Indexed: 05/08/2025]
Abstract
Research studies aimed at improving the outcomes of patients with pancreatic ductal adenocarcinoma (PDAC) have brought about limited progress, and in clinical practice, the optimized use of surgery, chemotherapy and supportive care have led to modest improvements in survival that have probably reached a plateau. As a result, PDAC is expected to be the second leading cause of cancer-related death in Western societies within a decade. The development of therapeutic advances in PDAC has been challenging owing to a lack of actionable molecular targets, a typically immunosuppressive microenvironment, and a disease course characterized by rapid progression and clinical deterioration. Yet, the progress in our understanding of PDAC and identification of novel therapeutic opportunities over the past few years is leading to a strong sense of optimism in the field. In this Perspective, we address the aforementioned challenges, including biological aspects of PDAC that make this malignancy particularly difficult to treat. We explore specific areas with potential for therapeutic advances, including targeting mutant KRAS, novel strategies to harness the antitumour immune response and approaches to early detection, and propose mechanisms to improve clinical trial design and to overcome various community and institutional barriers to progress.
Collapse
Affiliation(s)
- Stephan B Dreyer
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK
- West of Scotland Hepato-Biliary and Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
- Department of Hepatobiliary Surgery, Royal Liverpool University Hospital, Liverpool, UK
| | - Philip Beer
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK
- Hull York Medical School, University of York, York, UK
| | - Sunil R Hingorani
- Department of Internal Medicine, Division of Hemotology/Oncology, University of Nebraska Medical Center, Omaha, NE, USA
- Pancreatic Cancer Center of Excellence, University of Nebraska Medical Center, Omaha, NE, USA
| | - Andrew V Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK.
- West of Scotland Hepato-Biliary and Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK.
| |
Collapse
|
2
|
Liu D, Liu L, Zhao X, Zhang X, Chen X, Che X, Wu G. A comprehensive review on targeting diverse immune cells for anticancer therapy: Beyond immune checkpoint inhibitors. Crit Rev Oncol Hematol 2025; 210:104702. [PMID: 40122356 DOI: 10.1016/j.critrevonc.2025.104702] [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: 02/10/2025] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 03/25/2025] Open
Abstract
Although immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, primary resistance and acquired resistance continue to limit their efficacy for many patients. To address resistance and enhance the anti-tumor activity within the tumor immune microenvironment (TIME), numerous therapeutic strategies targeting both innate and adaptive immune cells have emerged. These include combination therapies with ICIs, chimeric antigen receptor T-cell (CAR-T), chimeric antigen receptor macrophages (CAR-Ms) or chimeric antigen receptor natural killer cell (CAR-NK) therapy, colony stimulating factor 1 receptor (CSF1R) inhibitors, dendritic cell (DC) vaccines, toll-like receptor (TLR) agonists, cytokine therapies, and chemokine inhibition. These approaches underscore the significant potential of the TIME in cancer treatment. This article provides a comprehensive and up-to-date review of the mechanisms of action of various innate and adaptive immune cells within the TIME, as well as the therapeutic strategies targeting each immune cell type, aiming to deepen the understanding of their therapeutic potential.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lei Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xinming Zhao
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaoman Zhang
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaochi Chen
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Xiangyu Che
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Guangzhen Wu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
3
|
Toghraie FS, Bayat M, Hosseini MS, Ramezani A. Tumor-infiltrating myeloid cells; mechanisms, functional significance, and targeting in cancer therapy. Cell Oncol (Dordr) 2025; 48:559-590. [PMID: 39998754 PMCID: PMC12119771 DOI: 10.1007/s13402-025-01051-y] [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] [Accepted: 02/20/2025] [Indexed: 02/27/2025] Open
Abstract
Tumor-infiltrating myeloid cells (TIMs), which encompass tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), myeloid-derived suppressor cells (MDSCs), and tumor-associated dendritic cells (TADCs), are of great importance in tumor microenvironment (TME) and are integral to both pro- and anti-tumor immunity. Nevertheless, the phenotypic heterogeneity and functional plasticity of TIMs have posed challenges in fully understanding their complexity roles within the TME. Emerging evidence suggested that the presence of TIMs is frequently linked to prevention of cancer treatment and improvement of patient outcomes and survival. Given their pivotal function in the TME, TIMs have recently been recognized as critical targets for therapeutic approaches aimed at augmenting immunostimulatory myeloid cell populations while depleting or modifying those that are immunosuppressive. This review will explore the important properties of TIMs related to immunity, angiogenesis, and metastasis. We will also document the latest therapeutic strategies targeting TIMs in preclinical and clinical settings. Our objective is to illustrate the potential of TIMs as immunological targets that may improve the outcomes of existing cancer treatments.
Collapse
Affiliation(s)
- Fatemeh Sadat Toghraie
- Institute of Biotechnology, Faculty of the Environment and Natural Sciences, Brandenburg University of Technology, Cottbus, Germany
| | - Maryam Bayat
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsa Sadat Hosseini
- Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Amin Ramezani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
4
|
Zhang J, Zhang Q, Lin G, Wang Y, Li J, Wang P, Qi J, Liang Y, He S, Gong Y, Feng N, Wang Y, Ma Y, Zhang M, Shi Y, Li X, Ci W, Zhou L. Single-Cell Analysis Reveals that Vitamin C Inhibits Bone Metastasis of Renal Cancer via Cell Cycle Arrest and Microenvironment Remodeling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e01011. [PMID: 40433925 DOI: 10.1002/advs.202501011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/24/2025] [Indexed: 05/29/2025]
Abstract
Bone metastasis is the second most common site of distant metastatic spread in renal cell carcinoma (RCC) patients, significantly contributing to cancer-related mortality. The metastatic process is driven by both intrinsic tumor cell properties, such as cancer stem cell-like characteristics, and the bone microenvironment. Understanding the complex interactions between cancer cells and their niche is crucial for identifying therapeutic targets to eliminate metastasis-initiating cells and prevent overt metastasis. In this study, a murine bone metastasis model is developed using renal cancer cells derived from fibrin gel-induced 3D tumor spheres, which exhibit stem-like phenotypes. It is found that a stable form of vitamin C, L-ascorbic acid 2-phosphate sesquimagnesium (APM), significantly inhibits the growth of renal cancer stem-like cells in vitro and the progression of RCC bone metastasis in vivo. Single-cell RNA sequencing revealed that APM induces cell cycle arrest and reduces the metastatic potential of cancer cells. Furthermore, APM remodels the tumor microenvironment by suppressing osteoclast differentiation and neutrophil recruitment. Combining APM with a CXCR2 antagonist, SB225002, further inhibits bone metastasis progression. This study provides a high-resolution profile of vitamin C's antitumor effects in the bone metastatic microenvironment and supports the rationale for clinical trials of vitamin C in bone metastatic RCC.
Collapse
Affiliation(s)
- Jianye Zhang
- Department of Urology, Peking University First Hospital, Beijing, 100034, P. R. China
- Institute of Urology, Peking University, Beijing, 100034, P. R. China
- National Urological Cancer Center, Beijing, 100034, P. R. China
| | - Qi Zhang
- China National Center for Bioinformation, Beijing, 100101, P. R. China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Gang Lin
- Department of Thoracic Surgery, Peking University First Hospital, Peking University, Beijing, 100034, P. R. China
| | - Ying Wang
- China National Center for Bioinformation, Beijing, 100101, P. R. China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Juan Li
- China National Center for Bioinformation, Beijing, 100101, P. R. China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Ping Wang
- China National Center for Bioinformation, Beijing, 100101, P. R. China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Jie Qi
- China National Center for Bioinformation, Beijing, 100101, P. R. China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Yuan Liang
- China National Center for Bioinformation, Beijing, 100101, P. R. China
| | - Shiming He
- Department of Urology, Peking University First Hospital, Beijing, 100034, P. R. China
- Institute of Urology, Peking University, Beijing, 100034, P. R. China
- National Urological Cancer Center, Beijing, 100034, P. R. China
| | - Yanqing Gong
- Department of Urology, Peking University First Hospital, Beijing, 100034, P. R. China
- Institute of Urology, Peking University, Beijing, 100034, P. R. China
- National Urological Cancer Center, Beijing, 100034, P. R. China
| | - Ninghan Feng
- Department of Urology, Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, 214002, P. R. China
| | - Yang Wang
- Department of Urology, Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, 214002, P. R. China
| | - Yuanyuan Ma
- Animal Center, Peking University First Hospital, Beijing, 100034, P. R. China
| | - Mei Zhang
- China National Center for Bioinformation, Beijing, 100101, P. R. China
| | - Yue Shi
- China National Center for Bioinformation, Beijing, 100101, P. R. China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Xuesong Li
- Department of Urology, Peking University First Hospital, Beijing, 100034, P. R. China
- Institute of Urology, Peking University, Beijing, 100034, P. R. China
- National Urological Cancer Center, Beijing, 100034, P. R. China
| | - Weimin Ci
- China National Center for Bioinformation, Beijing, 100101, P. R. China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- Department of Urology, Chinese PLA General Hospital, Beijing, 100039, P. R. China
| | - Liqun Zhou
- Department of Urology, Peking University First Hospital, Beijing, 100034, P. R. China
- Institute of Urology, Peking University, Beijing, 100034, P. R. China
- National Urological Cancer Center, Beijing, 100034, P. R. China
- Department of Urology, The First Affiliated Hospital of Henan University, Kaifeng, 475001, P. R. China
| |
Collapse
|
5
|
Kwak JW, Houghton AM. Targeting neutrophils for cancer therapy. Nat Rev Drug Discov 2025:10.1038/s41573-025-01210-8. [PMID: 40374764 DOI: 10.1038/s41573-025-01210-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2025] [Indexed: 05/18/2025]
Abstract
Neutrophils are among the most abundant immune cell types in the tumour microenvironment and have been associated with poor outcomes across multiple cancer types. Yet despite mounting evidence of their role in tumour progression, therapeutic strategies targeting neutrophils have only recently gained attention and remain limited in scope. This is probably due to the increasing number of distinct neutrophil subtypes identified in cancer and the limited understanding of the mechanisms by which these subsets influence tumour progression and immune evasion. In this Review, we discuss the spectrum of neutrophil subtypes - including those with antitumour activity - and their potential to polarize towards tumour-suppressive phenotypes. We explore the molecular pathways and effector functions by which neutrophils modulate cancer progression, with an emphasis on identifying tractable therapeutic targets. Finally, we examine emerging clinical trials aimed at modulating neutrophil lineages and consider their implications for patient outcomes.
Collapse
Affiliation(s)
- Jeff W Kwak
- Translational Science and Therapeutics Division and Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - A McGarry Houghton
- Translational Science and Therapeutics Division and Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
6
|
Roscigno G, Jacobs S, Toledo B, Borea R, Russo G, Pepe F, Serrano MJ, Calabrò V, Troncone G, Giovannoni R, Giovannetti E, Malapelle U. The potential application of stroma modulation in targeting tumor cells: focus on pancreatic cancer and breast cancer models. Semin Cancer Biol 2025:S1044-579X(25)00060-4. [PMID: 40373890 DOI: 10.1016/j.semcancer.2025.05.003] [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: 01/20/2025] [Revised: 04/08/2025] [Accepted: 05/04/2025] [Indexed: 05/17/2025]
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer development and spreading being considered as "the dark side of the tumor". Within this term tumor cells, immune components, supporting cells, extracellular matrix and a myriad of bioactive molecules that synergistically promote tumor development and therapeutic resistance, are included. Recent findings revealed the profound impacts of TME on cancer development, serving as physical support, critical mediator and biodynamic matrix in cancer evolution, immune modulation, and treatment outcomes. TME targeting strategies built on vasculature, immune checkpoints, and immuno-cell therapies, have paved the way for revolutionary clinical interventions. On this basis, the relevance of pre-clinical and clinical investigations has rapidly become fundamental for implementing novel therapeutical strategies breaking cell-cell and cell -mediators' interactions between TME components and tumor cells. This review summarizes the key players in the breast and pancreatic TME, elucidating the intricate interactions among cancer cells and their essential role for cancer progression and therapeutic resistance. Different tumors such breast and pancreatic cancer have both different and similar stroma features, that might affect therapeutic strategies. Therefore, this review aims to comprehensively evaluate recent findings for refining breast and pancreatic cancer therapies and improve patient prognoses by exploiting the TME's complexity in the next future.
Collapse
Affiliation(s)
- Giuseppina Roscigno
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy.
| | - Sacha Jacobs
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.
| | - Belen Toledo
- Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén E-23071, Spain.
| | - Roberto Borea
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| | - Gianluca Russo
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Francesco Pepe
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Maria Jose Serrano
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy; GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and Cancer Interception Group, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain.
| | - Viola Calabrò
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Roberto Giovannoni
- Department of Biology, Genetic Unit, University of Pisa, Via Derna 1, 56126 Pisa, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, the Netherlands; Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy.
| | - Umberto Malapelle
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| |
Collapse
|
7
|
Zhang M, Liu C, Tu J, Tang M, Ashrafizadeh M, Nabavi N, Sethi G, Zhao P, Liu S. Advances in cancer immunotherapy: historical perspectives, current developments, and future directions. Mol Cancer 2025; 24:136. [PMID: 40336045 PMCID: PMC12057291 DOI: 10.1186/s12943-025-02305-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 03/15/2025] [Indexed: 05/09/2025] Open
Abstract
Cancer immunotherapy, encompassing both experimental and standard-of-care therapies, has emerged as a promising approach to harnessing the immune system for tumor suppression. Experimental strategies, including novel immunotherapies and preclinical models, are actively being explored, while established treatments, such as immune checkpoint inhibitors (ICIs), are widely implemented in clinical settings. This comprehensive review examines the historical evolution, underlying mechanisms, and diverse strategies of cancer immunotherapy, highlighting both its clinical applications and ongoing preclinical advancements. The review delves into the essential components of anticancer immunity, including dendritic cell activation, T cell priming, and immune surveillance, while addressing the challenges posed by immune evasion mechanisms. Key immunotherapeutic strategies, such as cancer vaccines, oncolytic viruses, adoptive cell transfer, and ICIs, are discussed in detail. Additionally, the role of nanotechnology, cytokines, chemokines, and adjuvants in enhancing the precision and efficacy of immunotherapies were explored. Combination therapies, particularly those integrating immunotherapy with radiotherapy or chemotherapy, exhibit synergistic potential but necessitate careful management to reduce side effects. Emerging factors influencing immunotherapy outcomes, including tumor heterogeneity, gut microbiota composition, and genomic and epigenetic modifications, are also examined. Furthermore, the molecular mechanisms underlying immune evasion and therapeutic resistance are analyzed, with a focus on the contributions of noncoding RNAs and epigenetic alterations, along with innovative intervention strategies. This review emphasizes recent preclinical and clinical advancements, with particular attention to biomarker-driven approaches aimed at optimizing patient prognosis. Challenges such as immunotherapy-related toxicity, limited efficacy in solid tumors, and production constraints are highlighted as critical areas for future research. Advancements in personalized therapies and novel delivery systems are proposed as avenues to enhance treatment effectiveness and accessibility. By incorporating insights from multiple disciplines, this review aims to deepen the understanding and application of cancer immunotherapy, ultimately fostering more effective and widely accessible therapeutic solutions.
Collapse
Affiliation(s)
- Meiyin Zhang
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Chaojun Liu
- Department of Breast Surgery, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University; People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Jing Tu
- Department of Pulmonary and Critical Care Medicine, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Min Tang
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China
| | - Milad Ashrafizadeh
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, V8 V 1P7, Canada
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research (N2CR) Yong Loo Lin, School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| | - Peiqing Zhao
- Translational Medicine Center, Zibo Central Hospital Affiliated to Binzhou Medical University, No. 54 Communist Youth League Road, Zibo, China.
| | - Shijian Liu
- Department of General Medicine, The 2nd Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Harbin, 150081, China.
| |
Collapse
|
8
|
Kaplan Z, Prezioso E, Jain A, Lavu H, Yeo CJ, Bowne WB, Nevler A. Clinical Implications of Mismatch Repair Deficiency in Pancreatic Ductal Adenocarcinoma. Cancer Med 2025; 14:e70960. [PMID: 40366030 PMCID: PMC12076359 DOI: 10.1002/cam4.70960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Pancreatic cancer is a highly aggressive and lethal disease, characterized by a limited response to chemotherapy and overall poor prognosis. Pancreatic cancers with a distinct mismatch repair deficiency, although relatively rare, have been shown to be associated with markedly better outcomes in comparison. Furthermore, whereas pancreatic cancers are generally unresponsive to current immunotherapy, this specific group of tumors has been shown to have a notable susceptibility to immune checkpoint inhibitors. AIMS In this review, we aim to summarize the relevant literature regarding mismatch-repair associated pancreatic cancers, the impacted biological mechanisms, and the resulting vulnerabilities for potential opportunistic immunotherapeutic treatment approaches. We will also review the current clinical studies assessing survival outcomes of mismatch repair deficient pancreatic cancers and ongoing clinical trials in this emerging field. RESULTS AND CONCLUSIONS Patients with dMMR/MSI-H pancreatic cancers harbor a distinct phenotype that has increased immune activation, greater responsiveness to immune checkpoint inhibitor therapy and better overall survival when compared to other pancreatic cancers. Although this molecular subtype makes up a small minority of cases, emerging data suggest immunotherapy may offer benefit to these patients.
Collapse
Affiliation(s)
- Zachary Kaplan
- Sidney Kimmel Medical CollegePhiladelphiaPennsylvaniaUSA
| | | | - Aditi Jain
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Harish Lavu
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Charles J. Yeo
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Wilbur B. Bowne
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Avinoam Nevler
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
9
|
Rys RN, Calcinotto A. Senescent neutrophils: a hidden role in cancer progression. Trends Cell Biol 2025; 35:399-411. [PMID: 39362804 DOI: 10.1016/j.tcb.2024.09.001] [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: 06/03/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 10/05/2024]
Abstract
Neutrophils have recently received increased attention in cancer because they contribute to all stages of cancer. Neutrophils are so far considered to have a short half-life. However, a growing body of literature has shown that tumor-associated neutrophils (TANs) acquire a prolonged lifespan. This review discusses recent work surrounding the mechanisms by which neutrophils can persist in the tumor microenvironment (TME). It also highlights different scenarios for therapeutic targeting of protumorigenic neutrophils, supporting the idea that, in tumors, inhibition of neutrophil recruitment is not sufficient because these cells can persist and remain hidden from current interventions. Hence, the elimination of long-lived neutrophils should be pursued to increase the efficacy of standard therapy.
Collapse
Affiliation(s)
- Ryan N Rys
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Arianna Calcinotto
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland.
| |
Collapse
|
10
|
Tao M, Liu W, Chen J, Liu R, Zou J, Yu B, Wang C, Huang M, Chen Q, Zhang Z, Chen Z, Sun H, Zhou C, Tan S, Zheng Y, Wang H. Transcriptome Landscape of Cancer-Associated Fibroblasts in Human PDAC. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415196. [PMID: 40019403 PMCID: PMC12120754 DOI: 10.1002/advs.202415196] [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] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/27/2025] [Indexed: 03/01/2025]
Abstract
Cancer-associated fibroblasts (CAFs) play a crucial role in the progression of pancreatic ductal adenocarcinoma (PDAC). Here, integrated single-cell RNA sequencing analysis is utilized to comprehensively map CAFs in the human PDAC tumor microenvironment (TME). Normal fibroblasts (NFs) and nine distinct CAF subtypes are identified including newly identified CAF subtypes, CDCP1+FTL+ CAFs, transitional CAFs (tCAFs), interferon simulated genes (ISG)+ myofibroblastic CAFs (myCAFs), and proliferative CAFs (pCAFs). CDCP1+FTL+ CAFs, pCAFs, and ISG+ myCAFs are associated with unfavorable clinical outcomes. CDCP1+FTL+ CAFs exhibit enhanced glycolysis and iron metabolism, resisting ferroptosis. The antigen-presenting CAFs (apCAFs) show high heterogeneity, consisting of multiple subtypes expressing distinct immune cell signatures. The CAF subtypes display differentiation plasticity, transitioning from early normal-like CAFs (nCAFs) to inflammatory CAFs (iCAFs) and myCAFs, ultimately leading to more invasive pCAFs. AP-1 family members FOS and JUN regulate the malignant phenotype conversion of NFs to nCAFs, while transforming growth factor-β (TGFβ) and interferon-γ (IFNγ) signals trigger the interconversion between classic myCAFs and iCAFs, respectively. A close interaction between CAFs and myeloid cells (especially neutrophils) is further observed in PDAC-TME, mainly mediated by CXCR4-CXCL12 chemotaxis. This work depicts a detailed CAF map and its dynamic interconvertible shift, providing important insights for combined targeted CAFs therapy.
Collapse
Affiliation(s)
- Mengyu Tao
- Department of OncologyShanghai General HospitalShanghai Jiaotong University School of MedicineShanghai200800P. R. China
| | - Wenting Liu
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Jianhua Chen
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Rujiao Liu
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Jianling Zou
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Bo Yu
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Chenchen Wang
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Mingzhu Huang
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Qingjian Chen
- Department of OncologyShanghai General HospitalShanghai Jiaotong University School of MedicineShanghai200800P. R. China
| | - Zhe Zhang
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Zhiyu Chen
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Haoyu Sun
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Department of ImmunologySchool of Basic Medical SciencesFudan UniversityShanghai200032China
| | - Cheng Zhou
- Department of Radiation OncologyNanfang HospitalSouthern Medical UniversityGuangzhou510515P. R. China
| | - Shuguang Tan
- The Second Affiliated HospitalZhejiang University School of MedicineHangzhou310009China
| | - Yuxuan Zheng
- Human Phenome InstituteMinhang HosptialFudan UniversityShanghai201203P. R. China
| | - Hongxia Wang
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| |
Collapse
|
11
|
Zhao Z, Cutmore LC, Baleeiro RB, Hartlebury JJ, Brown N, Chard-Dunmall L, Lemoine N, Wang Y, Marshall JF. The Combination of Oncolytic Virus and Antibody Blockade of TGF-β Enhances the Efficacy of αvβ6-Targeting CAR T Cells Against Pancreatic Cancer in an Immunocompetent Model. Cancers (Basel) 2025; 17:1534. [PMID: 40361460 PMCID: PMC12070938 DOI: 10.3390/cancers17091534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/24/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES CAR T cell therapy, as a rapidly advancing immuno-oncology modality, has achieved significant success in the treatment of leukaemia and lymphoma. However, its application in solid tumours remains limited. The challenges include the heterogeneity of tumours, local immunosuppression, poor trafficking and infiltration, life-threatening toxicity and the lack of precise representative immunocompetent research models. Considering its typically dense and immunosuppressive tumour microenvironment (TME) and early metastasis, pancreatic ductal adenocarcinoma (PDAC) was employed as a model to address the challenges that hinder CAR T cell therapies against solid tumours and to expand immunotherapeutic options for advanced disease. METHODS A novel murine A20FMDV2 (A20) CAR T cell targeting integrin αvβ6 (mA20CART) was developed, demonstrating efficient and specific on-target cytotoxicity. The mA20CART cell as a monotherapy for orthotopic pancreatic cancer in an immunocompetent model demonstrated modest efficacy. Therefore, a novel triple therapy regimen, combining mA20CART cells with oncolytic vaccinia virus encoding IL-21 and a TGF-β-blocking antibody was evaluated in vivo. RESULTS The triple therapy improved overall survival, improved the safety profile of the CAR T cell therapy, attenuated metastasis and enhanced T cell infiltration. Notably, the potency of mA20CART was dependent on IL-2 supplementation. CONCLUSIONS This study presents an αvβ6-targeting murine CAR T cell, offering a novel approach to developing CAR T cell technologies for solid tumours and a potential adjuvant therapy for pancreatic cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yaohe Wang
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (Z.Z.); (L.C.C.); (R.B.B.); (J.J.H.); (N.B.); (L.C.-D.); (N.L.)
| | - John F. Marshall
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (Z.Z.); (L.C.C.); (R.B.B.); (J.J.H.); (N.B.); (L.C.-D.); (N.L.)
| |
Collapse
|
12
|
Cai W, Fan T, Xiao C, Deng Z, Liu Y, Li C, He J. Neutrophils in cancer: At the crucial crossroads of anti-tumor and pro-tumor. Cancer Commun (Lond) 2025. [PMID: 40296668 DOI: 10.1002/cac2.70027] [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: 09/19/2024] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025] Open
Abstract
Neutrophils are important components of the immune system and play a key role in defending against pathogenic infections and responding to inflammatory cues, including cancer. Their dysregulation indicates potential disease risk factors. However, their functional importance in disease progression has often been underestimated due to their short half-life, especially as there is limited information on the role of intratumoral neutrophils. Recent studies on their prominent role in cancer have led to a paradigm shift in our understanding of the functional diversity of neutrophils. These studies highlight that neutrophils have emerged as key components of the tumor microenvironment, where they can play a dual role in promoting and suppressing cancer. Moreover, several approaches to therapeutically target neutrophils have emerged, and clinical trials are investigating their efficacy. In this review, we discussed the involvement of neutrophils in cancer initiation and progression. We summarized recent advances in therapeutic strategies targeting neutrophils and, most importantly, suggested future research directions that could facilitate the manipulation of neutrophils for therapeutic purposes in cancer patients.
Collapse
Affiliation(s)
- Wenpeng Cai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Yixiao Liu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| |
Collapse
|
13
|
Alsaafeen BH, Ali BR, Elkord E. Combinational therapeutic strategies to overcome resistance to immune checkpoint inhibitors. Front Immunol 2025; 16:1546717. [PMID: 40342408 PMCID: PMC12058545 DOI: 10.3389/fimmu.2025.1546717] [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: 12/17/2024] [Accepted: 03/31/2025] [Indexed: 05/11/2025] Open
Abstract
Over the past few years, immune checkpoint inhibitors resulted in magnificent and durable successes in treating cancer; however, only a minority of patients respond favorably to the treatment due to a broad-spectrum of tumor-intrinsic and tumor-extrinsic factors. With the recent insights gained into the mechanisms of resistance, combination treatment strategies to overcome the resistance and enhance the therapeutic potential of immune checkpoint inhibitors are emerging and showing promising results in both pre-clinical and clinical settings. This has been derived through multiple interconnected mechanisms such as enhancing tumor immunogenicity, improving neoantigen processing and presentation in addition to augmenting T cell infiltration and cytotoxic potentials. In the clinical settings, several avenues of combination treatments involving immune checkpoint inhibitors were associated with considerable improvement in the therapeutic outcome in terms of patient's survival and tumor growth control. This, in turn, increased the spectrum of cancer patients benefiting from the unprecedented and durable effects of immune checkpoint inhibitors leading to their adoption as a first-line treatment for certain cancers. Moreover, the significance of precision medicine in cancer immunotherapy and the unmet demand to develop more personalized predictive biomarkers and treatment strategies are also highlighted in this review.
Collapse
Affiliation(s)
- Besan H. Alsaafeen
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Bassam R. Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Eyad Elkord
- Department of Biosciences and Bioinformatics & Suzhou Municipal Key Lab of Biomedical Sciences and Translational Immunology, School of Science, Xi’an Jiaotong-Liverpool University, Suzhou, China
- College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
- Biomedical Research Center, School of Science, Engineering and Environment, University of Salford, Manchester, United Kingdom
| |
Collapse
|
14
|
Raymakers L, Passchier EM, Verdonschot MEL, Evers M, Chan C, Kuijpers KC, Raicu GM, Molenaar IQ, van Santvoort HC, Strijbis K, Intven MPW, Daamen LA, Leusen JHW, Olofsen PA. The Efficacy of Targeted Monoclonal IgA Antibodies Against Pancreatic Ductal Adenocarcinoma. Cells 2025; 14:632. [PMID: 40358156 PMCID: PMC12071589 DOI: 10.3390/cells14090632] [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: 03/20/2025] [Revised: 04/11/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
The efficacy of immunotherapy in pancreatic ductal adenocarcinoma (PDAC) remains limited. The tumor microenvironment (TME), characterized by the accumulation of suppressive myeloid cells including neutrophils, attributes to immunotherapy resistance in PDAC. IgA monoclonal antibodies (mAbs) can activate neutrophils to kill tumor cells; this can be further enhanced by blocking the myeloid immune checkpoint CD47. In this study, we investigated the potential of this therapeutic strategy for PDAC. We determined the expression of tumor-associated antigens (TAAs) on PDAC cell lines and fresh patient samples, and the results showed that the TAAs epithelial cell adhesion molecule (EpCAM), trophoblast cell surface antigen 2 (TROP2) and mucin-1 (MUC1), as well as CD47 were consistently expressed on PDAC. In line with this, we showed that IgA mAbs against EpCAM can activate neutrophils to lyse various PDAC cell lines and tumor cells, which can be augmented by addition of CD47 blockade. In addition, we observed that neutrophils were present in patient tumors and expressed the receptor for IgA. In conclusion, our results indicate that a combination of IgA mAb with CD47 blockade is a promising preclinical treatment strategy for PDAC, which merits further investigation.
Collapse
Affiliation(s)
- Léon Raymakers
- Center for Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (L.R.); (E.M.P.); (M.E.L.V.); (M.E.); (P.A.O.)
- Division of Imaging & Oncology, University Medical Center Utrecht Cancer Center, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (M.P.W.I.); (L.A.D.)
| | - Elsemieke M. Passchier
- Center for Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (L.R.); (E.M.P.); (M.E.L.V.); (M.E.); (P.A.O.)
| | - Meggy E. L. Verdonschot
- Center for Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (L.R.); (E.M.P.); (M.E.L.V.); (M.E.); (P.A.O.)
| | - Mitchell Evers
- Center for Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (L.R.); (E.M.P.); (M.E.L.V.); (M.E.); (P.A.O.)
| | - Chilam Chan
- Center for Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (L.R.); (E.M.P.); (M.E.L.V.); (M.E.); (P.A.O.)
| | - Karel C. Kuijpers
- Department of Pathology, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, St. Antonius Hospital Nieuwegein, Koekoekslaan 1, 3435 CM Nieuwegein, The Netherlands (G.M.R.)
| | - G. Mihaela Raicu
- Department of Pathology, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, St. Antonius Hospital Nieuwegein, Koekoekslaan 1, 3435 CM Nieuwegein, The Netherlands (G.M.R.)
| | - I. Quintus Molenaar
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (I.Q.M.); (H.C.v.S.)
| | - Hjalmar C. van Santvoort
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (I.Q.M.); (H.C.v.S.)
| | - Karin Strijbis
- Department of Biomolecular Health Sciences, Division Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands;
| | - Martijn P. W. Intven
- Division of Imaging & Oncology, University Medical Center Utrecht Cancer Center, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (M.P.W.I.); (L.A.D.)
| | - Lois A. Daamen
- Division of Imaging & Oncology, University Medical Center Utrecht Cancer Center, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (M.P.W.I.); (L.A.D.)
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (I.Q.M.); (H.C.v.S.)
| | - Jeanette H. W. Leusen
- Center for Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (L.R.); (E.M.P.); (M.E.L.V.); (M.E.); (P.A.O.)
| | - Patricia A. Olofsen
- Center for Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (L.R.); (E.M.P.); (M.E.L.V.); (M.E.); (P.A.O.)
| |
Collapse
|
15
|
Cao L, Leclercq-Cohen G, Klein C, Sorrentino A, Bacac M. Mechanistic insights into resistance mechanisms to T cell engagers. Front Immunol 2025; 16:1583044. [PMID: 40330489 PMCID: PMC12053166 DOI: 10.3389/fimmu.2025.1583044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
T cell engagers (TCEs) represent a groundbreaking advancement in the treatment of B and plasma cell malignancies and are emerging as a promising therapeutic approach for the treatment of solid tumors. These molecules harness T cells to bind to and eliminate cancer cells, effectively bypassing the need for antigen-specific T cell recognition. Despite their established clinical efficacy, a subset of patients is either refractory to TCE treatment (e.g. primary resistance) or develops resistance during the course of TCE therapy (e.g. acquired or treatment-induced resistance). In this review we comprehensively describe the resistance mechanisms to TCEs, occurring in both preclinical models and clinical trials with a particular emphasis on cellular and molecular pathways underlying the resistance process. We classify these mechanisms into tumor intrinsic and tumor extrinsic ones. Tumor intrinsic mechanisms encompass changes within tumor cells that impact the T cell-mediated cytotoxicity, including tumor antigen loss, the expression of immune checkpoint inhibitory ligands and intracellular pathways that render tumor cells resistant to killing. Tumor extrinsic mechanisms involve factors external to tumor cells, including the presence of an immunosuppressive tumor microenvironment (TME) and reduced T cell functionality. We further propose actionable strategies to overcome resistance offering potential avenues for enhancing TCE efficacy in the clinic.
Collapse
Affiliation(s)
- Linlin Cao
- Roche Innovation Center, Zürich, Switzerland
| | | | | | | | | |
Collapse
|
16
|
Zhang F, Wang B, Wu M, Zhang L, Ji M. Current status of KRAS G12C inhibitors in NSCLC and the potential for combination with anti-PD-(L)1 therapy: a systematic review. Front Immunol 2025; 16:1509173. [PMID: 40303413 PMCID: PMC12037499 DOI: 10.3389/fimmu.2025.1509173] [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: 10/10/2024] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
In recent years, precision medicine for non-small cell lung cancer (NSCLC) has made significant strides, particularly with advancements in diagnostic and therapeutic technologies. Targeted 7therapies and Anti-PD-(L)1 Therapies have emerged as vital treatment options, yet KRAS mutations, especially KRAS G12C, have been historically difficult to address. Due to the unique activation mechanism of KRAS G12C has led to the development of specific inhibitors, such as AMG 510 and MRTX849, which show promising therapeutic potential. However, results from the CodeBreaK 200 Phase III trial indicated that AMG 510 did not significantly improve overall survival compared to docetaxel. Resistance after prolonged use of KRAS G12C inhibitors continues to pose a challenge, prompting interest in new drugs and combination strategies. KRAS mutations can impair tumor-infiltrating T cell function and create an immunosuppressive tumor microenvironment, making the combination of KRAS G12C inhibitors with anti-PD-(L)1 therapies particularly appealing. Preliminary data suggest these combinations may enhance both survival and quality of life, though safety concerns remain a barrier. Ongoing research is crucial to refine treatment regimens and identify suitable patient populations. This review focuses on the development of KRAS G12C inhibitors in monotherapy and combination therapies for NSCLC, discussing major clinical trials and future research directions.
Collapse
Affiliation(s)
| | | | | | | | - Mei Ji
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
17
|
Wahnou H, El Kebbaj R, Hba S, Ouadghiri Z, El Faqer O, Pinon A, Liagre B, Limami Y, Duval RE. Neutrophils and Neutrophil-Based Drug Delivery Systems in Anti-Cancer Therapy. Cancers (Basel) 2025; 17:1232. [PMID: 40227814 PMCID: PMC11988188 DOI: 10.3390/cancers17071232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/15/2025] Open
Abstract
Neutrophils, the most abundant white blood cells, play a dual role in cancer progression. While they can promote tumor growth, metastasis, and immune suppression, they also exhibit anti-tumorigenic properties by attacking cancer cells and enhancing immune responses. This review explores the complex interplay between neutrophils and the tumor microenvironment (TME), highlighting their ability to switch between pro- and anti-tumor phenotypes based on external stimuli. Pro-tumorigenic neutrophils facilitate tumor growth through mechanisms such as neutrophil extracellular traps (NETs), secretion of pro-inflammatory cytokines, and immune evasion strategies. They contribute to angiogenesis, tumor invasion, and metastasis by releasing vascular endothelial growth factor (VEGF) and matrix metalloproteinases (MMPs). Conversely, anti-tumor neutrophils enhance cytotoxicity by generating reactive oxygen species (ROS), promoting antibody-dependent cell-mediated cytotoxicity (ADCC), and activating other immune cells such as cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells. Recent advances in neutrophil-based drug delivery systems have harnessed their tumor-homing capabilities to improve targeted therapy. Neutrophil-mimicking nanoparticles and membrane-coated drug carriers offer enhanced drug accumulation in tumors, reduced systemic toxicity, and improved therapeutic outcomes. Additionally, strategies to modulate neutrophil activity, such as inhibiting their immunosuppressive functions or reprogramming them towards an anti-tumor phenotype, are emerging as promising approaches in cancer immunotherapy. Understanding neutrophil plasticity and their interactions with the TME provides new avenues for therapeutic interventions. Targeting neutrophil-mediated mechanisms could enhance existing cancer treatments and lead to the development of novel immunotherapies, ultimately improving patient survival and clinical outcomes.
Collapse
Affiliation(s)
- Hicham Wahnou
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
| | - Riad El Kebbaj
- Sciences and Engineering of Biomedicals, Biophysics and Health Laboratory, Higher Institute of Health Sciences, Hassan First University, Settat 26000, Morocco;
| | - Soufyane Hba
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (A.P.); (B.L.)
| | - Zaynab Ouadghiri
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
| | - Othman El Faqer
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
| | - Aline Pinon
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (A.P.); (B.L.)
| | - Bertrand Liagre
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (A.P.); (B.L.)
| | - Youness Limami
- Sciences and Engineering of Biomedicals, Biophysics and Health Laboratory, Higher Institute of Health Sciences, Hassan First University, Settat 26000, Morocco;
| | | |
Collapse
|
18
|
Jeong Y, Yoon SY, Jung SP, Nam SJ, Lee JE, Kim S. Inhibition of Interleukin-8/C-X-C Chemokine Receptor 2 Signaling Axis Prevents Tumor Growth and Metastasis in Triple-Negative Breast Cancer Cells. Pharmacology 2025:1-13. [PMID: 40188812 DOI: 10.1159/000545659] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 03/27/2025] [Indexed: 05/28/2025]
Abstract
INTRODUCTION Previously, we reported that interleukin-8 (IL-8) was associated with poor prognosis of basal-like breast cancer patients and has been identified as a pro-tumorigenic factor, facilitating cell invasion and migration. Here, we investigated the pharmacological impact of inhibitors targeting the chemokine receptors, C-X-C chemokine receptor 1 (CXCR1) and C-X-C chemokine receptor 2 (CXCR2), which are activated by IL-8. METHODS The survival rates of triple-negative breast cancer (TNBC) patients by IL-8 were analyzed by the Kaplan-Meier plotter. The levels of mRNA and protein expression were analyzed by real-time PCR and Western blotting. The alteration of apoptotic cell death-related proteins by SB225002 was analyzed by the Proteome Profiler Human Apoptosis Array. Cell growth was analyzed by MTT and colony-forming assay. Apoptosis and cell cycle were analyzed by fluorescence-activated cell sorting. RESULTS Aberrant IL-8 expression is involved with the prognosis of TNBC patients. Basal IL-8 levels are markedly elevated in TNBC cells compared to those in HER2+ and/or ER+ breast cancer cells. Furthermore, recombinant human IL-8 treatment enhanced cell invasiveness in TNBC cells. To counteract the tumor-promoting effects of IL-8, we assessed the therapeutic potential of CXCR1 and CXCR2 inhibitors. Notably, while reparixin, a CXCR1-specific inhibitor, exhibited no impact on cell viability, SB225002, a CXCR2-specific inhibitor, significantly reduced cell viability in a dose-dependent manner. There was a noticeable reduction in the levels of anti-apoptotic biomarkers, including Bcl-2, cIAP-1, cIAP-2, Survivin, XIAP, HIF-1α, and HO-1, following SB225002 treatment. Our findings indicate an increase in the apoptotic cell population with SB225002 treatment in TNBC cells. In xenograft models, SB225002 effectively diminished the metastatic potential of 4T1 cells, which are known to metastasize to the lung and liver. CONCLUSION Our results underscore the significant role of the IL-8/CXCR2 signaling axis in the metastasis of TNBC and suggest that CXCR2 inhibitors such as SB225002 may be promising therapeutic agents for TNBC patients.
Collapse
Affiliation(s)
- Yisun Jeong
- Department of Breast Cancer Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sun Young Yoon
- Department of Breast Cancer Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Seung Pil Jung
- Division of Breast and Endocrine Surgery, Department of Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seok Jin Nam
- Department of Breast Cancer Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jeong Eon Lee
- Department of Breast Cancer Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Sangmin Kim
- Department of Breast Cancer Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
19
|
McMorrow R, de Bruijn HS, Farina S, van Ardenne RJ, Que I, Mastroberardino PG, Robinson DJ, Mezzanotte L, Löwik CW. Combination of Bremachlorin PDT and Immune Checkpoint Inhibitor Anti-PD-1 Shows Response in Murine Immunological T-cell-High and T-cell-Low PDAC Models. Mol Cancer Ther 2025; 24:605-617. [PMID: 39704624 PMCID: PMC11962392 DOI: 10.1158/1535-7163.mct-23-0733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 04/12/2024] [Accepted: 12/17/2024] [Indexed: 12/21/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most challenging types of cancer with little or no response to immune checkpoint inhibitors (ICI). Photodynamic therapy (PDT) has been shown to ablate tumors and induce an immune response. In our study, we investigated the effect of PDT using the photosensitizer Bremachlorin, in its ability to reduce tumor burden and immunologically sensitize T-cell-high and T-cell-low murine PDAC tumors to the ICIs that blocks PD-1 immune checkpoint. In addition, we monitored the effect on survival and investigated if there was a response in PDT-treated and non-PDT-treated distant tumors. Our results showed that Bremachlorin PDT induces direct tumor killing that increased survival in both "hot" T-cell-high and "cold" T-cell-low PDAC tumors and that it can make T-cell-high tumors more sensitive to ICIs blocking PD-1. We found that T-cell-high tumor-bearing mice had an overall greater response to therapy than did T-cell-low tumor-bearing mice. One mouse with T-cell-high tumors exhibited complete tumor regression in both the treated and nontreated distant tumor 90 days after treatment. These results indicate that combining ICIs with Bremachlorin PDT could be a promising therapeutic intervention for enhancing PDAC's response to therapy.
Collapse
Affiliation(s)
- Roisin McMorrow
- Department of Radiology and Nuclear Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
- Department of Molecular Genetics, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Henriette S. de Bruijn
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Stefania Farina
- Department of Molecular Genetics, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Ruben J.L. van Ardenne
- Department of Radiology and Nuclear Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Ivo Que
- Department of Radiology and Nuclear Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
- Department of Molecular Genetics, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Pier G. Mastroberardino
- Department of Molecular Genetics, Erasmus Medical Centre, Rotterdam, the Netherlands
- IFOM-The FIRC Institute of Molecular Oncology, Milan, Italy
- Department of Life, Health, and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Dominic J. Robinson
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Laura Mezzanotte
- Department of Radiology and Nuclear Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
- Department of Molecular Genetics, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Clemens W.G.M. Löwik
- Department of Radiology and Nuclear Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
| |
Collapse
|
20
|
Zhang F, Jozani KA, Chakravarty A, Lin D, Hollinger A, Rajasekar S, Zhang B. Immune-Infiltrated Cancer Spheroid Model with Vascular Recirculation Reveals Temporally Dependent and Tissue-Specific Macrophage Recruitment. Adv Healthc Mater 2025; 14:e2402946. [PMID: 39962817 PMCID: PMC11973944 DOI: 10.1002/adhm.202402946] [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: 08/07/2024] [Revised: 01/20/2025] [Indexed: 04/08/2025]
Abstract
Immune cell infiltration in tumors has been reported to influence tumor progression and clinical outcomes. Considerable efforts have been made to understand interactions between tumors and the immune system. However, current models are either not comprehensive or limited to short-term studies. Recognizing thedynamic and long-term nature of tumor-immune interactions, an immune-infiltrated cancer spheroid model is developed by continuously perfusing and recirculating immune cells with gravity-driven flow through a tubular blood vessel adjacent to a cancer spheroid. Fibroblasts and pericytes are embedded in the gel matrix to support endothelial cells and enhance the vascular barrier. With continuous monocyte recirculation, monocyte adhesion, transendothelium migration, differentiation, and macrophage recruitment into breast carcinoma and hepatoma spheroids is successfully demonstrated over a week. The macrophage recruitment process is temporally dependent and tissue-specific, leading to the formation of cancer-macrophage heterospheroids. Elevated secretion of granulocyte-macrophage colony-stimulating factor (GM-CSF), which regulates monocyte recruitment and macrophage activation, is observed in the breast carcinoma model. Increased levels of Interleukin 6 (IL-6) and Interleukin 8 (IL-8) are detected, indicating a pro-inflammatory environment associated with tumor progression and metastasis. This platform provides a valuable framework for investigating immune cell infiltration and differentiation within the tumor microenvironment, supporting the advancement of cancer immunotherapies.
Collapse
Affiliation(s)
- Feng Zhang
- School of Biomedical EngineeringMcMaster UniversityHamiltonOntarioL8S 4L8Canada
| | - Kimia Asadi Jozani
- School of Biomedical EngineeringMcMaster UniversityHamiltonOntarioL8S 4L8Canada
| | - Anushree Chakravarty
- Department of Chemical EngineeringMcMaster UniversityHamiltonOntarioL8S 4L8Canada
| | - Dawn Lin
- Department of Chemical EngineeringMcMaster UniversityHamiltonOntarioL8S 4L8Canada
| | - Andrew Hollinger
- School of Biomedical EngineeringMcMaster UniversityHamiltonOntarioL8S 4L8Canada
| | - Shravanthi Rajasekar
- Department of Chemical EngineeringMcMaster UniversityHamiltonOntarioL8S 4L8Canada
| | - Boyang Zhang
- School of Biomedical EngineeringMcMaster UniversityHamiltonOntarioL8S 4L8Canada
- Department of Chemical EngineeringMcMaster UniversityHamiltonOntarioL8S 4L8Canada
- The Centre for Discovery in Cancer ResearchMcMaster University1280 Main Street WestHamiltonOntarioL8S 4M1Canada
| |
Collapse
|
21
|
Rahimizadeh P, Kim S, Yoon BJ, Jeong Y, Lim S, Jeon H, Lim HJ, Park SH, Park SI, Kong DH, Park JR, Song YB. Novel CXCR2 antibodies exhibit enhanced anti-tumor activity in pancreatic cancer. Biomed Pharmacother 2025; 185:117966. [PMID: 40058151 DOI: 10.1016/j.biopha.2025.117966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/22/2025] [Accepted: 03/05/2025] [Indexed: 03/23/2025] Open
Abstract
G protein-coupled receptors (GPCRs) are crucial in several physiological and pathological processes and are associated with numerous diseases across all therapeutic areas. Antibodies are well-established therapeutics with better selectivity, stability, and half-life than small molecules and peptides. CXC motif chemokine receptor 2 (CXCR2) and its ligands play functional roles in the progression and metastasis of tumors and activation and trafficking of inflammatory mediators. Several chemical-based antagonists that inhibit the CXCLs/CXCR2 signaling axis are under clinical study in cancer or inflammatory disease research; however, therapeutic antibodies have not yet been successfully established. In this study, we used phage display technology to identify single-chain variable fragment proteins that target the N-terminal domain of human CXCR2. Subsequently, we performed an enzyme-linked immunosorbent assay to validate the interaction of these IgG1 candidates and bio-layer interferometry to determine their affinity. The association of these antibodies, IgG18 and IgG56, with stable HEK293 cell lines expressing hCXCR2 and various cancer cell lines, including pancreatic cancer cells, was further analyzed. We found that IgG18 and IgG56 antibodies antagonized CXCL8-induced CXCR2 signaling, suppressed CXCL8-mediated cell proliferation and migration, and induced apoptosis in pancreatic cancer cells. In a xenograft model of pancreatic cancer, CXCR2 antibodies, IgG18 and IgG56, decreased tumor growth and induced apoptosis in vivo. Our results indicate that CXCR2 inhibitory antibodies attenuate tumor progression and may be potential candidates for anti-tumor therapeutics.
Collapse
Affiliation(s)
- Parastou Rahimizadeh
- Division of Research Center, Scripps Korea Antibody Institute, Chuncheon 24341, Republic of Korea
| | - Seheon Kim
- Division of Research Center, Scripps Korea Antibody Institute, Chuncheon 24341, Republic of Korea
| | - Byeong Jun Yoon
- Division of Research Center, Scripps Korea Antibody Institute, Chuncheon 24341, Republic of Korea
| | - Younsik Jeong
- Division of Research Center, Scripps Korea Antibody Institute, Chuncheon 24341, Republic of Korea
| | - Seoyoen Lim
- Division of Research Center, Scripps Korea Antibody Institute, Chuncheon 24341, Republic of Korea
| | - Hyeyoon Jeon
- Division of Research Center, Scripps Korea Antibody Institute, Chuncheon 24341, Republic of Korea
| | - Hyung Jin Lim
- Division of Research Center, Scripps Korea Antibody Institute, Chuncheon 24341, Republic of Korea
| | - Sang Ho Park
- Division of Research Center, Scripps Korea Antibody Institute, Chuncheon 24341, Republic of Korea
| | - Sang-In Park
- Division of Research Center, Scripps Korea Antibody Institute, Chuncheon 24341, Republic of Korea
| | - Deok-Hoon Kong
- Division of Research Center, Scripps Korea Antibody Institute, Chuncheon 24341, Republic of Korea
| | - Jeong-Ran Park
- Division of Research Center, Scripps Korea Antibody Institute, Chuncheon 24341, Republic of Korea
| | - Yong Bhum Song
- Division of Research Center, Scripps Korea Antibody Institute, Chuncheon 24341, Republic of Korea.
| |
Collapse
|
22
|
Hua Q, Li Z, Weng Y, Wu Y, Zheng L. Myeloid cells: key players in tumor microenvironments. Front Med 2025; 19:265-296. [PMID: 40048137 DOI: 10.1007/s11684-025-1124-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/16/2024] [Indexed: 05/04/2025]
Abstract
Cancer is the result of evolving crosstalk between neoplastic cell and its immune microenvironment. In recent years, immune therapeutics targeting T lymphocytes, such as immune checkpoint blockade (ICB) and CAR-T, have made significant progress in cancer treatment and validated targeting immune cells as a promising approach to fight human cancers. However, responsiveness to the current immune therapeutic agents is limited to only a small proportion of solid cancer patients. As major components of most solid tumors, myeloid cells played critical roles in regulating the initiation and sustentation of adaptive immunity, thus determining tumor progression as well as therapeutic responses. In this review, we discuss emerging data on the diverse functions of myeloid cells in tumor progression through their direct effects or interactions with other immune cells. We explain how different metabolic reprogramming impacts the characteristics and functions of tumor myeloid cells, and discuss recent progress in revealing different mechanisms-chemotaxis, proliferation, survival, and alternative sources-involved in the infiltration and accumulation of myeloid cells within tumors. Further understanding of the function and regulation of myeloid cells is important for the development of novel strategies for therapeutic exploitation in cancer.
Collapse
Affiliation(s)
- Qiaomin Hua
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Zhixiong Li
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yulan Weng
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yan Wu
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Limin Zheng
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
23
|
Walsh RM, Ambrose J, Jack JL, Eades AE, Bye BA, Tannus Ruckert M, Messaggio F, Olou AA, Chalise P, Pei D, VanSaun MN. Depletion of tumor-derived CXCL5 improves T cell infiltration and anti-PD-1 therapy response in an obese model of pancreatic cancer. J Immunother Cancer 2025; 13:e010057. [PMID: 40121029 PMCID: PMC11931939 DOI: 10.1136/jitc-2024-010057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND CXCR1/2 inhibitors are being implemented with immunotherapies in PDAC clinical trials. CXC-ligands are a family of cytokines responsible for stimulating these receptors; while typically secreted by activated immune cells, fibroblasts, and even adipocytes, they are also secreted by immune-evasive cancer cells. CXC-ligand release is known to occur in response to inflammatory stimuli. Adipose tissue is an endocrine organ and a source of inflammatory signaling peptides. Importantly, adipose-derived cytokines and chemokines are implicated as potential drivers of tumor cell immune evasion; cumulatively, these findings suggest that targeting CXC-ligands may be beneficial in the context of obesity. METHODS RNA-sequencing of human PDAC cell lines was used to assess influences of adipose conditioned media on the cancer cell transcriptome. The adipose-induced secretome of PDAC cells was validated with ELISA for induction of CXCL5 secretion. Human tissue data from CPTAC was used to correlate IL-1β and TNF expression with both CXCL5 mRNA and protein levels. CRISPR-Cas9 was used to knockout CXCL5 from a murine PDAC KPC cell line to assess orthotopic tumor studies in syngeneic, diet-induced obese mice. Flow cytometry and immunohistochemistry were used to compare the immune profiles between tumors with or without CXCL5. Mice-bearing CXCL5 competent or deficient tumors were monitored for differential tumor size in response to anti-PD-1 immune checkpoint blockade therapy. RESULTS Human adipose tissue conditioned media stimulates CXCL5 secretion from PDAC cells via either IL-1β or TNF; neutralization of both is required to significantly block the release of CXCL5 from tumor cells. Ablation of CXCL5 from tumors promoted an enriched immune phenotype with an unanticipatedly increased number of exhausted CD8 T cells. Application of anti-PD-1 treatment to control tumors failed to alter tumor growth, yet treatment of CXCL5-deficient tumors showed response by significantly diminished tumor mass. CONCLUSIONS In summary, our findings show that both TNF and IL-1β can stimulate CXCL5 release from PDAC cells in vitro, which correlates with expression in patient data. CXCL5 depletion in vivo alone is sufficient to promote T cell infiltration into tumors, increasing efficacy and requiring checkpoint blockade inhibition to alleviate tumor burden.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fanuel Messaggio
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Prabhakar Chalise
- Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
- The University of Kansas Cancer Center, Kansas City, Kansas, USA
| | - Dong Pei
- Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
- The University of Kansas Cancer Center, Kansas City, Kansas, USA
| | - Michael N VanSaun
- Cancer Biology, KUMC, Kansas City, Kansas, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
24
|
Santibanez JF. Myeloid-Derived Suppressor Cells: Implications in Cancer Immunology and Immunotherapy. FRONT BIOSCI-LANDMRK 2025; 30:25203. [PMID: 40152373 DOI: 10.31083/fbl25203] [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: 06/11/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 03/29/2025]
Abstract
Myeloid-derived suppressor cells (MDSCs) are believed to be key promoters of tumor development and are recognized as a hallmark of cancer cells' ability to evade the immune system evasion. MDSC levels often increase in peripheral blood and the tumor microenvironment (TME). These cells exert immunosuppressive functions, weakening the anticancer immune surveillance system, in part by repressing T-cell immunity. Moreover, MDSCs may promote tumor progression and interact with cancer cells, increasing MDSC expansion and favoring an immunotolerant TME. This review analyzes the primary roles of MDSCs in cancer and T-cell immunity, discusses the urgent need to develop effective MDSC-targeted therapies, and highlights the potential synergistic combination of MDSC targeting with chimeric antigen receptors and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Juan F Santibanez
- Group for Molecular Oncology, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia
- Integrative Center for Biology and Applied Chemistry (CIBQA), Bernardo O'Higgins University, 8370993 Santiago, Chile
| |
Collapse
|
25
|
Chambers CR, Watakul S, Schofield P, Howell AE, Zhu J, Tran AMH, Kuepper N, Reed DA, Murphy KJ, Channon LM, Pereira BA, Tyma VM, Lee V, Trpceski M, Henry J, Melenec P, Abdulkhalek L, Nobis M, Metcalf XL, Ritchie S, Cadell A, Stoehr J, Magenau A, Chacon-Fajardo D, Chitty JL, O’Connell S, Zaratzian A, Tayao M, Da Silva A, Lyons RJ, Goldstein LD, Dale A, Rookyard A, Connolly A, Crossett B, Tran YTH, Kaltzis P, Vennin C, Dinevska M, Australian Pancreatic Cancer Genome Initiative (APGI), Australian Pancreatic Cancer Matrix Atlas (APMA), Croucher DR, Samra J, Mittal A, Weatheritt RJ, Philp A, Del Monte-Nieto G, Zhang L, Enriquez RF, Cox TR, Shi YCC, Pinese M, Waddell N, Sim HW, Chtanova T, Wang Y, Joshua AM, Chantrill L, Evans TRJ, Gill AJ, Morton JP, Pajic M, Christ D, Herzog H, Timpson P, Herrmann D. Targeting the NPY/NPY1R signaling axis in mutant p53-dependent pancreatic cancer impairs metastasis. SCIENCE ADVANCES 2025; 11:eadq4416. [PMID: 40073121 PMCID: PMC11900870 DOI: 10.1126/sciadv.adq4416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 01/29/2025] [Indexed: 03/14/2025]
Abstract
Pancreatic cancer (PC) is a highly metastatic malignancy. More than 80% of patients with PC present with advanced-stage disease, preventing potentially curative surgery. The neuropeptide Y (NPY) system, best known for its role in controlling energy homeostasis, has also been shown to promote tumorigenesis in a range of cancer types, but its role in PC has yet to be explored. We show that expression of NPY and NPY1R are up-regulated in mouse PC models and human patients with PC. Moreover, using the genetically engineered, autochthonous KPR172HC mouse model of PC, we demonstrate that pancreas-specific and whole-body knockout of Npy1r significantly decreases metastasis to the liver. We identify that treatment with the NPY1R antagonist BIBO3304 significantly reduces KPR172HC migratory capacity on cell-derived matrices. Pharmacological NPY1R inhibition in an intrasplenic model of PC metastasis recapitulated the results of our genetic studies, with BIBO3304 significantly decreasing liver metastasis. Together, our results reveal that NPY/NPY1R signaling is a previously unidentified antimetastatic target in PC.
Collapse
Affiliation(s)
- Cecilia R. Chambers
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Supitchaya Watakul
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Peter Schofield
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Immune Biotherapies Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Anna E. Howell
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Jessie Zhu
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Alice M. H. Tran
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Nadia Kuepper
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Daniel A. Reed
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Kendelle J. Murphy
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Lily M. Channon
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Brooke A. Pereira
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Victoria M. Tyma
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Victoria Lee
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Michael Trpceski
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Jake Henry
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Immune Biotherapies Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Pauline Melenec
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Lea Abdulkhalek
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Max Nobis
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
| | - Xanthe L. Metcalf
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Shona Ritchie
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Antonia Cadell
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Translational Oncology Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Janett Stoehr
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Astrid Magenau
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Diego Chacon-Fajardo
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Translational Oncology Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Jessica L. Chitty
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Savannah O’Connell
- Immune Biotherapies Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Anaiis Zaratzian
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Michael Tayao
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Andrew Da Silva
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Ruth J. Lyons
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Leonard D. Goldstein
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Data Science Platform, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Ashleigh Dale
- Sydney Mass Spectrometry, University of Sydney, Sydney, New South Wales, Australia
| | - Alexander Rookyard
- Sydney Mass Spectrometry, University of Sydney, Sydney, New South Wales, Australia
| | - Angela Connolly
- Sydney Mass Spectrometry, University of Sydney, Sydney, New South Wales, Australia
| | - Ben Crossett
- Sydney Mass Spectrometry, University of Sydney, Sydney, New South Wales, Australia
| | - Yen T. H. Tran
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Peter Kaltzis
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Claire Vennin
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Marija Dinevska
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, Australia
- Department of Surgery, University of Melbourne, Melbourne, Australia
| | | | | | - David R. Croucher
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Translational Oncology Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Jaswinder Samra
- Royal North Shore Hospital, St Leonards, Sydney, New South Wales, Australia
| | - Anubhav Mittal
- Royal North Shore Hospital, St Leonards, Sydney, New South Wales, Australia
| | - Robert J. Weatheritt
- Immune Biotherapies Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Andrew Philp
- Centre for Healthy Ageing, Centenary Institute, Sydney, New South Wales, Australia
- School of Sport, Exercise and Rehabilitation Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Gonzalo Del Monte-Nieto
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Lei Zhang
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- St. Vincent’s Centre for Applied Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Ronaldo F. Enriquez
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Thomas R. Cox
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Yan-Chuan C. Shi
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Mark Pinese
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Nicola Waddell
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Hao-Wen Sim
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Tatyana Chtanova
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia
| | - Yingxiao Wang
- Department of Bioengineering & Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Anthony M. Joshua
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Translational Oncology Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Lorraine Chantrill
- Department of Medical Oncology and Illawarra Shoalhaven Local Health District, Wollongong, New South Wales, Australia
| | - Thomas R. Jeffry Evans
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | - Anthony J. Gill
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- Royal North Shore Hospital, St Leonards, Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Jennifer P. Morton
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | - Marina Pajic
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Translational Oncology Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Daniel Christ
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Immune Biotherapies Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Herbert Herzog
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- St. Vincent’s Centre for Applied Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Paul Timpson
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - David Herrmann
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| |
Collapse
|
26
|
Meier C, Brieger A. The role of IL-8 in cancer development and its impact on immunotherapy resistance. Eur J Cancer 2025; 218:115267. [PMID: 39899909 DOI: 10.1016/j.ejca.2025.115267] [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: 12/18/2024] [Revised: 01/28/2025] [Accepted: 01/28/2025] [Indexed: 02/05/2025]
Abstract
Tumors are structures of high complexity. Plurality of their structural and functional components - heterogeneity, diversity, directionality, interdependence and integration of signaling pathways - seem to follow isolated local rules, whereby a superordinate structure remains largely unknown. Understanding the complexity of cancer is the mainstay in finding determinants and developing effective therapies. Interleukin 8 (IL-8) is a potent pro-inflammatory chemokine that is significantly elevated in many different tumor entities. In contrast to its initially postulated anti-tumor properties, an increasing number of studies have been published in recent years linking this chemokine with tumor-promoting features and poor prognosis. This review summarizes the current state and diversity of the role of IL-8 in the development of cancer.
Collapse
Affiliation(s)
- Clara Meier
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Biomedical Research Laboratory, Frankfurt am Main, Germany
| | - Angela Brieger
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Biomedical Research Laboratory, Frankfurt am Main, Germany.
| |
Collapse
|
27
|
Seo R, de Guzman ACV, Park S, Lee JY, Kang SJ. Cancer-intrinsic Cxcl5 orchestrates a global metabolic reprogramming for resistance to oxidative cell death in 3D. Cell Death Differ 2025:10.1038/s41418-025-01466-y. [PMID: 40050422 DOI: 10.1038/s41418-025-01466-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 01/10/2025] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
Pancreatic ductal adenocarcinoma is characterized by a three-dimensional (3D) tumor microenvironment devoid of oxygen and nutrients but enriched in extracellular matrix, which acts as a physical and chemical barrier. In 3D, cancer cells reprogram their metabolic pathways in ways that help them survive hostile conditions. However, little is known about the metabolic phenotypes of cancer cells in 3D and the intrinsic cues that modulate them. We found that Cxcl5 deletion restricted pancreatic tumor growth in a 3D spheroid-in-Matrigel culture system without affecting cancer cell growth in 2D culture. Cxcl5 deletion impaired 3D-specific global metabolic reprogramming, resistance to hypoxia-induced cell death, and upregulation of Hif1α and Myc. Overexpression of Hif1α and Myc, however, effectively restored 3D culture-induced metabolic reconfiguration, growth, redox homeostasis, and mitochondrial function in Cxcl5-/- cells, reducing ferroptosis. We also found that pancreatic cancer patients with higher expression of hypoxia and metabolism-related genes whose expression is well-correlated with CXCL5 generally have poorer prognosis. Together, our findings identify an unanticipated role of Cxcl5 in orchestrating the cancer metabolic reprogramming in 3D culture that is required for energy and biomass maintenance and that restricts oxidative cell death. Thus, our results provide a rationale for targeting CXCL5 as a promising therapeutic strategy.
Collapse
Affiliation(s)
- Ramin Seo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Arvie Camille V de Guzman
- College of Pharmacy, Natural Product Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sunghyouk Park
- College of Pharmacy, Natural Product Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ji Youn Lee
- Biometrology Group, Division of Biomedical Metrology, Korea Research Institute of Standards and Science, Daejeon, 34113, Republic of Korea
| | - Suk-Jo Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
28
|
Gerlza T, Trojacher C, Fuchs T, Atlic A, Weis R, Adage T, Kungl AJ. Designing a CXCL8-hsa chimera as potential immunmodulator of the tumor micro-environment. Front Immunol 2025; 16:1539733. [PMID: 40124384 PMCID: PMC11926544 DOI: 10.3389/fimmu.2025.1539733] [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: 12/04/2024] [Accepted: 02/13/2025] [Indexed: 03/25/2025] Open
Abstract
Introduction CXCL8, belonging to inflammatory chemokines, is expressed by various cell types and plays a key role in leukocyte trafficking during infections, inflammatory processes, tissue injury and tumor progression. Chemokines interact not only with G-protein coupled receptors but also with glycosaminoglycans (GAGs), which are polyanionic linear polysaccharides. Chemokine-GAG interactions are critical for creating localized concentration gradients, protecting chemokines from degradation, and maintaining their efficacy in vivo. Methods We have previously engineered a CXCL8-based dominant-negative decoy ("PA401") with strongly increased GAG binding affinity combined with complete GPCR knockout, which was originally developed for the treatment of COPD. Here we have optimized our engineering protocol by minimizing CXCL8 mutations while conserving its in vitro dominant-negative activities. This novel CXCL8-based decoy (mtCXCL8) was further fused to human serum albumin (HSA) to overcome the typically very short serum half-life of chemokine-based biologics. We are therefore able to present here an entirely novel CXCL8-based biologic (hsa/mtCXCL8) which reflects our threefold modification strategy - increasing GAG-binding affinity by minimal mutagenesis, GPCR knockout, and fusion to HSA - thus representing a comprehensive and novel approach towards addressing chronic CXCL8-driven diseases. Results In the current study, we have investigated the immunomodulatory potential of our new decoy in a 3-D cellular tumor model ("BioMAP") which relates the biomarker interaction profile of immune and tumor cells to a data-base mirrored biomarker read-out. The obtained BioMAP results suggest an impact of hsa/mtCXCL8 on the immune compartment of the VascHT29 cell model by modulating cytokine levels and inhibiting immune cell activation markers. When combined with Keytruda (Pembrolizumab), a PD-1 inhibitor, it enhances some of its known activities, indicating potential synergistic effects, but further investigation is needed due to the observed increase in soluble IL-6 and limitations in dose selection for future in vivo studies. Discussion By prolonging the presence of engineered chemokine mutants in the bloodstream and optimizing their stability, these strategies aim to enhance the therapeutic efficacy of CXCL8-based interventions, offering promising avenues for the treatment of several CXCL8-mediated pathologies, including cancer.
Collapse
Affiliation(s)
- Tanja Gerlza
- Karl-Franzens-University Graz, Institute of Pharmaceutical Sciences, Pharmaceutical Chemistry, Graz, Austria
| | - Christina Trojacher
- Karl-Franzens-University Graz, Institute of Pharmaceutical Sciences, Pharmaceutical Chemistry, Graz, Austria
| | - Thomas Fuchs
- Medical University Graz, Otto Loewi Research Center, Graz, Austria
| | | | | | | | - Andreas J. Kungl
- Karl-Franzens-University Graz, Institute of Pharmaceutical Sciences, Pharmaceutical Chemistry, Graz, Austria
- Antagonis Biotherapeutics GmbH, Graz, Austria
| |
Collapse
|
29
|
Park SY, Pylaeva E, Bhuria V, Gambardella AR, Schiavoni G, Mougiakakos D, Kim SH, Jablonska J. Harnessing myeloid cells in cancer. Mol Cancer 2025; 24:69. [PMID: 40050933 PMCID: PMC11887392 DOI: 10.1186/s12943-025-02249-2] [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: 12/19/2024] [Accepted: 01/28/2025] [Indexed: 03/09/2025] Open
Abstract
Cancer-associated myeloid cells due to their plasticity play dual roles in both promoting and inhibiting tumor progression. Myeloid cells with immunosuppressive properties play a critical role in anti-cancer immune regulation. Cells of different origin, such as tumor associated macrophages (TAMs), tumor associated neutrophils (TANs), myeloid derived suppressor cells (also called MDSCs) and eosinophils are often expanded in cancer patients and significantly influence their survival, but also the outcome of anti-cancer therapies. For this reason, the variety of preclinical and clinical studies to modulate the activity of these cells have been conducted, however without successful outcome to date. In this review, pro-tumor activity of myeloid cells, myeloid cell-specific therapeutic targets, in vivo studies on myeloid cell re-polarization and the impact of myeloid cells on immunotherapies/genetic engineering are addressed. This paper also summarizes ongoing clinical trials and the concept of chimeric antigen receptor macrophage (CAR-M) therapies, and suggests future research perspectives, offering new opportunities in the development of novel clinical treatment strategies.
Collapse
Affiliation(s)
- Su-Yeon Park
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ekaterina Pylaeva
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, Essen, 45147, Germany
- German Cancer Consortium (DKTK) Partner Site Düsseldorf/Essen, Essen, Germany
| | - Vikas Bhuria
- Department of Hematology, Oncology, and Cell Therapy, Otto-Von-Guericke University, Magdeburg, Germany
| | | | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore Di Sanità, Rome, Italy
| | - Dimitrios Mougiakakos
- Department of Hematology, Oncology, and Cell Therapy, Otto-Von-Guericke University, Magdeburg, Germany
| | - Sung-Hoon Kim
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jadwiga Jablonska
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, Essen, 45147, Germany.
- German Cancer Consortium (DKTK) Partner Site Düsseldorf/Essen, Essen, Germany.
| |
Collapse
|
30
|
Xu W, Xu J, Liu J, Wang N, Zhou L, Guo J. Liver Metastasis in Cancer: Molecular Mechanisms and Management. MedComm (Beijing) 2025; 6:e70119. [PMID: 40027151 PMCID: PMC11868442 DOI: 10.1002/mco2.70119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Liver metastasis is a leading cause of mortality from malignant tumors and significantly impairs the efficacy of therapeutic interventions. In recent years, both preclinical and clinical research have made significant progress in understanding the molecular mechanisms and therapeutic strategies of liver metastasis. Metastatic tumor cells from different primary sites undergo highly similar biological processes, ultimately achieving ectopic colonization and growth in the liver. In this review, we begin by introducing the inherent metastatic-friendly features of the liver. We then explore the panorama of liver metastasis and conclude the three continuous, yet distinct phases based on the liver's response to metastasis. This includes metastatic sensing stage, metastatic stress stage, and metastasis support stage. We discuss the intricate interactions between metastatic tumor cells and various resident and recruited cells. In addition, we emphasize the critical role of spatial remodeling of immune cells in liver metastasis. Finally, we review the recent advancements and the challenges faced in the clinical management of liver metastasis. Future precise antimetastatic treatments should fully consider individual heterogeneity and implement different targeted interventions based on stages of liver metastasis.
Collapse
Affiliation(s)
- Wenchao Xu
- Department of General SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Key Laboratory of Research in Pancreatic TumorChinese Academy of Medical SciencesBeijingChina
- National Infrastructures for Translational MedicinePeking Union Medical College HospitalBeijingChina
- State Key Laboratory of ComplexSevere, and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jia Xu
- State Key Laboratory of Fine ChemicalsDepartment of Pharmaceutical SciencesSchool of Chemical EngineeringDalian University of TechnologyDalianChina
| | - Jianzhou Liu
- Department of General SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Key Laboratory of Research in Pancreatic TumorChinese Academy of Medical SciencesBeijingChina
- National Infrastructures for Translational MedicinePeking Union Medical College HospitalBeijingChina
- State Key Laboratory of ComplexSevere, and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Nanzhou Wang
- Department of Colorectal SurgeryState Key Laboratory of Oncology in South ChinaSun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerGuangzhouChina
| | - Li Zhou
- Department of General SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Key Laboratory of Research in Pancreatic TumorChinese Academy of Medical SciencesBeijingChina
- National Infrastructures for Translational MedicinePeking Union Medical College HospitalBeijingChina
- State Key Laboratory of ComplexSevere, and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Junchao Guo
- Department of General SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Key Laboratory of Research in Pancreatic TumorChinese Academy of Medical SciencesBeijingChina
- National Infrastructures for Translational MedicinePeking Union Medical College HospitalBeijingChina
- State Key Laboratory of ComplexSevere, and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
31
|
Li Y, Liu F, Cai Q, Deng L, Ouyang Q, Zhang XHF, Zheng J. Invasion and metastasis in cancer: molecular insights and therapeutic targets. Signal Transduct Target Ther 2025; 10:57. [PMID: 39979279 PMCID: PMC11842613 DOI: 10.1038/s41392-025-02148-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 12/24/2024] [Accepted: 01/16/2025] [Indexed: 02/22/2025] Open
Abstract
The progression of malignant tumors leads to the development of secondary tumors in various organs, including bones, the brain, liver, and lungs. This metastatic process severely impacts the prognosis of patients, significantly affecting their quality of life and survival rates. Research efforts have consistently focused on the intricate mechanisms underlying this process and the corresponding clinical management strategies. Consequently, a comprehensive understanding of the biological foundations of tumor metastasis, identification of pivotal signaling pathways, and systematic evaluation of existing and emerging therapeutic strategies are paramount to enhancing the overall diagnostic and treatment capabilities for metastatic tumors. However, current research is primarily focused on metastasis within specific cancer types, leaving significant gaps in our understanding of the complex metastatic cascade, organ-specific tropism mechanisms, and the development of targeted treatments. In this study, we examine the sequential processes of tumor metastasis, elucidate the underlying mechanisms driving organ-tropic metastasis, and systematically analyze therapeutic strategies for metastatic tumors, including those tailored to specific organ involvement. Subsequently, we synthesize the most recent advances in emerging therapeutic technologies for tumor metastasis and analyze the challenges and opportunities encountered in clinical research pertaining to bone metastasis. Our objective is to offer insights that can inform future research and clinical practice in this crucial field.
Collapse
Affiliation(s)
- Yongxing Li
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fengshuo Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA
- Graduate School of Biomedical Science, Cancer and Cell Biology Program, Baylor College of Medicine, Houston, TX, USA
| | - Qingjin Cai
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lijun Deng
- Department of Medicinal Chemistry, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qin Ouyang
- Department of Medicinal Chemistry, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA.
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
32
|
Nishinakamura H, Shinya S, Irie T, Sakihama S, Naito T, Watanabe K, Sugiyama D, Tamiya M, Yoshida T, Hase T, Yoshida T, Karube K, Koyama S, Nishikawa H. Coactivation of innate immune suppressive cells induces acquired resistance against combined TLR agonism and PD-1 blockade. Sci Transl Med 2025; 17:eadk3160. [PMID: 39937883 DOI: 10.1126/scitranslmed.adk3160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/02/2024] [Accepted: 01/03/2025] [Indexed: 02/14/2025]
Abstract
Immune checkpoint blockade therapy has been successfully applied in clinical settings as a standard therapy for many cancer types, but its clinical efficacy is restricted to patients with immunologically hot tumors. Various strategies to modify the tumor microenvironment (TME), such as Toll-like receptor (TLR) agonists that can stimulate innate immunity, have been explored but have not been successful. Here, we show a mechanism of acquired resistance to combination treatment consisting of an agonist for multiple TLRs, OK-432 (Picibanil), and programmed cell death protein 1 (PD-1) blockade. Adding the TLR agonist failed to convert the TME from immunogenically cold to hot and did not augment antitumor immunity, particularly CD8+ T cell responses, in multiple animal models. The failure was attributed to the coactivation of innate suppressive cells, such as polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) expressing CXCR2, through high CXCL1 production by macrophages in the TME upon OK-432 treatment. A triple combination treatment with OK-432, PD-1 blockade, and a CXCR2 neutralizing antibody overcame the resistance induced by PMN-MDSCs, resulting in a stronger antitumor effect than that of any dual combinations or single treatments. The accumulation of PMN-MDSCs was similarly observed in the pleural effusions of patients with lung cancer after OK-432 administration. We propose that successful combination cancer immunotherapy intended to stimulate innate antitumor immunity requires modulation of unwanted activation of innate immune suppressive cells, including PMN-MDSCs.
Collapse
Affiliation(s)
- Hitomi Nishinakamura
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center Japan, Tokyo, 104-0045/Chiba 277-8577, Japan
| | - Sayoko Shinya
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center Japan, Tokyo, 104-0045/Chiba 277-8577, Japan
- Discovery and Research, Ono Pharmaceutical Co. Ltd., Osaka, 618-8585, Japan
| | - Takuma Irie
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center Japan, Tokyo, 104-0045/Chiba 277-8577, Japan
| | - Shugo Sakihama
- Laboratory of Hemato-Immunology, Graduate School of Health Sciences, University of the Ryukyus, Nishihara, 903-0125, Japan
| | - Takeo Naito
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center Japan, Tokyo, 104-0045/Chiba 277-8577, Japan
| | - Keisuke Watanabe
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center Japan, Tokyo, 104-0045/Chiba 277-8577, Japan
| | - Daisuke Sugiyama
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Motohiro Tamiya
- Respiratory Medicine, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Tatsuya Yoshida
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Tetsunari Hase
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Takao Yoshida
- Discovery and Research, Ono Pharmaceutical Co. Ltd., Osaka, 618-8585, Japan
| | - Kennosuke Karube
- Department of Pathology and Laboratory Medicine, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shohei Koyama
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center Japan, Tokyo, 104-0045/Chiba 277-8577, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center Japan, Tokyo, 104-0045/Chiba 277-8577, Japan
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Division of Cancer Immune Multicellular System Regulation, Center for Cancer Immunotherapy and Immunobiology (CCII), Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
33
|
Aleksandrova Y, Neganova M. Antioxidant Senotherapy by Natural Compounds: A Beneficial Partner in Cancer Treatment. Antioxidants (Basel) 2025; 14:199. [PMID: 40002385 PMCID: PMC11851806 DOI: 10.3390/antiox14020199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Aging is a general biological process inherent in all living organisms. It is characterized by progressive cellular dysfunction. For many years, aging has been widely recognized as a highly effective mechanism for suppressing the progression of malignant neoplasms. However, in recent years, increasing evidence suggests a "double-edged" role of aging in cancer development. According to these data, aging is not only a tumor suppressor that leads to cell cycle arrest in neoplastic cells, but also a cancer promoter that ensures a chronic proinflammatory and immunosuppressive microenvironment. In this regard, in our review, we discuss recent data on the destructive role of senescent cells in the pathogenesis of cancer. We also identify for the first time correlations between the modulation of the senescence-associated secretory phenotype and the antitumor effects of naturally occurring molecules.
Collapse
Affiliation(s)
| | - Margarita Neganova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Vavilova St. 28, Bld. 1, Moscow 119991, Russia;
| |
Collapse
|
34
|
D'Alessio-Sands L, Gaynier J, Michel-Milian V, Agbowuro AA, Brackett CM. Current Strategies and Future Dimensions in the Development of KRAS Inhibitors for Targeted Anticancer Therapy. Drug Dev Res 2025; 86:e70042. [PMID: 39799558 DOI: 10.1002/ddr.70042] [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: 09/10/2024] [Revised: 11/05/2024] [Accepted: 12/15/2024] [Indexed: 01/15/2025]
Abstract
KRAS is a proto-oncogene that is found to be mutated in 15% of all metastatic cancers with high prevalence in pancreatic, lung, and colorectal cancers. Additionally, patients harboring KRAS mutations respond poorly to standard cancer therapy. As a result, KRAS is seen as an attractive target for targeted anticancer therapy. Over the last decade, this protein has evolved from being termed "undruggable" to producing two clinically approved drugs along with several more in clinical development, and many under preclinical investigations. This review details the development of various KRAS-targeted molecules with emphasis on the different drug design strategies employed by examining the following areas: (1) Direct inhibition of KRAS mutants using small molecule binders, (2) Inhibiting the activated state of KRAS mutants using a binary complex of small molecule binders and cyclophilin A, and (3) Targeted degradation of KRAS mutants using the PROTAC approach. We assess the pharmacological attributes and possible clinical benefits of the different molecules and look to the next frontiers in the application of KRAS inhibitors as anticancer agents.
Collapse
Affiliation(s)
| | - Joshua Gaynier
- South University School of Pharmacy, Savannah, Giorgia, USA
| | | | | | | |
Collapse
|
35
|
Wu X, Pan B, Chu C, Zhang Y, Ma J, Xing Y, Ma Y, Zhu W, Zhong H, Alimu A, Zhou G, Liu S, Chen W, Li X, Puyi S. CXCL16/CXCR6/TGF-β Feedback Loop Between M-MDSCs and Treg Inhibits Anti-Bacterial Immunity During Biofilm Infection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409537. [PMID: 39716908 PMCID: PMC11831521 DOI: 10.1002/advs.202409537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/29/2024] [Indexed: 12/25/2024]
Abstract
Staphylococcus aureus (S. aureus) is a leading cause of Periprosthetic joint infection (PJI), a severe complication after joint arthroplasty. Immunosuppression is a major factor contributing to the infection chronicity of S. aureus PJI, posing significant treatment challenges. This study investigates the relationship between the immunosuppressive biofilm milieu and S. aureus PJI outcomes in both discovery and validation cohorts. This scRNA-seq analysis of synovium from PJI patients reveals an expansion and heightened activity of monocyte-related myeloid-derived suppressor cells (M-MDSCs) and regulatory T cells (Treg). Importantly, CXCL16 is significantly upregulated in M-MDSCs, with its corresponding CXCR6 receptor also elevated on Treg. M-MDSCs recruit Treg and enhance its activity via CXCL16-CXCR6 interactions, while Treg secretes TGF-β, inducing M-MDSCs proliferation and immunosuppressive activity. Interfering with this cross-talk in vivo using Treg-specific CXCR6 knockout PJI mouse model reduces M-MDSCs/Treg-mediated immunosuppression and alleviates bacterial burden. Immunohistochemistry and recurrence analysis show that PJI patients with CXCR6high synovium have poor prognosis. This findings highlight the critical role of CXCR6 in Treg in orchestrating an immunosuppressive microenvironment and biofilm persistence during PJI, offering potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Xiaoyu Wu
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Baiqi Pan
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Chenghan Chu
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Yangchun Zhang
- Department of OrthopedicsThe People's Hospital of Baoan ShenzhenShenzhenGuangdong518101China
- Department of OrthopedicsThe Second Affiliated Hospital of Shenzhen UniversityShenzhenGuangdong518101China
| | - Jinjin Ma
- Technology School of MedicineSouth China University of TechnologyGuangzhouGuangdong510640China
- Shien‐ming Wu School of Intelligent EngineeringSouth China University of TechnologyGuangzhouGuangdong510640China
| | - Yang Xing
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Yuanchen Ma
- Department of OrthopedicsGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouGuangdong519041China
| | - Wengang Zhu
- Department of Joint OrthopedicsYuebei People's HospitalShaoguanGuangdong512099China
| | - Huan Zhong
- Department of Joint SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong524002China
| | - Aerman Alimu
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Guanming Zhou
- Department of OrthopedicsFoshan Hospital of Traditional Chinese MedicineGuangzhouGuangdong528051China
| | - Shuying Liu
- Department of Histology and EmbryologyZhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Weishen Chen
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Xiang Li
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Department of Spine SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Sheng Puyi
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| |
Collapse
|
36
|
Luyang H, Zeng F, Lei Y, He Q, Zhou Y, Xu J. Bidirectional role of neutrophils in tumor development. Mol Cancer 2025; 24:22. [PMID: 39819428 PMCID: PMC11737241 DOI: 10.1186/s12943-025-02228-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025] Open
Abstract
Neutrophils, traditionally considered as non-specific components of the innate immune system, have garnered considerable research interest due to their dual roles in both promoting and inhibiting tumor progression. This paper seeks to clarify the specific mechanisms by which neutrophils play a bidirectional role in tumor immunity and the factors that influence these roles. By conducting a comprehensive analysis and synthesis of a vast array of relevant literature, it has become evident that neutrophils can influence tumor development and invasive migration through various mechanisms, thereby exerting their anti-tumor effects. Conversely, they can also facilitate tumorigenesis and proliferation, as well as affect the normal physiological functions of other immune cells, thus exerting pro-tumor effects. Moreover, neutrophils are influenced by tumor cells and their unique microenvironment, which in turn affects their heterogeneity and plasticity. Neutrophils interact with tumor cells to regulate various aspects of their life activities precisely. This paper also identifies unresolved issues in the research concerning the bidirectional role of neutrophils in tumorigenesis and tumor development, offering new opportunities and challenges for advancing our understanding. This, in turn, can aid in the proper application of these insights to clinical treatment strategies.
Collapse
Affiliation(s)
- Haoxin Luyang
- Department of critical care medicine, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, China
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410011, Hunan, China
| | - Feng Zeng
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410011, Hunan, China
| | - Yan Lei
- Department of Blood Transfusion, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, China
| | - Qian He
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, China
| | - Yanhong Zhou
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410011, Hunan, China.
| | - Juan Xu
- Department of critical care medicine, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, China.
| |
Collapse
|
37
|
Ng M, Cerezo-Wallis D, Ng LG, Hidalgo A. Adaptations of neutrophils in cancer. Immunity 2025; 58:40-58. [PMID: 39813993 DOI: 10.1016/j.immuni.2024.12.009] [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: 09/02/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025]
Abstract
There is a renewed interest in neutrophil biology, largely instigated by their prominence in cancer. From an immunologist's perspective, a conceptual breakthrough is the realization that prototypical inflammatory, cytotoxic leukocytes can be tamed to promote the survival and growth of other cells. This has sparked interest in defining the biological principles and molecular mechanisms driving the adaptation of neutrophils to cancer. Yet, many questions remain: is this adaptation mediated by reprogramming mature neutrophils inside the tumoral mass, or rather by rewiring granulopoiesis in the bone marrow? Why, in some instances, are neutrophils beneficial and in others detrimental to cancer? How many different functional programs can be induced in neutrophils by tumors, and is this dependent on the type of tumor? This review summarizes what we know about these questions and discusses therapeutic strategies based on our incipient knowledge of how neutrophils adapt to cancer.
Collapse
Affiliation(s)
- Melissa Ng
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore, Singapore.
| | - Daniela Cerezo-Wallis
- Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| | - Lai Guan Ng
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Andres Hidalgo
- Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
38
|
Glaviano A, Lau HSH, Carter LM, Lee EHC, Lam HY, Okina E, Tan DJJ, Tan W, Ang HL, Carbone D, Yee MYH, Shanmugam MK, Huang XZ, Sethi G, Tan TZ, Lim LHK, Huang RYJ, Ungefroren H, Giovannetti E, Tang DG, Bruno TC, Luo P, Andersen MH, Qian BZ, Ishihara J, Radisky DC, Elias S, Yadav S, Kim M, Robert C, Diana P, Schalper KA, Shi T, Merghoub T, Krebs S, Kusumbe AP, Davids MS, Brown JR, Kumar AP. Harnessing the tumor microenvironment: targeted cancer therapies through modulation of epithelial-mesenchymal transition. J Hematol Oncol 2025; 18:6. [PMID: 39806516 PMCID: PMC11733683 DOI: 10.1186/s13045-024-01634-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025] Open
Abstract
The tumor microenvironment (TME) is integral to cancer progression, impacting metastasis and treatment response. It consists of diverse cell types, extracellular matrix components, and signaling molecules that interact to promote tumor growth and therapeutic resistance. Elucidating the intricate interactions between cancer cells and the TME is crucial in understanding cancer progression and therapeutic challenges. A critical process induced by TME signaling is the epithelial-mesenchymal transition (EMT), wherein epithelial cells acquire mesenchymal traits, which enhance their motility and invasiveness and promote metastasis and cancer progression. By targeting various components of the TME, novel investigational strategies aim to disrupt the TME's contribution to the EMT, thereby improving treatment efficacy, addressing therapeutic resistance, and offering a nuanced approach to cancer therapy. This review scrutinizes the key players in the TME and the TME's contribution to the EMT, emphasizing avenues to therapeutically disrupt the interactions between the various TME components. Moreover, the article discusses the TME's implications for resistance mechanisms and highlights the current therapeutic strategies toward TME modulation along with potential caveats.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Hannah Si-Hui Lau
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Lukas M Carter
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Donavan Jia Jie Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Wency Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Michelle Yi-Hui Yee
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Xiao Zi Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Lina H K Lim
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Ruby Yun-Ju Huang
- School of Medicine and Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, 23538, Lübeck, Germany
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Experimental Therapeutics (ET) Graduate Program, University at Buffalo & Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Tullia C Bruno
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Bin-Zhi Qian
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, The Human Phenome Institute, Zhangjiang-Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London, W12 0BZ, UK
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Salem Elias
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Saurabh Yadav
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Minah Kim
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Caroline Robert
- Department of Cancer Medicine, Inserm U981, Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif, France
- Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, Paris, France
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Tao Shi
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, NY, USA
| | - Simone Krebs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironment Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Matthew S Davids
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jennifer R Brown
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
| |
Collapse
|
39
|
Liu Y, Han J, Hsu WH, LaBella KA, Deng P, Shang X, de Lara PT, Cai L, Jiang S, DePinho RA. Combined KRAS Inhibition and Immune Therapy Generates Durable Complete Responses in an Autochthonous PDAC Model. Cancer Discov 2025; 15:162-178. [PMID: 39348506 PMCID: PMC11858029 DOI: 10.1158/2159-8290.cd-24-0489] [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: 04/08/2024] [Revised: 08/27/2024] [Accepted: 09/26/2024] [Indexed: 10/02/2024]
Abstract
SIGNIFICANCE Clinically available KRAS* inhibitors and IO agents alleviated the immunosuppressive tumor microenvironment in PDAC. Profound tumor regression and prolonged survival in an autochthonous PDAC model provide a compelling rationale for combining KRAS* inhibition with IO agents targeting multiple arms of the immunity cycle to combat PDAC.
Collapse
Affiliation(s)
- Yonghong Liu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Jincheng Han
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Wen-Hao Hsu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Kyle A. LaBella
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Pingna Deng
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Xiaoying Shang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Paulino Tallón de Lara
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Li Cai
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Shan Jiang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Ronald A. DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| |
Collapse
|
40
|
Ge X, Zhang K, Zhu J, Chen Y, Wang Z, Wang P, Xu P, Yao J. Targeting protein modification: a new direction for immunotherapy of pancreatic cancer. Int J Biol Sci 2025; 21:63-74. [PMID: 39744438 PMCID: PMC11667816 DOI: 10.7150/ijbs.101861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 11/03/2024] [Indexed: 01/11/2025] Open
Abstract
Post-translational modifications (PTMs) alter protein conformation by covalently attaching functional groups to substrates, influencing their biological activity, mechanisms of action, and functional performance. PTMs and their interactions are essential to many critical signal transduction processes, including tumor transformation, cancer progression, and metastasis in pancreatic cancer. Additionally, advancements in tumor immunotherapy indicate that PTMs are essential in immune cell activation, transport, and energy metabolism. This study aimed to investigate the effects of different PTMs on immunotherapy for pancreatic cancer, providing new perspectives and suggesting directions for future research.
Collapse
Affiliation(s)
- Xinyu Ge
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu 225000, China
| | - Ke Zhang
- The Yangzhou School of Clinical Medicine of Dalian Medical University, Jiangsu 225000, China
| | - Jie Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu 225000, China
| | - Yuan Chen
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu 225000, China
| | - Zhengwang Wang
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu 225000, China
| | - Peng Wang
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu 225000, China
| | - Peng Xu
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu 225000, China
| | - Jie Yao
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu 225000, China
| |
Collapse
|
41
|
Coperchini F, Greco A, Petrosino E, Croce L, Teliti M, Marchesi N, Pascale A, Calì B, Pignatti P, Magri F, Uddin M, Rotondi M. Selective anti-CXCR2 receptor blockade by AZD5069 inhibits CXCL8-mediated pro-tumorigenic activity in human thyroid cancer cells in vitro. J Endocrinol Invest 2025; 48:53-65. [PMID: 38900374 PMCID: PMC11729135 DOI: 10.1007/s40618-024-02410-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Thyroid cancer is the most common endocrine malignancy. Current therapies are successful, however some patients progress to therapeutically refractive disease. The immunotherapeutic potential of the CXCL8-chemokine/CXCR2-chemokine-receptor system is currently being explored in numerous human cancers. This study aimed to evaluate if the targeting of CXCR2 by its selective antagonist, AZD5069, could modulate CXCL8-mediated pro-tumorigenic effects in thyroid-cancer (TC) cells in vitro. METHODS Normal human primary thyroid cells (NHT) and TC cell lines TPC-1 (RET/PTC), BCPAP, 8505C and 8305C (BRAFV600e) were treated with AZD5069 (100 pM-10 µM) over a time-course. Viability and proliferation were assessed by WST-1 and crystal violet assays. CXCL8 and CXCR2 mRNA were evaluated by RT-PCR. CXCL8-protein concentrations were measured in cell culture supernatants by ELISA. CXCR2 on cell surface was evaluated by flow-cytometry. Cell-migration was assessed by trans-well-migration chamber-system. RESULTS AZD5069 exerted negligible effects on cell proliferation or viability. AZD5069 significantly reduced CXCR2, (but not CXCL8) mRNAs in all cell types. CXCR2 was reduced on the membrane of some TC cell lines. A significant reduction of the CXCL8 secretion was found in TPC-1 cells (basal-secretion) and NHT (TNFα-induced secretion). AZD5069 significantly reduced basal and CXCL8-induced migration in NHT and different TC cells. CONCLUSIONS Our findings confirm the involvement of the CXCL8/CXCR2-axis in promoting pro-tumorigenic effects in TC cells, further demonstrating its immunotherapeutic significance in human cancer.
Collapse
Affiliation(s)
- F Coperchini
- Department of Internal Medicine and Therapeutics, University of Pavia, Via S. Maugeri 4, 27100, Pavia, Italy
| | - A Greco
- Department of Internal Medicine and Therapeutics, University of Pavia, Via S. Maugeri 4, 27100, Pavia, Italy
| | - E Petrosino
- Department of Internal Medicine and Therapeutics, University of Pavia, Via S. Maugeri 4, 27100, Pavia, Italy
| | - L Croce
- Department of Internal Medicine and Therapeutics, University of Pavia, Via S. Maugeri 4, 27100, Pavia, Italy
- Unit of Endocrinology and Metabolism, Laboratory for Endocrine Disruptors, Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy
| | - M Teliti
- Department of Internal Medicine and Therapeutics, University of Pavia, Via S. Maugeri 4, 27100, Pavia, Italy
- Unit of Endocrinology and Metabolism, Laboratory for Endocrine Disruptors, Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy
| | - N Marchesi
- Unit of Pharmacology, Department of Drug Sciences, University of Pavia, 27100, Pavia, Italy
| | - A Pascale
- Unit of Pharmacology, Department of Drug Sciences, University of Pavia, 27100, Pavia, Italy
| | - B Calì
- Department of General and Minimally Invasive Surgery, Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia (PV), Italy
| | - P Pignatti
- Allergy and Immunology Unit, Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy
| | - F Magri
- Department of Internal Medicine and Therapeutics, University of Pavia, Via S. Maugeri 4, 27100, Pavia, Italy
- Unit of Endocrinology and Metabolism, Laboratory for Endocrine Disruptors, Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy
| | - M Uddin
- AstraZeneca Gothenburg, Biopharmaceuticals R&D, Mӧlndal, Sweden
| | - M Rotondi
- Department of Internal Medicine and Therapeutics, University of Pavia, Via S. Maugeri 4, 27100, Pavia, Italy.
- Unit of Endocrinology and Metabolism, Laboratory for Endocrine Disruptors, Istituti Clinici Scientifici Maugeri IRCCS, 27100, Pavia, Italy.
| |
Collapse
|
42
|
Hanahan D, Michielin O, Pittet MJ. Convergent inducers and effectors of T cell paralysis in the tumour microenvironment. Nat Rev Cancer 2025; 25:41-58. [PMID: 39448877 DOI: 10.1038/s41568-024-00761-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/23/2024] [Indexed: 10/26/2024]
Abstract
Tumorigenesis embodies the formation of a heterotypic tumour microenvironment (TME) that, among its many functions, enables the evasion of T cell-mediated immune responses. Remarkably, most TME cell types, including cancer cells, fibroblasts, myeloid cells, vascular endothelial cells and pericytes, can be stimulated to deploy immunoregulatory programmes. These programmes involve regulatory inducers (signals-in) and functional effectors (signals-out) that impair CD8+ and CD4+ T cell activity through cytokines, growth factors, immune checkpoints and metabolites. Some signals target specific cell types, whereas others, such as transforming growth factor-β (TGFβ) and prostaglandin E2 (PGE2), exert broad, pleiotropic effects; as signals-in, they trigger immunosuppressive programmes in most TME cell types, and as signals-out, they directly inhibit T cells and also modulate other cells to reinforce immunosuppression. This functional diversity and redundancy pose a challenge for therapeutic targeting of the immune-evasive TME. Fundamentally, the commonality of regulatory programmes aimed at abrogating T cell activity, along with paracrine signalling between cells of the TME, suggests that many normal cell types are hard-wired with latent functions that can be triggered to prevent inappropriate immune attack. This intrinsic capability is evidently co-opted throughout the TME, enabling tumours to evade immune destruction.
Collapse
Affiliation(s)
- Douglas Hanahan
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, Switzerland.
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.
| | - Olivier Michielin
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
- Department of Medicine, University of Geneva (UNIGE), Geneva, Switzerland
| | - Mikael J Pittet
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
- Department of Pathology and Immunology, University of Geneva (UNIGE), Geneva, Switzerland
| |
Collapse
|
43
|
Fan G, Na J, Shen Z, Lin F, Zhong L. Heterogeneity of tumor-associated neutrophils in hepatocellular carcinoma. Mol Immunol 2025; 177:1-16. [PMID: 39642781 DOI: 10.1016/j.molimm.2024.11.009] [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: 09/19/2024] [Revised: 11/08/2024] [Accepted: 11/17/2024] [Indexed: 12/09/2024]
Abstract
Neutrophils are the most abundant cell type in human blood and play a crucial role in the immune system and development of tumors. This review begins with the generation and development of neutrophils, traces their release from the bone marrow into the bloodstream, and finally discusses their role in the hepatocellular carcinoma (HCC) microenvironment. It elaborates in detail the mechanisms by which tumor-associated neutrophils (TANs) exert antitumor or protumor effects under the influence of various mediators in the tumor microenvironment. Neutrophils can exert antitumor effects through direct cytotoxic action. However, they can also accelerate the formation and progression of HCC by being recruited and infiltrated, promoting tumor angiogenesis, and maintaining an immunosuppressive microenvironment. Therefore, based on the heterogeneity and plasticity of neutrophils in tumor development, this review summarizes the current immunotherapies targeting TANs, discusses potential opportunities and challenges, and provides new insights into exploring more promising strategies for treating HCC.
Collapse
Affiliation(s)
- Guixiang Fan
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, Guangxi 530021, China; Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jintong Na
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Zhen Shen
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, Guangxi 530021, China; Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Faquan Lin
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China.
| | - Liping Zhong
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, Guangxi 530021, China; Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi 530021, China.
| |
Collapse
|
44
|
Koenderman L, Vrisekoop N. Neutrophils in cancer: from biology to therapy. Cell Mol Immunol 2025; 22:4-23. [PMID: 39653768 PMCID: PMC11686117 DOI: 10.1038/s41423-024-01244-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/21/2024] [Indexed: 12/12/2024] Open
Abstract
The view of neutrophils has shifted from simple phagocytic cells, whose main function is to kill pathogens, to very complex cells that are also involved in immune regulation and tissue repair. These cells are essential for maintaining and regaining tissue homeostasis. Neutrophils can be viewed as double-edged swords in a range of situations. The potent killing machinery necessary for immune responses to pathogens can easily lead to collateral damage to host tissues when inappropriately controlled. Furthermore, some subtypes of neutrophils are potent pathogen killers, whereas others are immunosuppressive or can aid in tissue healing. Finally, in tumor immunology, many examples of both protumorigenic and antitumorigenic properties of neutrophils have been described. This has important consequences for cancer therapy, as targeting neutrophils can lead to either suppressed or stimulated antitumor responses. This review will discuss the current knowledge regarding the pro- and antitumorigenic roles of neutrophils, leading to the concept of a confused state of neutrophil-driven pro-/antitumor responses.
Collapse
Affiliation(s)
- Leo Koenderman
- Dept. Respiratory Medicine and Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Nienke Vrisekoop
- Dept. Respiratory Medicine and Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
45
|
Santiago-Sánchez GS, Fabian KP, Hodge JW. A landscape of checkpoint blockade resistance in cancer: underlying mechanisms and current strategies to overcome resistance. Cancer Biol Ther 2024; 25:2308097. [PMID: 38306161 PMCID: PMC10841019 DOI: 10.1080/15384047.2024.2308097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/17/2024] [Indexed: 02/03/2024] Open
Abstract
The discovery of immune checkpoints and the development of immune checkpoint inhibitors (ICI) have achieved a durable response in advanced-stage cancer patients. However, there is still a high proportion of patients who do not benefit from ICI therapy due to a lack of response when first treated (primary resistance) or detection of disease progression months after objective response is observed (acquired resistance). Here, we review the current FDA-approved ICI for the treatment of certain solid malignancies, evaluate the contrasting responses to checkpoint blockade in different cancer types, explore the known mechanisms associated with checkpoint blockade resistance (CBR), and assess current strategies in the field that seek to overcome these mechanisms. In order to improve current therapies and develop new ones, the immunotherapy field still has an unmet need in identifying other molecules that act as immune checkpoints, and uncovering other mechanisms that promote CBR.
Collapse
Affiliation(s)
- Ginette S. Santiago-Sánchez
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kellsye P. Fabian
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James W. Hodge
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
46
|
Paczkowska J, Tang M, Wright KT, Song L, Luu K, Shanmugam V, Welsh EL, Weirather JL, Besson N, Olszewski H, Porter BA, Pfaff KL, Redd RA, Cader FZ, Mandato E, Ouyang J, Calabretta E, Bai G, Lawton LN, Armand P, Rodig SJ, Liu XS, Shipp MA. Cancer-specific innate and adaptive immune rewiring drives resistance to PD-1 blockade in classic Hodgkin lymphoma. Nat Commun 2024; 15:10740. [PMID: 39737927 PMCID: PMC11686379 DOI: 10.1038/s41467-024-54512-7] [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: 02/07/2024] [Accepted: 11/11/2024] [Indexed: 01/01/2025] Open
Abstract
Hodgkin Reed-Sternberg (HRS) cells of classic Hodgkin lymphoma (cHL), like many solid tumors, elicit ineffective immune responses. However, patients with cHL are highly responsive to PD-1 blockade, which largely depends on HRS cell-specific retention of MHC class II and implicates CD4+ T cells and additional MHC class I-independent immune effectors. Here, we utilize single-cell RNA sequencing and spatial analysis to define shared circulating and microenvironmental features of the immune response to PD-1 blockade in cHL. Compared with non-responders, responding patients have more circulating CD4+ naïve and central memory T cells and B cells, as well as more diverse CD4+ T cell and B cell receptor repertoires. Importantly, a population of circulating and tumor-infiltrating IL1β+ monocytes/macrophages is detectable in patients with cHL but not healthy donors, and a proinflammatory, tumor-promoting signature of these circulating IL1β+ monocytes is associated with resistance to PD-1 blockade in cHL. Altogether, our findings reveal extensive immune rewiring and complementary roles of CD4+ T cells, B cells and IL1β+ monocytes in the response to PD-1 blockade and suggest that these features can be captured with a peripheral blood test.
Collapse
Affiliation(s)
- Julia Paczkowska
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ming Tang
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Astra Zeneca, Waltham, MA, USA
| | - Kyle T Wright
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Li Song
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biomedical Data Science, Dartmouth College, Hanover, NH, USA
| | - Kelsey Luu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- PathAI, Boston, MA, USA
| | - Vignesh Shanmugam
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Emma L Welsh
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jason L Weirather
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Naomi Besson
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Harrison Olszewski
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Billie A Porter
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kathleen L Pfaff
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Robert A Redd
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Fathima Zumla Cader
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- AstraZeneca, City House, Cambridge, UK
| | - Elisa Mandato
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jing Ouyang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Mechanisms of Cancer Resistance Thematic Center, Bristol Myers Squibb, Cambridge, MA, USA
| | - Eleonora Calabretta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gali Bai
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Lee N Lawton
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Philippe Armand
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Scott J Rodig
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Xiaole Shirley Liu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
- GV20 Therapeutics, LLC, Cambridge, MA, USA
| | - Margaret A Shipp
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
47
|
Diao X, Zhan C, Ye H, Wu H, Yi C, Lin J, Mao H, Chen W, Yang X. Single-cell transcriptomic reveals the peritoneal microenvironmental change in long-term peritoneal dialysis patients with ultrafiltration failure. iScience 2024; 27:111383. [PMID: 39687014 PMCID: PMC11647153 DOI: 10.1016/j.isci.2024.111383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 07/02/2024] [Accepted: 11/11/2024] [Indexed: 12/18/2024] Open
Abstract
The microenvironmental changes in peritoneal dialysis effluent (PDE) after long-term vintage (LV) of PD in patients with ultrafiltration failure (LV_UF) are unclear. Single-cell sequencing revealed that peritoneal neutrophils were elevated in LV_UF patients, while MRC1-macrophage subcluster decreased compared with PD patients with short vintage (SV) and LV without ultrafiltration failure (LV_NOT_UF). Compared with the LV_NOT_UF group, the upregulated differentially expressed genes (DEGs) of monocytes/macrophages in the LV_UF group were involved in inflammatory response and EMT progress. LV_UF patients had a higher proportion of epithelial-like mesothelial cells (E-MCs), which were characterized by autophagy activation, inflammation, and upregulation of neutrophil- and autophagy-related DEGs compared to the LV_NOT_UF group. Additionally, mesenchymal-like MCs and AQP1 expression were reduced in the LV_UF group compared with the other groups. Both neutrophils and monocytes/macrophages interacted with MCs. Our study provides insights into the roles of peritoneal mesothelial cells and inflammatory cells in PD patients with UF.
Collapse
Affiliation(s)
- Xiangwen Diao
- Department of Emergency, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Cuixia Zhan
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, Guangdong, China
| | - Hongjian Ye
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, Guangdong, China
| | - Haishan Wu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, Guangdong, China
| | - Chunyan Yi
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, Guangdong, China
| | - Jianxiong Lin
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, Guangdong, China
| | - Haiping Mao
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, Guangdong, China
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, Guangdong, China
| | - Xiao Yang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, Guangdong, China
| |
Collapse
|
48
|
Kang W, Wang C, Wang M, Liu M, Hu W, Liang X, Yang J, Zhang Y. A key regulator of tumor-associated neutrophils: the CXCR2 chemokine receptor. J Mol Histol 2024; 55:1051-1061. [PMID: 39269537 DOI: 10.1007/s10735-024-10260-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024]
Abstract
In recent years, with the advance of research, the role of tumor-associated neutrophils (TANs) in tumors has become a research hotspot. As important effector cells in the innate immune system, neutrophils play a key role in the immune and inflammatory responses of the body. As the first line of defense against bacterial and fungal infections, neutrophils have the ability to kill invading pathogens. In the pathological state of malignant tumors, the phenotype of neutrophils is altered and has an important regulatory function in tumor development. The C-X-C motif chemokine receptor 2(CXCR2) is a key molecule that mediates the migration and aggregation signaling pathway of immune cells, especially neutrophils. This review focuses on the regulation of CXCR2 on TANs in the process of tumorigenesis and development, and emphasizes the application significance of CXCR2 inhibitors in blocking the migration of TANs to tumors.
Collapse
Affiliation(s)
- Wenyan Kang
- Department of Gynecology, The First Affiliated Hospital, Hengyang School of Medicine, University of South China, Hengyang, 421001, Hunan, P.R. China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China
| | - Chengkun Wang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China
| | - Minhui Wang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China
| | - Meiqi Liu
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China
| | - Wei Hu
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China
| | - Xiaoqiu Liang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China
| | - Juanli Yang
- Department of Gynecology, The First Affiliated Hospital, Hengyang School of Medicine, University of South China, Hengyang, 421001, Hunan, P.R. China.
| | - Yang Zhang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China.
| |
Collapse
|
49
|
Ye Y, Yu S, Guo T, Zhang S, Shen X, Han G. Epithelial-Mesenchymal Transition in Non-Small Cell Lung Cancer Management: Opportunities and Challenges. Biomolecules 2024; 14:1523. [PMID: 39766230 PMCID: PMC11673737 DOI: 10.3390/biom14121523] [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: 10/07/2024] [Revised: 11/07/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
Lung cancer, the leading cause of death worldwide, is associated with the highest morbidity. Non-small cell lung cancer (NSCLC) accounts for 80-85% of lung cancer cases. Advances in the domain of cancer treatment have improved the prognosis and quality of life of patients with metastatic NSCLC. Nevertheless, tumor progression or metastasis owing to treatment failure caused by primary or secondary drug resistance remains the cause of death in the majority of cases. Epithelial-mesenchymal transition (EMT), a vital biological process wherein epithelial cancer cells lose their inherent adhesion and transform into more invasive mesenchymal-like cells, acts as a powerful engine driving tumor metastasis. EMT can also induce immunosuppression in the tumor environment, thereby promoting cancer development and poor prognosis among patients with NSCLC. This review aims to elucidate the effect of EMT on metastasis and the tumor immune microenvironment. Furthermore, it explores the possible roles of EMT inhibition in improving the treatment efficacy of NSCLC. Targeting EMT may be an ideal mechanism to inhibit tumor growth and progression at multiple steps.
Collapse
Affiliation(s)
- Yunyao Ye
- Department of Oncology, Taizhou People’s Hospital Affiliated to Nanjing Medical University, Taizhou 225300, China; (Y.Y.); (S.Y.); (S.Z.); (X.S.)
| | - Shanxun Yu
- Department of Oncology, Taizhou People’s Hospital Affiliated to Nanjing Medical University, Taizhou 225300, China; (Y.Y.); (S.Y.); (S.Z.); (X.S.)
| | - Ting Guo
- Central Lab, Taizhou People’s Hospital Affiliated to Nanjing Medical University, Taizhou 225300, China;
| | - Sihui Zhang
- Department of Oncology, Taizhou People’s Hospital Affiliated to Nanjing Medical University, Taizhou 225300, China; (Y.Y.); (S.Y.); (S.Z.); (X.S.)
| | - Xiaozhou Shen
- Department of Oncology, Taizhou People’s Hospital Affiliated to Nanjing Medical University, Taizhou 225300, China; (Y.Y.); (S.Y.); (S.Z.); (X.S.)
| | - Gaohua Han
- Department of Oncology, Taizhou People’s Hospital Affiliated to Nanjing Medical University, Taizhou 225300, China; (Y.Y.); (S.Y.); (S.Z.); (X.S.)
| |
Collapse
|
50
|
Li AL, Sugiura K, Nishiwaki N, Suzuki K, Sadeghian D, Zhao J, Maitra A, Falvo D, Chandwani R, Pitarresi JR, Sims PA, Rustgi AK. FRA1 controls acinar cell plasticity during murine Kras G12D-induced pancreatic acinar to ductal metaplasia. Dev Cell 2024; 59:3025-3042.e7. [PMID: 39178842 PMCID: PMC11576252 DOI: 10.1016/j.devcel.2024.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 04/17/2024] [Accepted: 07/30/2024] [Indexed: 08/26/2024]
Abstract
Acinar cells have been proposed as a cell-of-origin for pancreatic ductal adenocarcinoma (PDAC) after undergoing acinar-to-ductal metaplasia (ADM). ADM can be triggered by pancreatitis, causing acinar cells to de-differentiate to a ductal-like state. We identify FRA1 (gene name Fosl1) as the most active transcription factor during KrasG12D acute pancreatitis-mediated injury, and we have elucidated a functional role of FRA1 by generating an acinar-specific Fosl1 knockout mouse expressing KrasG12D. Using a gene regulatory network and pseudotime trajectory inferred from single-nuclei ATAC-seq and bulk RNA sequencing (RNA-seq), we hypothesized a regulatory model of the acinar-ADM-pancreatic intraepithelial neoplasia (PanIN) continuum and experimentally validated that Fosl1 knockout mice are delayed in the onset of ADM and neoplastic transformation. Our study also identifies that pro-inflammatory cytokines, such as granulocyte colony stimulating factor (G-CSF), can regulate FRA1 activity to modulate ADM. Our findings identify that FRA1 is a mediator of acinar cell plasticity and is critical for acinar cell de-differentiation and transformation.
Collapse
Affiliation(s)
- Alina L Li
- Divison of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kensuke Sugiura
- Divison of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Noriyuki Nishiwaki
- Divison of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kensuke Suzuki
- Divison of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of General Surgery, Chiba University, Chiba 260-0856, Japan
| | - Dorsay Sadeghian
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Sheikh Ahmed Pancreatic Cancer Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jun Zhao
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Sheikh Ahmed Pancreatic Cancer Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anirban Maitra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Sheikh Ahmed Pancreatic Cancer Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - David Falvo
- Department of Surgery and of Cell and Developmental Biology, Meyer Cancer Center, Weill-Cornell Medicine, New York, NY 10065, USA
| | - Rohit Chandwani
- Department of Surgery and of Cell and Developmental Biology, Meyer Cancer Center, Weill-Cornell Medicine, New York, NY 10065, USA
| | - Jason R Pitarresi
- Division of Hematology-Oncology, Department of Medicine, University of Massachusetts Chan School of Medicine, Worchester, MA 01655, USA
| | - Peter A Sims
- Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Anil K Rustgi
- Divison of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|