1
|
Bräutigam K, Skok K, Szymonski K, Rift CV, Karamitopoulou E. Tumor immune microenvironment in pancreatic ductal adenocarcinoma revisited - Exploring the "Space". Cancer Lett 2025; 622:217699. [PMID: 40204149 DOI: 10.1016/j.canlet.2025.217699] [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: 12/21/2024] [Revised: 03/24/2025] [Accepted: 04/04/2025] [Indexed: 04/11/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most deadly malignancies with a highly immunosuppressive tumor immune microenvironment (TIME) that hinders effective therapy. PDAC is characterized by significant heterogeneity in immune cell composition, spatial distribution and activation states, which impacts tumor progression and treatment response. Tumour-infiltrating lymphocytes (TILs), including CD4+ T-helper cells, CD8+ cytotoxic T-cells and FOXP3+ regulatory T-cells, play a key role in immune regulation, yet PDAC is largely an immunologically "cold" tumour with limited effector T-cell infiltration. The surrounding cellular microenvironment, particularly Cancer Associated Fibroblasts (CAFs) and macrophages, contributes to immune evasion by promoting a fibrotic and desmoplastic barrier that limits TIL infiltration. The prognostic significance of TILs is increasingly recognized, with higher densities correlating with improved survival, whereas regulatory T-cell infiltration and immunosuppressive stromal interactions are associated with poor outcomes. Emerging therapeutic strategies targeting the TIME (e.g., CAFs), immune checkpoint inhibitors, and TIL-based therapies offer the potential to overcome resistance. Future research must focus on optimizing immunotherapy strategies and unravelling the complex stromal-immune interactions to improve clinical translation.
Collapse
Affiliation(s)
- Konstantin Bräutigam
- Institute of Cancer Research, Centre for Evolution and Cancer, London, SM2 5NG, United Kingdom; Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland.
| | - Kristijan Skok
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria; Institute of Biomedical Sciences, Medical Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Krzysztof Szymonski
- Department of Pathomorphology, Jagiellonian University Medical College, Krakow, Poland
| | | | - Eva Karamitopoulou
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
2
|
Guan X, Mu Y, Jin X, Wang C. Integrated multi-omics profiling to establish an IGFBP-based prognostic score for pancreatic ductal adenocarcinoma: unraveling prognostic biomarkers, immune microenvironment crosstalk, and therapeutic implications. Front Immunol 2025; 16:1600527. [PMID: 40443651 PMCID: PMC12119506 DOI: 10.3389/fimmu.2025.1600527] [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: 03/26/2025] [Accepted: 04/24/2025] [Indexed: 06/02/2025] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is accompanied by endocrine dysfunction, particularly involving dysregulation of the insulin and insulin-like growth factor (IGF) signaling pathways. Clinical manifestations such as hyperglycemia and insulin resistance are common and have been linked to aberrant expression of insulin-like growth factor-binding proteins (IGFBPs). However, the specific roles and mechanisms of IGFBP family genes in PDAC remain unclear. Method We conducted a multi-dimensional integrative analysis using publicly available PDAC cohorts, stratifying patients based on IGFBP gene expression profiles. A prognostic model was constructed to classify patients into risk groups. To explore the biological mechanisms underlying IGFBP involvement in PDAC, we further incorporated single-cell transcriptomic sequencing and spatial transcriptomic data to investigate the relationship between IGFBP expression and the tumor immune microenvironment. Result Our prognostic model effectively stratified PDAC patients into distinct risk categories with significant survival differences. High-risk patients demonstrated specific IGFBP expression patterns associated with aggressive tumor biology. Single-cell and spatial transcriptomic analyses revealed that IGFBP family genes modulate immune cell infiltration and spatial immune heterogeneity within the tumor microenvironment. Conclusion This study identified the IGFBP family genes as key modulators of PDAC progression and immune landscape remodeling. These findings supported the potential of IGFBP family genes as prognostic biomarkers and therapeutic targets, offering new insights into PDAC biology and opportunities for personalized treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Chengfeng Wang
- State Key Lab of Molecular Oncology and Department of Pancreatic and Gastric Surgery,
National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
3
|
Pan D, Li X, Qiao X, Wang Q. Immunosuppressive tumor microenvironment in pancreatic cancer: mechanisms and therapeutic targets. Front Immunol 2025; 16:1582305. [PMID: 40443678 PMCID: PMC12119487 DOI: 10.3389/fimmu.2025.1582305] [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: 02/24/2025] [Accepted: 04/17/2025] [Indexed: 06/02/2025] Open
Abstract
Pancreatic cancer is projected to become the second leading cause of cancer-related death by 2030. Conventional interventions including surgery, radiotherapy, and chemotherapy provide only modest survival benefits, underscoring an urgent need for more effective therapies. Although immunotherapy has revolutionized the management of several solid tumors, its clinical benefit in pancreatic cancer has so far been disappointing. Mounting evidence indicates that a highly immunosuppressive tumor microenvironment (TME), dominated by tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs), drives immune evasion, tumor progression, metastasis, and chemoresistance through complex cytokine and chemokine networks. This review summarizes current knowledge of these immunosuppressive mechanisms and provides emerging strategies aimed at re-educating or depleting these cellular constituents to enhance the efficacy of immunotherapy in pancreatic cancer.
Collapse
Affiliation(s)
- Da Pan
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou, China
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, China
| | - Xinyue Li
- First College for Clinical Medicine, Xuzhou Medical University, Jiangsu, Xuzhou, China
| | - Xiao Qiao
- Department of Gastroenterology, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian, China
| | - Qiqi Wang
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou, China
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
4
|
Cramer GM, Davis RW, Papasavvas E, Klampatsa A, Miller JM, Carter S, Ikpe R, Yuan M, Widura S, Majumdar RS, McNulty S, Putt M, Kossenkov AV, Montaner LJ, Singhal S, Moon EK, Albelda SM, Cengel KA, Busch TM. PD-1 Blockade Mitigates Surgery-Induced Immunosuppression and Increases the Efficacy of Photodynamic Therapy for Pleural Mesothelioma. CANCER RESEARCH COMMUNICATIONS 2025; 5:841-856. [PMID: 40331601 PMCID: PMC12099492 DOI: 10.1158/2767-9764.crc-24-0571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/25/2025] [Accepted: 05/02/2025] [Indexed: 05/08/2025]
Abstract
Lung-sparing radical pleurectomy with intraoperative photodynamic therapy (PDT) demonstrates remarkable survival for patients with pleural mesothelioma. Nevertheless, most patients treated with this multimodal approach will develop local tumor recurrence. An understanding of potential causes of treatment failure is central to developing mitigation strategies. Surgery importantly reduces disease burden but also produces tumor-promoting inflammation, as demonstrated through transcriptomic analysis of pleural mesothelioma specimens. Using preclinical models in the setting of combination therapy, we separated the benefit of surgical resection from its counterproductive effects on therapeutic outcome. Specifically, we evaluated mechanisms by which surgically induced inflammation can be therapy-limiting in a murine model of tumor incision (TI) introduced by a surgical cut across the tumor. In this TI model, we identified distinct TI-altered patterns in innate and adaptive inflammatory cells in murine mesothelioma tumors, and we studied changes in these patterns with the addition of PDT. TI introduction of an immunosuppressive environment is established via upregulation of PD-1/PD-L1 expression on tumor cells, T cells, and myeloid cells that is partially resolved by PDT. Immune dysfunction is further mitigated by the addition of PD-1 blockade, leading to curative potential in a process that requires Ly6G+ neutrophils and CD8+ T cells. Overall, these studies suggest that, without PDT, surgical modulation of immune cell trafficking and functionality leads to systemic immunosuppression. This immunosuppressive state potentially interferes with the generation of antitumor immunity by PDT. However, targeted inhibition of surgery-induced signaling in the PD-l/PD-L1 pathway counteracts surgery's immunosuppressive outcomes to enhance PDT efficacy in the intraoperative setting. SIGNIFICANCE Surgery combined with PDT extends survival for patients with mesothelioma, but these patients are still at risk for tumor recurrence, in part due to the immunosuppressive effects of surgery. We find, in a mouse model, that combining surgery, PDT, and immune checkpoint blockade maximizes the efficacy of these therapies.
Collapse
Affiliation(s)
- Gwendolyn M. Cramer
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Richard W. Davis
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | | | - Astero Klampatsa
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joann M. Miller
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Shirron Carter
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Ruth Ikpe
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Min Yuan
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Sandy Widura
- The Wistar Institute, Philadelphia, Pennsylvania
| | | | - Sally McNulty
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Mary Putt
- Department of Biostatistics, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | - Sunil Singhal
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Edmund K. Moon
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steven M. Albelda
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Keith A. Cengel
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Theresa M. Busch
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Wang N, Zhu M, Jiang Y, Zhang D, Lin J, Wu J, Lin N, Gao J. Neutrophil-Mediated Tumor Discrimination Biomimetic Nanodevice for Precise Tumor Eradication and Metastasis Cascade Perturbing. ACS APPLIED MATERIALS & INTERFACES 2025; 17:23680-23690. [PMID: 40219943 DOI: 10.1021/acsami.5c01866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2025]
Abstract
The deficient discrimination of tumor cells, limited accumulation of therapeutic agents in tumor foci, and undesirable tumor metastasis result in compromised therapeutic efficacy in antitumor therapy. Neutrophils, the most abundant circulating leukocytes, are key players in tumor progression and have a high affinity to tumors. Herein, a biomimetic tumor discrimination nanodevice (NL-FSB) is developed by harboring a pH-sensitive Fenton agent (FSB) in an activated neutrophil membrane-incorporated liposome for tumor-specific ablation. Inheriting the biointerfacing properties of neutrophils, NL-FSB is endowed with high affinity to tumors. On one hand, NL-FSB can be recruited to acidic tumor sites mediated by chemotaxis attraction and selectively generate reactive oxygen species via amplified Fenton chemical reaction for specific tumor eradication. On the other hand, the biomimetic NL-FSB can also target and bind tumor vascular endothelium or circulating tumor cells (CTCs) in circulation, executing pseudo escort to perturb CTC-neutrophil cluster formation and tumor metastasis cascade. Unprecedentedly, the neutrophil-mediated nanoagent can effectively inhibit the already-formed tumor and prevent tumor metastasis with high specificity. Our study represents a promising yet simple strategy for tumor-specific killing and metastasis cascade blockage via a nanobioengineering functionalization strategy.
Collapse
Affiliation(s)
- Nan Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
- Department of Pharmacy, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Mingjian Zhu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yiming Jiang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Daoming Zhang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiachen Lin
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiahe Wu
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
- Department of Pharmacy, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Nengming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
- Zheiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou 310058, China
- Department of Pharmacy, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Jianqing Gao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Zheiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
8
|
Lloyd EG, Jihad M, Manansala JS, Li W, Cheng PS, Mucciolo G, Zaccaria M, Teles SP, Henríquez JA, Harish S, Brais R, Ashworth S, Luo W, Johnson PM, Veghini L, Vallespinos M, Corbo V, Biffi G. SMAD4 and KRAS Status Shapes Cancer Cell-Stromal Cross-Talk and Therapeutic Response in Pancreatic Cancer. Cancer Res 2025; 85:1368-1389. [PMID: 39841099 PMCID: PMC7617379 DOI: 10.1158/0008-5472.can-24-2330] [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: 10/21/2024] [Revised: 12/09/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) contains an extensive stroma that modulates response to therapy, contributing to the dismal prognosis associated with this cancer. Evidence suggests that PDAC stromal composition is shaped by mutations within malignant cells, but most previous work has focused on preclinical models driven by KrasG12D and mutant Trp53. Elucidation of the contribution of additional known oncogenic drivers, including KrasG12V mutation and Smad4 loss, is needed to increase the understanding of malignant cell-stromal cell cross-talk in PDAC. In this study, we used single-cell RNA sequencing to analyze the cellular landscape of Trp53-mutant mouse models driven by KrasG12D or KrasG12V, in which Smad4 was wild type or deleted. KrasG12DSmad4-deleted PDAC developed a fibro-inflammatory rich stroma with increased malignant JAK/STAT cell signaling and enhanced therapeutic response to JAK/STAT inhibition. SMAD4 loss in KrasG12V PDAC differently altered the tumor microenvironment compared with KrasG12D PDAC, and the malignant compartment lacked JAK/STAT signaling dependency. Thus, malignant cell genotype affects cancer cell and stromal cell phenotypes in PDAC, directly affecting therapeutic efficacy. Significance: SMAD4 loss differentially impacts malignant cell-stromal cell signaling and treatment sensitivity of pancreatic tumors driven by KRASG12D or KRASG12V, highlighting the importance of understanding genotype-phenotype relationships for precision therapy.
Collapse
Affiliation(s)
- Eloise G. Lloyd
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Muntadher Jihad
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Judhell S. Manansala
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Wenlong Li
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Priscilla S.W. Cheng
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Gianluca Mucciolo
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Marta Zaccaria
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Sara Pinto Teles
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Joaquín Araos Henríquez
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Sneha Harish
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Rebecca Brais
- Histopathology, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke’s Hospital, Cambridge, UK
| | - Sally Ashworth
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Weike Luo
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Paul M. Johnson
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Lisa Veghini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Mireia Vallespinos
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
- ARC-Net Research Centre, University of Verona, Verona, Italy
| | - Giulia Biffi
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| |
Collapse
|
9
|
Huang C, Qiu H, Xu C, Tan Z, Jin M, Hu J, Huang Z, Zhou Y, Ge S, Hu X. Downregulation of tropomyosin 2 promotes the progression of lung adenocarcinoma by regulating neutrophil infiltration through neutrophil elastase. Cell Death Dis 2025; 16:264. [PMID: 40199876 PMCID: PMC11978998 DOI: 10.1038/s41419-025-07531-1] [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: 11/12/2024] [Revised: 02/12/2025] [Accepted: 03/12/2025] [Indexed: 04/10/2025]
Abstract
Lung adenocarcinoma (LUAD) is a common malignant tumor in the lung that seriously endangers the health of people worldwide. The neutrophil-associated inflammatory microenvironment contributes to the activation of tumor cells. In this study, we report a role of tumor-associated neutrophils (TANs) promote tumor progression of LUAD by crosstalk between neutrophils and tumor cells. Mechanistically, in co-culture with tumor cells, downregulation of TPM2 on tumor cells increases neutrophil elastase (ELANE) levels in neutrophils regulated by p38/ MAPK signaling activation, and ELANE promotes tumor cell progression through the Hippo pathway. Furthermore, downregulation of TPM2 activates ELANE of neutrophils to facilitate ERK1/2 activation, thus enhancing IL1β and IL8 secretion for chemoattraction of more neutrophils to tumor microenvironment. The new studies identify an accomplice role for the interaction between TPM2 and ELANE in promoting LUAD progression and provide potential strategies in the prevention and/or treatment of LUAD and other cancers.
Collapse
Affiliation(s)
- Caixiu Huang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Hao Qiu
- The First People's Hospital of Changde City, Changde, PR China
| | - Changting Xu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Zilong Tan
- Nanchang University Second Affiliated Hospital, Nanchang, PR China
| | - Mei Jin
- Pingxiang People's Hospital, Pingxiang, PR China
| | - Jing Hu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | | | - Yuwei Zhou
- Nanchang University Second Affiliated Hospital, Nanchang, PR China
| | - Shengyou Ge
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China.
| | - Xiaoyuan Hu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China.
| |
Collapse
|
10
|
Breaza GM, Closca RM, Cindrea AC, Hut FE, Cretu O, Sima LV, Rakitovan M, Zara F. Immunohistochemical Evaluation of the Tumor Immune Microenvironment in Pancreatic Ductal Adenocarcinoma. Diagnostics (Basel) 2025; 15:646. [PMID: 40075893 PMCID: PMC11899021 DOI: 10.3390/diagnostics15050646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Background: Pancreatic ductal adenocarcinoma is an aggressive neoplasm with a complex carcinogenesis process that must be understood through the interactions between tumor cells and tumor microenvironment cells. Methods: This study was retrospective with a chronological extension period of 16 years and included 56 cases of pancreatic ductal adenocarcinoma. This study identified, quantified, and correlated the cells of the tumor immune microenvironment in pancreatic ductal adenocarcinoma with major prognostic factors as well as overall survival, using an extensive panel of immunohistochemical markers. Results: Three tumor immunotypes were identified: subtype A (hot immunotype), subtype B (intermediate immunotype), and subtype C (cold immunotype). Patients with immunotype C exhibit considerably higher rates of both pancreatic fistulas and acute pancreatitis. Immunotypes B and C significantly increased the risk of this complication by factors of 3.68 (p = 0.002) and 3.94 (p = 0.001), respectively. The estimated probabilities of fistula formation for each immunotype are as follows: 2.5% for immunotype A, 25% for immunotype B, and 28% for immunotype C. There was a statistically significant difference in median survival times according to tumor immunotype (p < 0.001). Specifically, patients with immunotype C tumors had a median survival time of only 120.5 days, compared to 553.5 days for those with immunotype A and 331.5 for immunotype B tumors. Conclusions: The identification of the immunotype of pancreatic ductal adenocarcinoma can be a predictive factor for the occurrence of complications such as pancreatic fistula as well as for overall survival.
Collapse
Affiliation(s)
- Gelu Mihai Breaza
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
| | - Raluca Maria Closca
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- Department of Pathology, Emergency City Hospital, 300254 Timisoara, Romania
| | - Alexandru Cristian Cindrea
- Department of Surgery, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania;
- Emergency Department, Emergency Clinical Municipal Hospital, 300079 Timisoara, Romania
| | - Florin Emil Hut
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
- Center for Hepato-Bilio-Pancreatic Surgery, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania
| | - Octavian Cretu
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
| | - Laurentiu Vasile Sima
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
| | - Marina Rakitovan
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- Oro-Maxillo-Facial Surgery Clinic, Emergency City Hospital, 300062 Timisoara, Romania
| | - Flavia Zara
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- Department of Pathology, Emergency City Hospital, 300254 Timisoara, Romania
| |
Collapse
|
11
|
Dolskii A, dos Santos SAA, Andrake M, Franco-Barraza J, Dunbrack RL, Cukierman E. Exploring the potential role of palladin in modulating human CAF/ECM functional units. Cytoskeleton (Hoboken) 2025; 82:175-185. [PMID: 39239855 PMCID: PMC11882928 DOI: 10.1002/cm.21926] [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: 06/09/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024]
Abstract
Fibroblasts, crucial for maintaining tissue homeostasis, significantly shape the tumor microenvironment (TME). In pancreatic cancer, a highly aggressive malignancy, cancer-associated fibroblast (CAF)/extracellular matrix (ECM) units dominate the TME, influencing tumor initiation, progression, and treatment responses. Palladin, an actin-associated protein, is vital for fibroblast structural integrity and activation, playing a key role in CAF/ECM functionality. Palladin interacts with cytoskeletal proteins such as alpha-actinin (α-Act) and can therefore regulate other proteins like syndecans, modulating cytoskeletal features, cell adhesion, integrin recycling, and signaling. In this review, we propose that targeting the palladin/α-Act/syndecan interaction network could modulate CAF/ECM units, potentially shifting the TME from a tumor-promoting to a tumor-suppressive state. In silico data and reported studies to suggest that stabilizing palladin-α-Act interactions, via excess palladin, influences syndecan functions; potentially modulating integrin endocytosis via syndecan engagement with protein kinase C alpha as opposed to syndecan binding to α-Act. This mechanism can then affect the distribution of active α5β1-integrin between the plasma membrane and known intracellular vesicular compartments, thereby influencing the tumor-suppressive versus tumor-promoting functions of CAF/ECM units. Understanding these interactions offers likely future therapeutic avenues for stroma normalization in pancreatic and other cancers, aiming to inhibit tumor progression and improve future treatment outcomes.
Collapse
Affiliation(s)
| | | | - Mark Andrake
- Cancer Signaling and Microenvironment, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Lewis Katz School of Medicine, Temple Health, Philadelphia, PA
| | - Janusz Franco-Barraza
- Cancer Signaling and Microenvironment, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Lewis Katz School of Medicine, Temple Health, Philadelphia, PA
| | - Roland L. Dunbrack
- Cancer Signaling and Microenvironment, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Lewis Katz School of Medicine, Temple Health, Philadelphia, PA
| | - Edna Cukierman
- Cancer Signaling and Microenvironment, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Lewis Katz School of Medicine, Temple Health, Philadelphia, PA
| |
Collapse
|
12
|
Gabashvili AN, Vasiukova AA, Rakitina AS, Garanina AS. The Issue on Dualistic Role of Neutrophils in Carcinogenesis and Their Possible Use for Treatment of Malignant Neoplasms. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:303-320. [PMID: 40367075 DOI: 10.1134/s000629792460368x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/04/2024] [Accepted: 12/24/2024] [Indexed: 05/16/2025]
Abstract
Neutrophils are phagocytic leukocytes of the myeloid series, which are the most common myeloid cells in human blood, normally accounting from 65 to 80% of all circulating leukocytes. Over the years of investigation of these cells, more and more evidence has emerged indicating functional plasticity of neutrophils and their ambiguous role in the tumor development. Similarly to the M1/M2 classification of macrophages, the N1/N2 paradigm could be applied to neutrophils, where N1-neutrophils exhibit tumor-suppressive properties, and N2-neutrophils contribute to tumor development and immune suppression. An important natural feature of neutrophils is their mobility and ability to overcome physical barriers, thus these cells, as well as their vesicles and membranes, could be used to deliver therapeutic drugs to tumor cells. In addition, neutrophils themselves could be activated and mobilized to fight the tumor. This review describes current state of research on the role of neutrophils in carcinogenesis, as well as possible approaches of using these cells and their derivatives as systems for targeted delivery of therapeutic drugs for treatment of malignant neoplasms.
Collapse
Affiliation(s)
- Anna N Gabashvili
- Prokhorov General Physics Institute, Russian Academy of Sciences, Moscow, 119991, Russia
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Anastasiia A Vasiukova
- Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, 119034, Russia
| | - Aleksandra S Rakitina
- Prokhorov General Physics Institute, Russian Academy of Sciences, Moscow, 119991, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russia
| | | |
Collapse
|
13
|
Kwantwi LB. SLAM family-mediated crosstalk between tumor and immune cells in the tumor microenvironment: a promising biomarker and a potential therapeutic target for immune checkpoint therapies. Clin Transl Oncol 2025; 27:901-908. [PMID: 39212911 DOI: 10.1007/s12094-024-03675-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
Immune cells infiltrating the tumor microenvironment are physiologically important in controlling cancers. However, emerging studies have shown that cancer cells can evade immune surveillance and establish a balance in which these immune cells support tumor progression and therapeutic resistance. The signaling lymphocytic activation molecule family members have been recognized as mediators of tumor microenvironment interactions, and a promising therapeutic target for cancer immunotherapy. This review is focused on the role of SLAM family in tumor and immune cell interactions and discusses how such crosstalk affects tumor behavior. This will shed insight into the next step toward improving cancer immunotherapy.
Collapse
Affiliation(s)
- Louis Boafo Kwantwi
- Department of Anatomy and Neurobiology, College of Medicine, Northeast Ohio Medical University, Rootstown, OH, 44272, USA.
| |
Collapse
|
14
|
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
|
15
|
Giurini EF, Ralph O, Pappas SG, Gupta KH. Looking Beyond Checkpoint Inhibitor Monotherapy: Uncovering New Frontiers for Pancreatic Cancer Immunotherapy. Mol Cancer Ther 2025; 24:18-32. [PMID: 39311547 DOI: 10.1158/1535-7163.mct-24-0311] [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: 05/03/2024] [Revised: 08/01/2024] [Accepted: 09/09/2024] [Indexed: 01/03/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) stands out as one of the most aggressive and challenging tumors, characterized by a bleak prognosis with a mere 11% survival rate over 5 years in the United States. Its formidable nature is primarily attributed to its highly aggressive behavior and poor response to existing therapies. PDAC, being notably resistant to immune interventions, presents a significant obstacle in treatment strategies. While immune checkpoint inhibitor therapies have revolutionized outcomes for various cancers, their efficacy in PDAC remains exceedingly low, benefiting less than 1% of patients. The consistent failure of these therapies in PDAC has prompted intensive investigation, particularly at the preclinical level, to unravel the intricate mechanisms of resistance inherent in this cancer type. This pursuit aims to pave the way for the development of novel immunotherapeutic strategies tailored to the distinct characteristics of PDAC. This review endeavors to provide a comprehensive exploration of these emerging immunotherapy approaches in PDAC, with a specific emphasis on elucidating their underlying immunological mechanisms. Additionally, it sheds light on the recently identified factors driving resistance to immunotherapy and evasion of the immune system in PDAC, offering insights beyond the conventional drivers that have been extensively studied.
Collapse
Affiliation(s)
- Eileena F Giurini
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, Illinois
| | - Oliver Ralph
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, Illinois
| | - Sam G Pappas
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, Illinois
| | - Kajal H Gupta
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, Illinois
- Division of Pediatric Surgery, Department of Surgery, Rush University Medical Center, Chicago, Illinois
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| |
Collapse
|
16
|
Dai W, Zhou X, Zhao J, Lei L, Huang Y, Jia F, Tang Z, Ji J, Jin Q. Tumor microenvironment-modulated nanoparticles with cascade energy transfer as internal light sources for photodynamic therapy of deep-seated tumors. Biomaterials 2025; 312:122743. [PMID: 39111233 DOI: 10.1016/j.biomaterials.2024.122743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024]
Abstract
Photodynamic therapy (PDT) is an appealing modality for cancer treatments. However, the limited tissue penetration depth of external-excitation light makes PDT impossible in treating deep-seated tumors. Meanwhile, tumor hypoxia and intracellular reductive microenvironment restrain the generation of reactive oxygen species (ROS). To overcome these limitations, a tumor-targeted self-illuminating supramolecular nanoparticle T-NPCe6-L-N is proposed by integrating photosensitizer Ce6 with luminol and nitric oxide (NO) for chemiluminescence resonance energy transfer (CRET)-activated PDT. The high H2O2 level in tumor can trigger chemiluminescence of luminol to realize CRET-activated PDT without exposure of external light. Meanwhile, the released NO significantly relieves tumor hypoxia via vascular normalization and reduces intracellular reductive GSH level, further enhancing ROS abundance. Importantly, due to the different ROS levels between cancer cells and normal cells, T-NPCe6-L-N can selectively trigger PDT in cancer cells while sparing normal cells, which ensured low side effect. The combination of CRET-based photosensitizer-activation and tumor microenvironment modulation overcomes the innate challenges of conventional PDT, demonstrating efficient inhibition of orthotopic and metastatic tumors on mice. It also provoked potent immunogenic cell death to ensure long-term suppression effects. The proof-of-concept research proved as a new strategy to solve the dilemma of PDT in treatment of deep-seated tumors.
Collapse
Affiliation(s)
- Wenbin Dai
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, PR China
| | - Xianchi Zhou
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, PR China
| | - Jinchao Zhao
- Department of Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, PR China
| | - Lei Lei
- Department of Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, PR China
| | - Yue Huang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, PR China
| | - Fan Jia
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, PR China.
| | - Zhe Tang
- Department of Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, PR China; Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, PR China
| | - Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, PR China.
| |
Collapse
|
17
|
Yao J, Ji L, Wang G, Ding J. Effect of neutrophils on tumor immunity and immunotherapy resistance with underlying mechanisms. Cancer Commun (Lond) 2025; 45:15-42. [PMID: 39485719 PMCID: PMC11758154 DOI: 10.1002/cac2.12613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 09/08/2024] [Accepted: 09/17/2024] [Indexed: 11/03/2024] Open
Abstract
Neutrophils are key mediators of the immune response and play essential roles in the development of tumors and immune evasion. Emerging studies indicate that neutrophils also play a critical role in the immunotherapy resistance in cancer. In this review, firstly, we summarize the novel classification and phenotypes of neutrophils and describe the regulatory relationships between neutrophils and tumor metabolism, flora microecology, neuroendocrine and tumor therapy from a new perspective. Secondly, we review the mechanisms by which neutrophils affect drug resistance in tumor immunotherapy from the aspects of the immune microenvironment, tumor antigens, and epigenetics. Finally, we propose several promising strategies for overcoming tumor immunotherapy resistance by targeting neutrophils and provide new research ideas in this area.
Collapse
Affiliation(s)
- Jiali Yao
- Clinical Cancer InstituteCenter for Translational MedicineNaval Medical UniversityShanghaiChina
| | - Linlin Ji
- Clinical Cancer InstituteCenter for Translational MedicineNaval Medical UniversityShanghaiChina
| | - Guang Wang
- Clinical Cancer InstituteCenter for Translational MedicineNaval Medical UniversityShanghaiChina
| | - Jin Ding
- Clinical Cancer InstituteCenter for Translational MedicineNaval Medical UniversityShanghaiChina
| |
Collapse
|
18
|
Hashimoto A, Hashimoto S. Plasticity and Tumor Microenvironment in Pancreatic Cancer: Genetic, Metabolic, and Immune Perspectives. Cancers (Basel) 2024; 16:4094. [PMID: 39682280 DOI: 10.3390/cancers16234094] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer has long been believed to be a genetic disease caused by the accumulation of mutations in key genes involved in cellular processes. However, recent advances in sequencing technology have demonstrated that cells with cancer driver mutations are also present in normal tissues in response to aging, environmental damage, and chronic inflammation, suggesting that not only intrinsic factors within cancer cells, but also environmental alterations are important key factors in cancer development and progression. Pancreatic cancer tissue is mostly comprised of stromal cells and immune cells. The desmoplasmic microenvironment characteristic of pancreatic cancer is hypoxic and hypotrophic. Pancreatic cancer cells may adapt to this environment by rewiring their metabolism through epigenomic changes, enhancing intrinsic plasticity, creating an acidic and immunosuppressive tumor microenvironment, and inducing noncancerous cells to become tumor-promoting. In addition, pancreatic cancer has often metastasized to local and distant sites by the time of diagnosis, suggesting that a similar mechanism is operating from the precancerous stage. Here, we review key recent findings on how pancreatic cancers acquire plasticity, undergo metabolic reprogramming, and promote immunosuppressive microenvironment formation during their evolution. Furthermore, we present the following two signaling pathways that we have identified: one based on the small G-protein ARF6 driven by KRAS/TP53 mutations, and the other based on the RNA-binding protein Arid5a mediated by inflammatory cytokines, which promote both metabolic reprogramming and immune evasion in pancreatic cancer. Finally, the striking diversity among pancreatic cancers in the relative importance of mutational burden and the tumor microenvironment, their clinical relevance, and the potential for novel therapeutic strategies will be discussed.
Collapse
Affiliation(s)
- Ari Hashimoto
- Department of Molecular Biology, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Shigeru Hashimoto
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0818, Japan
| |
Collapse
|
19
|
Xu Y, Wang X, Li Y, Mao Y, Su Y, Mao Y, Yang Y, Gao W, Fu C, Chen W, Ye X, Liang F, Bai P, Sun Y, Li S, Xu R, Tian R. Multimodal single cell-resolved spatial proteomics reveal pancreatic tumor heterogeneity. Nat Commun 2024; 15:10100. [PMID: 39572534 PMCID: PMC11582669 DOI: 10.1038/s41467-024-54438-0] [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: 10/20/2023] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
Despite the advances in antibody-guided cell typing and mass spectrometry-based proteomics, their integration is hindered by challenges for processing rare cells in the heterogeneous tissue context. Here, we introduce Spatial and Cell-type Proteomics (SCPro), which combines multiplexed imaging and flow cytometry with ion exchange-based protein aggregation capture technology to characterize spatial proteome heterogeneity with single-cell resolution. The SCPro is employed to explore the pancreatic tumor microenvironment and reveals the spatial alternations of over 5000 proteins by automatically dissecting up to 100 single cells guided by multi-color imaging of centimeter-scale formalin-fixed, paraffin-embedded tissue slide. To enhance cell-type resolution, we characterize the proteome of 14 different cell types by sorting up to 1000 cells from the same tumor, which allows us to deconvolute the spatial distribution of immune cell subtypes and leads to the discovery of subtypes of regulatory T cells. Together, the SCPro provides a multimodal spatial proteomics approach for profiling tissue proteome heterogeneity.
Collapse
Affiliation(s)
- Yanfen Xu
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Xi Wang
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
- Department of Oncology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Yuan Li
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Yiheng Mao
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Yiran Su
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Yize Mao
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Yun Yang
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Weina Gao
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Changying Fu
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Wendong Chen
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Xueting Ye
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Fuchao Liang
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Panzhu Bai
- Department of System Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Ying Sun
- Department of System Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Shengping Li
- Department of Pancreatobiliary Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Ruilian Xu
- Department of Oncology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Ruijun Tian
- State Key Laboratory of Medical Proteomics and Shenzhen Key Laboratory of Functional Proteomics, Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science and Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
20
|
Pan X, Wang Q, Sun B. Multifaceted roles of neutrophils in tumor microenvironment. Biochim Biophys Acta Rev Cancer 2024; 1879:189231. [PMID: 39615862 DOI: 10.1016/j.bbcan.2024.189231] [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: 06/12/2024] [Revised: 11/20/2024] [Accepted: 11/24/2024] [Indexed: 12/14/2024]
Abstract
Neutrophils, the most abundant leukocyte population in circulation, play a crucial role in detecting and responding to foreign cells, such as pathogens and tumor cells. However, the impact of neutrophils on cancer pathogenesis has been overlooked because of their short lifespan, terminal differentiation, and limited transcriptional activity. Within the tumor microenvironment (TME), neutrophils can be influenced by tumor cells or other stromal cells to acquire either protumor or antitumor properties via the cytokine environment. Despite progress in neutrophil-related research, a comprehensive understanding of tissue-specific neutrophil diversity and adaptability in the TME is still lacking, which poses a significant obstacle to the development of neutrophil-based cancer therapies. This review evaluated the current studies on the dual roles of neutrophils in cancer progression, emphasizing their importance in predicting clinical outcomes, and explored various approaches for targeting neutrophils in cancer treatment, including their potential synergy with cancer immunotherapy.
Collapse
Affiliation(s)
- Xueyin Pan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Innovative Institute of Tumor Immunity and Medicine (ITIM), Anhui Provincial Innovation Institute for Pharmaceutical Basic Research, Hefei, Anhui, China; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China
| | - Qiang Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Innovative Institute of Tumor Immunity and Medicine (ITIM), Anhui Provincial Innovation Institute for Pharmaceutical Basic Research, Hefei, Anhui, China; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China.
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Innovative Institute of Tumor Immunity and Medicine (ITIM), Anhui Provincial Innovation Institute for Pharmaceutical Basic Research, Hefei, Anhui, China; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China.
| |
Collapse
|
21
|
Guo M, Sheng W, Yuan X, Wang X. Neutrophils as promising therapeutic targets in pancreatic cancer liver metastasis. Int Immunopharmacol 2024; 140:112888. [PMID: 39133956 DOI: 10.1016/j.intimp.2024.112888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/01/2024] [Accepted: 08/04/2024] [Indexed: 09/01/2024]
Abstract
Pancreatic cancer is characterized by an extremely poor prognosis and presents significant treatment challenges. Liver metastasis is the leading cause of death in patients with pancreatic cancer. Recent studies have highlighted the significant impact of neutrophils on tumor occurrence and progression, as well as their crucial role in the pancreatic cancer tumor microenvironment. Neutrophil infiltration plays a critical role in the progression and prognosis of pancreatic cancer. Neutrophils contribute to pancreatic cancer liver metastasis through various mechanisms, including angiogenesis, immune suppression, immune evasion, and epithelial-mesenchymal transition (EMT). Therefore, targeting neutrophils holds promise as an important therapeutic strategy for inhibiting pancreatic cancer liver metastasis. This article provides a summary of research findings on the involvement of neutrophils in pancreatic cancer liver metastasis and analyzes their potential as therapeutic targets. This research may provide new insights for the treatment of pancreatic cancer and improve the prognosis of patients with this disease.
Collapse
Affiliation(s)
- Minjie Guo
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Wanying Sheng
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiao Yuan
- Cancer Institute of Jiangsu University, Zhenjiang, China.
| | - Xu Wang
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| |
Collapse
|
22
|
Zhang M, Qin H, Wu Y, Gao Q. Complex role of neutrophils in the tumor microenvironment: an avenue for novel immunotherapies. Cancer Biol Med 2024; 21:j.issn.2095-3941.2024.0192. [PMID: 39297568 PMCID: PMC11523270 DOI: 10.20892/j.issn.2095-3941.2024.0192] [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: 05/24/2024] [Accepted: 08/26/2024] [Indexed: 11/01/2024] Open
Abstract
Neutrophils, which originate from the bone marrow and are characterized by a segmented nucleus and a brief lifespan, have a crucial role in the body's defense against infections and acute inflammation. Recent research has uncovered the complex roles of neutrophils as regulators in tumorigenesis, during which neutrophils exhibit a dualistic nature that promotes or inhibits tumor progression. This adaptability is pivotal within the tumor microenvironment (TME). In this review, we provide a comprehensive characterization of neutrophil plasticity and heterogeneity, aiming to illuminate current research findings and discuss potential therapeutic avenues. By delineating the intricate interplay of neutrophils in the TME, this review further underscores the urgent need to understand the dual functions of neutrophils with particular emphasis on the anti-tumor effects to facilitate the development of effective therapeutic strategies against cancer.
Collapse
Affiliation(s)
- Mao Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Haokai Qin
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Yingcheng Wu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai 200433, China
| |
Collapse
|
23
|
Ju Y, Xu D, Liao MM, Sun Y, Bao WD, Yao F, Ma L. Barriers and opportunities in pancreatic cancer immunotherapy. NPJ Precis Oncol 2024; 8:199. [PMID: 39266715 PMCID: PMC11393360 DOI: 10.1038/s41698-024-00681-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 08/27/2024] [Indexed: 09/14/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents a fatal clinical challenge characterized by a dismal 5-year overall survival rate, primarily due to the lack of early diagnosis and limited therapeutic efficacy. Immunotherapy, a proven success in multiple cancers, has yet to demonstrate significant benefits in PDAC. Recent studies have revealed the immunosuppressive characteristics of the PDAC tumor microenvironment (TME), including immune cells with suppressive properties, desmoplastic stroma, microbiome influences, and PDAC-specific signaling pathways. In this article, we review recent advances in understanding the immunosuppressive TME of PDAC, TME differences among various mouse models of pancreatic cancer, and the mechanisms underlying resistance to immunotherapeutic interventions. Furthermore, we discuss the potential of targeting cancer cell-intrinsic pathways and TME components to sensitize PDAC to immune therapies, providing insights into strategies and future perspectives to break through the barriers in improving pancreatic cancer treatment.
Collapse
Affiliation(s)
- Yixin Ju
- Hubei Hongshan Laboratory, College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Shenzhen, Guangdong, 518000, China
| | - Dongzhi Xu
- Hubei Hongshan Laboratory, College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Shenzhen, Guangdong, 518000, China
| | - Miao-Miao Liao
- Hubei Hongshan Laboratory, College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Yutong Sun
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Wen-Dai Bao
- Hubei Hongshan Laboratory, College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Fan Yao
- Hubei Hongshan Laboratory, College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Shenzhen, Guangdong, 518000, China.
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, Guangdong, 518000, China.
| | - Li Ma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, 77030, USA.
| |
Collapse
|
24
|
Poyia F, Neophytou CM, Christodoulou MI, Papageorgis P. The Role of Tumor Microenvironment in Pancreatic Cancer Immunotherapy: Current Status and Future Perspectives. Int J Mol Sci 2024; 25:9555. [PMID: 39273502 PMCID: PMC11395109 DOI: 10.3390/ijms25179555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Pancreatic cancer comprises different subtypes, where most cases include ductal adenocarcinoma (PDAC). It is one of the deadliest tumor types, with a poor prognosis. In the majority of patients, the disease has already spread by the time of diagnosis, making full recovery unlikely and increasing mortality risk. Despite developments in its detection and management, including chemotherapy, radiotherapy, and targeted therapies as well as advances in immunotherapy, only in about 13% of PDAC patients does the overall survival exceed 5 years. This may be attributed, at least in part, to the highly desmoplastic tumor microenvironment (TME) that acts as a barrier limiting perfusion, drug delivery, and immune cell infiltration and contributes to the establishment of immunologically 'cold' conditions. Therefore, there is an urgent need to unravel the complexity of the TME that promotes PDAC progression and decipher the mechanisms of pancreatic tumors' resistance to immunotherapy. In this review, we provide an overview of the major cellular and non-cellular components of PDAC TME, as well as their biological interplays. We also discuss the current state of PDAC therapeutic treatments and focus on ongoing and future immunotherapy efforts and multimodal treatments aiming at remodeling the TME to improve therapeutic efficacy.
Collapse
Affiliation(s)
- Fotini Poyia
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 2404, Cyprus
| | - Christiana M Neophytou
- Apoptosis and Cancer Chemoresistance Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 2404, Cyprus
| | - Maria-Ioanna Christodoulou
- Tumor Immunology and Biomarkers Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 2404, Cyprus
| | - Panagiotis Papageorgis
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 2404, Cyprus
| |
Collapse
|
25
|
Jin Y, Christenson ES, Zheng L, Li K. Neutrophils in pancreatic ductal adenocarcinoma: bridging preclinical insights to clinical prospects for improved therapeutic strategies. Expert Rev Clin Immunol 2024; 20:945-958. [PMID: 38690749 DOI: 10.1080/1744666x.2024.2348605] [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: 01/20/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy characterized by a dismal five-year survival rate of less than 10%. Neutrophils are key components of the innate immune system, playing a pivotal role in the PDAC immune microenvironment. AREAS COVERED This review provides a comprehensive survey of the pivotal involvement of neutrophils in the tumorigenesis and progression of PDAC. Furthermore, it synthesizes preclinical and clinical explorations aimed at targeting neutrophils within the milieu of PDAC, subsequently proposing a conceptual framework to propel further inquiry focused on enhancing the therapeutic efficacy of PDAC through neutrophil-targeted strategies. PubMed and Web of Science databases were utilized for researching neutrophils in pancreatic cancer publications prior to 2024. EXPERT OPINION Neutrophils play roles in promoting tumor growth and metastasis in PDAC and are associated with poor prognosis. However, the heterogeneity and plasticity of neutrophils and their complex relationships with other immune cells and extracellular matrix also provide new insights for immunotherapy targeting neutrophils to achieve a better prognosis for PDAC.
Collapse
Affiliation(s)
- Yi Jin
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Eric S Christenson
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lei Zheng
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Cancer Convergence Institute at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Keyu Li
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
26
|
Wu Q, Mao H, Jiang Z, Tang D. Tumour-associated neutrophils: Potential therapeutic targets in pancreatic cancer immunotherapy. Immunology 2024; 172:343-361. [PMID: 38402904 DOI: 10.1111/imm.13765] [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: 11/08/2023] [Accepted: 01/31/2024] [Indexed: 02/27/2024] Open
Abstract
Pancreatic cancer (PC) is a highly malignant tumour of the digestive system with poor therapeutic response and low survival rates. Immunotherapy has rapidly developed in recent years and has achieved significant outcomes in numerous malignant neoplasms. However, responses to immunotherapy in PC are rare, and the immunosuppressive and desmoplastic tumour microenvironment (TME) significantly hinders their efficacy in PC. Tumour-associated neutrophils (TANs) play a crucial role in the PC microenvironment and exert a profound influence on PC immunotherapy by establishing a robust stromal shelter and restraining immune cells to assist PC cells in immune escape, which may subvert the current status of PC immunotherapy. The present review aims to offer a comprehensive summary of the latest progress in understanding the involvement of TANs in PC desmoplastic and immunosuppressive functions and to emphasise the potential therapeutic implications of focusing on TANs in the immunotherapy of this deleterious disease. Finally, we provide an outlook for the future use of TANs in PC immunotherapy.
Collapse
Affiliation(s)
- Qihang Wu
- Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Han Mao
- Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Zhengting Jiang
- Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, China
| |
Collapse
|
27
|
Liu R, Li J, Liu L, Wang W, Jia J. Tumor-associated macrophages (TAMs): Constructing an immunosuppressive microenvironment bridge for pancreatic ductal adenocarcinoma (PDAC). CANCER PATHOGENESIS AND THERAPY 2024. [DOI: 10.1016/j.cpt.2024.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2025]
|
28
|
Shao S, Delk NA, Jones CN. A microphysiological system reveals neutrophil contact-dependent attenuation of pancreatic tumor progression by CXCR2 inhibition-based immunotherapy. Sci Rep 2024; 14:14142. [PMID: 38898176 PMCID: PMC11187156 DOI: 10.1038/s41598-024-64780-4] [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: 11/28/2023] [Accepted: 06/12/2024] [Indexed: 06/21/2024] Open
Abstract
Cancer cells recruit neutrophils from the bloodstream into the tumor tissue, where these immune cells promote the progression of numerous solid tumors. Studies in mice suggest that blocking neutrophil recruitment to tumors by inhibition of neutrophil chemokine receptor CXCR2 could be a potential immunotherapy for pancreatic cancer. Yet, the mechanisms by which neutrophils promote tumor progression in humans, as well as how CXCR2 inhibition could potentially serve as a cancer therapy, remain elusive. In this study, we developed a human cell-based microphysiological system to quantify neutrophil-tumor spheroid interactions in both "separated" and "contact" scenarios. We found that neutrophils promote the invasion of tumor spheroids through the secretion of soluble factors and direct contact with cancer cells. However, they promote the proliferation of tumor spheroids solely through direct contact. Interestingly, treatment with AZD-5069, a CXCR2 inhibitor, attenuates invasion and proliferation of tumor spheroids by blocking direct contact with neutrophils. Our findings also show that CXCR2 inhibition reduces neutrophil migration toward tumor spheroids. These results shed new light on the tumor-promoting mechanisms of human neutrophils and the tumor-suppressive mechanisms of CXCR2 inhibition in pancreatic cancer and may aid in the design and optimization of novel immunotherapeutic strategies based on neutrophils.
Collapse
Affiliation(s)
- Shuai Shao
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, 75080, USA
- Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Nikki A Delk
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Caroline N Jones
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, 75080, USA.
- Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, TX, 75235, USA.
| |
Collapse
|
29
|
Gao L, Shay C, Teng Y. Cell death shapes cancer immunity: spotlighting PANoptosis. J Exp Clin Cancer Res 2024; 43:168. [PMID: 38877579 PMCID: PMC11179218 DOI: 10.1186/s13046-024-03089-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/05/2024] [Indexed: 06/16/2024] Open
Abstract
PANoptosis represents a novel type of programmed cell death (PCD) with distinctive features that incorporate elements of pyroptosis, apoptosis, and necroptosis. PANoptosis is governed by a newly discovered cytoplasmic multimeric protein complex known as the PANoptosome. Unlike each of these PCD types individually, PANoptosis is still in the early stages of research and warrants further exploration of its specific regulatory mechanisms and primary targets. In this review, we provide a brief overview of the conceptual framework and molecular components of PANoptosis. In addition, we highlight recent advances in the understanding of the molecular mechanisms and therapeutic applications of PANoptosis. By elucidating the complex crosstalk between pyroptosis, apoptosis and necroptosis and summarizing the functional consequences of PANoptosis with a special focus on the tumor immune microenvironment, this review aims to provide a theoretical basis for the potential application of PANoptosis in cancer therapy.
Collapse
Affiliation(s)
- Lixia Gao
- National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, College of Pharmacy, Chongqing University of Arts and Sciences, Chongqing, 402160, People's Republic of China
| | - Chloe Shay
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| | - Yong Teng
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA.
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 201 Dowman Dr, Atlanta, GA, 30322, USA.
| |
Collapse
|
30
|
Farhangnia P, Khorramdelazad H, Nickho H, Delbandi AA. Current and future immunotherapeutic approaches in pancreatic cancer treatment. J Hematol Oncol 2024; 17:40. [PMID: 38835055 PMCID: PMC11151541 DOI: 10.1186/s13045-024-01561-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024] Open
Abstract
Pancreatic cancer is a major cause of cancer-related death, but despondently, the outlook and prognosis for this resistant type of tumor have remained grim for a long time. Currently, it is extremely challenging to prevent or detect it early enough for effective treatment because patients rarely exhibit symptoms and there are no reliable indicators for detection. Most patients have advanced or spreading cancer that is difficult to treat, and treatments like chemotherapy and radiotherapy can only slightly prolong their life by a few months. Immunotherapy has revolutionized the treatment of pancreatic cancer, yet its effectiveness is limited by the tumor's immunosuppressive and hard-to-reach microenvironment. First, this article explains the immunosuppressive microenvironment of pancreatic cancer and highlights a wide range of immunotherapy options, including therapies involving oncolytic viruses, modified T cells (T-cell receptor [TCR]-engineered and chimeric antigen receptor [CAR] T-cell therapy), CAR natural killer cell therapy, cytokine-induced killer cells, immune checkpoint inhibitors, immunomodulators, cancer vaccines, and strategies targeting myeloid cells in the context of contemporary knowledge and future trends. Lastly, it discusses the main challenges ahead of pancreatic cancer immunotherapy.
Collapse
Affiliation(s)
- Pooya Farhangnia
- Reproductive Sciences and Technology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hamid Nickho
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali-Akbar Delbandi
- Reproductive Sciences and Technology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Zhang J, Gu J, Wang X, Ji C, Yu D, Wang M, Pan J, Santos HA, Zhang H, Zhang X. Engineering and Targeting Neutrophils for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310318. [PMID: 38320755 DOI: 10.1002/adma.202310318] [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: 10/05/2023] [Revised: 01/29/2024] [Indexed: 02/22/2024]
Abstract
Neutrophils are the most abundant white blood cells in the circulation and act as the first line of defense against infections. Increasing evidence suggests that neutrophils possess heterogeneous phenotypes and functional plasticity in human health and diseases, including cancer. Neutrophils play multifaceted roles in cancer development and progression, and an N1/N2 paradigm of neutrophils in cancer is proposed, where N1 neutrophils exert anti-tumor properties while N2 neutrophils display tumor-supportive and immune-suppressive functions. Selective activation of beneficial neutrophil population and targeted inhibition or re-polarization of tumor-promoting neutrophils has shown an important potential in tumor therapy. In addition, due to the natural inflammation-responsive and physical barrier-crossing abilities, neutrophils and their derivatives (membranes and extracellular vesicles (EVs)) are regarded as advanced drug delivery carriers for enhanced tumor targeting and improved therapeutic efficacy. In this review, the recent advances in engineering neutrophils for drug delivery and targeting neutrophils for remodeling tumor microenvironment (TME) are comprehensively presented. This review will provide a broad understanding of the potential of neutrophils in cancer therapy.
Collapse
Affiliation(s)
- Jiahui Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Jianmei Gu
- Departmemt of Clinical Laboratory Medicine, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, 226361, China
| | - Xu Wang
- Department of Radiation Oncology, Jiangsu University Cancer Institute, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China
| | - Cheng Ji
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Dan Yu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Maoye Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Jianming Pan
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen/University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory, Abo Akademi University, Turku, 20520, Finland
- Turku Bioscience Centre, University of Turku and Abo Akademi University, Turku, 20520, Finland
| | - Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| |
Collapse
|
32
|
Ni Y, Li R, Shen X, Yi D, Ren Y, Wang F, Geng Y, You Q. Diaphorobacter nitroreducens synergize with oxaliplatin to reduce tumor burden in mice with lung adenocarcinoma. mSystems 2024; 9:e0132323. [PMID: 38483163 PMCID: PMC11019951 DOI: 10.1128/msystems.01323-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/29/2024] [Indexed: 04/17/2024] Open
Abstract
Lung adenocarcinoma (LADC) is the most common lung cancer and the leading cause of cancer-related deaths globally. Accumulating evidence suggests that the gut microbiota regulates the host response to chemotherapeutic drugs and can be targeted to reduce the toxicity of current chemotherapeutic agents. However, the effect of Diaphorobacter nitroreducens synergized with oxaliplatin on the gut microbiota and their impact on LADC have never been explored. This study aimed to evaluate the anti-cancer effects of D. nitroreducens, oxaliplatin, and their combined treatment on tumor growth in tumor-bearing mice. The composition of gut microbiota and the immune infiltration of tumors were evaluated by using 16S rRNA gene high-throughput sequencing and immunofluorescence, respectively. The inhibitory effect of the combination treatment with D. nitroreducens and oxaliplatin was significantly stronger than that of oxaliplatin alone in tumor-bearing mice. Furthermore, we observed that the combination treatment significantly increased the relative abundance of Lactobacillus and Akkermansia in the gut microbiota. Meanwhile, the combination treatment significantly increased the proportions of macrophage but decreased the proportion of regulatory T cells in the LADC tumor tissues of mice. These findings underscored the relationship between D. nitroreducens and the gut microbiota-immune cell-LADC axis, highlighting potential therapeutic avenues for LADC treatment. IMPORTANCE Oxaliplatin is widely used as an effective chemotherapeutic agent in cancer treatment, but its side effects and response rate still need to be improved. Conventional probiotics potentially benefit cancer chemotherapy by regulating gut microbiota and tumor immune infiltration. This study was novel in reporting a more significant inhibitory effect of Diaphorobacter nitroreducens on lung adenocarcinoma (LADC) cells compared with common traditional probiotics and validating its potential as an adjuvant therapy for LADC chemotherapy in mice. This study investigated the impact of D. nitroreducens combined with oxaliplatin on the gut microbiota and immune infiltration of tumors as a potential mechanism to improve anticancer effects.
Collapse
Affiliation(s)
- Yalan Ni
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
| | - Rui Li
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
| | - Xiaoyu Shen
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Deli Yi
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yilin Ren
- Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Fudong Wang
- Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yan Geng
- School of Life Science and Health Engineering, Jiangnan University, Wuxi, China
| | - Qingjun You
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
33
|
Shu Y, Huang H, Gao M, Xu W, Cao X, Jia X, Deng B. Lipid Metabolism-Related Gene Markers Used for Prediction Prognosis, Immune Microenvironment, and Tumor Stage of Pancreatic Cancer. Biochem Genet 2024; 62:931-949. [PMID: 37505298 PMCID: PMC11031448 DOI: 10.1007/s10528-023-10457-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: 02/27/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023]
Abstract
Recently, more and more evidence shows that lipid metabolism disorder has been observed in tumor, which impacts tumor cell proliferation, survival, invasion, metastasis, and response to the tumor microenvironment (TME) and tumor treatment. However, hitherto there has not been sufficient research to demonstrate the role of lipid metabolism in pancreatic cancer. This study contrives to get an insight into the relationship between the characteristics of lipid metabolism and pancreatic cancer. We collected samples of patients with pancreatic cancer from the Gene Expression Omnibus (GEO), the Therapeutically Applicable Research to Generate Effective Treatments (TARGET), and the International Cancer Genome Consortium (ICGC) databases. Firstly, we implemented univariate regression analysis to get prognosis-related lipid metabolism genes screened and a construction of protein-protein interaction (PPI) network ensued. Then, contingent on our screening results, we explored the molecular subtypes mediated by lipid metabolism-related genes and the correlated TME cell infiltration. Additionally, we studied the disparately expressed genes among disparate lipid metabolism subtypes and established a scoring model of lipid metabolism-related characteristics using the least absolute shrinkage and selection operator (LASSO) regression analysis. At last, we explored the relationship between the scoring model and disease prognosis, tumor stage, tumor microenvironment, and immunotherapy. Two subtypes, C1 and C2, were identified, and lipid metabolism-related genes were studied. The result indicated that the patients with subtype C2 have a significantly lower survival rate than that of the patients with subtype C1, and we found difference in abundance of different immune-infiltrating cells. The gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses revealed the association of these differentially expressed genes with functions and pathways related to lipid metabolism. Finally, we established a scoring model of lipid metabolism-related characteristics based on the disparately expressed genes. The results show that our scoring model have a substantial effect on forecasting the prognosis of patients with pancreatic cancer. The lipid metabolism model is an important biomarker of pancreatic cancer. Using the model, the relationship between disease prognosis, molecular subtypes, TME cell infiltration characteristics, and immunotherapy in pancreatic cancer patients could be explored.
Collapse
Affiliation(s)
- Yuan Shu
- The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi, 330000, People's Republic of China
- Departments of Endocrine, The First Hospital of Nanchang, Nanchang, Jiangxi, 330008, People's Republic of China
| | - Haiqiang Huang
- The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi, 330000, People's Republic of China
- Departments of Endocrine, The First Hospital of Nanchang, Nanchang, Jiangxi, 330008, People's Republic of China
| | - Minjie Gao
- The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi, 330000, People's Republic of China
- Departments of Endocrine, The First Hospital of Nanchang, Nanchang, Jiangxi, 330008, People's Republic of China
| | - Wenjie Xu
- The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi, 330000, People's Republic of China
- Departments of Endocrine, The First Hospital of Nanchang, Nanchang, Jiangxi, 330008, People's Republic of China
| | - Xiang Cao
- The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi, 330000, People's Republic of China
- Departments of Endocrine, The First Hospital of Nanchang, Nanchang, Jiangxi, 330008, People's Republic of China
| | - Xiaoze Jia
- Internet of Things Engineering, College of Wuxi University, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Bo Deng
- Departments of Endocrine, The First Hospital of Nanchang, Nanchang, Jiangxi, 330008, People's Republic of China.
| |
Collapse
|
34
|
Basnet A, Landreth KM, Nohoesu R, Santiago SP, Geldenhuys WJ, Boone BA, Liu TW. Targeting myeloperoxidase limits myeloid cell immunosuppression enhancing immune checkpoint therapy for pancreatic cancer. Cancer Immunol Immunother 2024; 73:57. [PMID: 38367056 PMCID: PMC10874341 DOI: 10.1007/s00262-024-03647-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/29/2024] [Indexed: 02/19/2024]
Abstract
Pancreatic ductal adenocarcinoma is a devastating disease characterized by an extreme resistance to current therapies, including immune checkpoint therapy. The limited success of immunotherapies can be attributed to a highly immunosuppressive pancreatic cancer microenvironment characterized by an extensive infiltration of immune suppressing myeloid cells. While there are several pathways through which myeloid cells contribute to immunosuppression, one important mechanism is the increased production of reactive oxygen species. Here, we evaluated the contribution of myeloperoxidase, a myeloid-lineage restricted enzyme and primary source of reactive oxygen species, to regulate immune checkpoint therapy response in preclinical pancreatic cancer models. We compared treatment outcome, immune composition and characterized myeloid cells using wild-type, myeloperoxidase-deficient, and myeloperoxidase inhibitor treated wild-type mice using established subcutaneous pancreatic cancer models. Loss of host myeloperoxidase and pharmacological inhibition of myeloperoxidase in combination with immune checkpoint therapy significantly delayed tumor growth. The tumor microenvironment and systemic immune landscape demonstrated significant decreases in myeloid cells, exhausted T cells and T regulatory cell subsets when myeloperoxidase was deficient. Loss of myeloperoxidase in isolated myeloid cell subsets from tumor-bearing mice resulted in decreased reactive oxygen species production and T cell suppression. These data suggest that myeloperoxidase contributes to an immunosuppressive microenvironment and immune checkpoint therapy resistance where myeloperoxidase inhibitors have the potential to enhance immunotherapy response. Repurposing myeloperoxidase specific inhibitors may provide a promising therapeutic strategy to expand therapeutic options for pancreatic cancer patients to include immunotherapies.
Collapse
Affiliation(s)
- Angisha Basnet
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA
| | - Kaitlyn M Landreth
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA
| | - Remi Nohoesu
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA
| | - Stell P Santiago
- Department of Pathology, Anatomy and Laboratory Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Werner J Geldenhuys
- WVU Cancer Institute, West Virginia University, Morgantown, WV, 26506, USA
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, 26506, USA
| | - Brian A Boone
- WVU Cancer Institute, West Virginia University, Morgantown, WV, 26506, USA
- Division of Surgical Oncology, Department of Surgery, West Virginia University, Morgantown, WV, 26506, USA
| | - Tracy W Liu
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA.
- WVU Cancer Institute, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
35
|
Yang F, He Y, Ge N, Guo J, Yang F, Sun S. Exploring KRAS-mutant pancreatic ductal adenocarcinoma: a model validation study. Front Immunol 2024; 14:1203459. [PMID: 38268915 PMCID: PMC10805828 DOI: 10.3389/fimmu.2023.1203459] [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: 04/10/2023] [Accepted: 12/22/2023] [Indexed: 01/26/2024] Open
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) has the highest mortality rate among all solid tumors. Tumorigenesis is promoted by the oncogene KRAS, and KRAS mutations are prevalent in patients with PDAC. Therefore, a comprehensive understanding of the interactions between KRAS mutations and PDAC may expediate the development of therapeutic strategies for reversing the progression of malignant tumors. Our study aims at establishing and validating a prediction model of KRAS mutations in patients with PDAC based on survival analysis and mRNA expression. METHODS A total of 184 and 412 patients with PDAC from The Cancer Genome Atlas (TCGA) database and the International Cancer Genome Consortium (ICGC), respectively, were included in the study. RESULTS After tumor mutation profile and copy number variation (CNV) analyses, we established and validated a prediction model of KRAS mutations, based on survival analysis and mRNA expression, that contained seven genes: CSTF2, FAF2, KIF20B, AKR1A1, APOM, KRT6C, and CD70. We confirmed that the model has a good predictive ability for the prognosis of overall survival (OS) in patients with KRAS-mutated PDAC. Then, we analyzed differential biological pathways, especially the ferroptosis pathway, through principal component analysis, pathway enrichment analysis, Gene Ontology (GO) enrichment analysis, and gene set enrichment analysis (GSEA), with which patients were classified into low- or high-risk groups. Pathway enrichment results revealed enrichment in the cytokine-cytokine receptor interaction, metabolism of xenobiotics by cytochrome P450, and viral protein interaction with cytokine and cytokine receptor pathways. Most of the enriched pathways are metabolic pathways predominantly enriched by downregulated genes, suggesting numerous downregulated metabolic pathways in the high-risk group. Subsequent tumor immune infiltration analysis indicated that neutrophil infiltration, resting CD4 memory T cells, and resting natural killer (NK) cells correlated with the risk score. After verifying that the seven gene expression levels in different KRAS-mutated pancreatic cancer cell lines were similar to that in the model, we screened potential drugs related to the risk score. DISCUSSION This study established, analyzed, and validated a model for predicting the prognosis of PDAC based on risk stratification according to KRAS mutations, and identified differential pathways and highly effective drugs.
Collapse
Affiliation(s)
- Fan Yang
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yanjie He
- Department of Surgery, New York University School of Medicine and NYU-Langone Medical Center, New York, NY, United States
| | - Nan Ge
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jintao Guo
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fei Yang
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Siyu Sun
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
36
|
Feng S, Luo H, Li C, Geng Y, Yang Z, Zhao X, Wang L, Liu R, Zhang Y, Che T, Zhang Q, Wang X. Regulatory role of RGMb in lung injury promoted by the combination of carbon ion irradiation and anti-PD-1 antibody through Erk1/2 and p38 MAPK pathways. Biochem Biophys Res Commun 2024; 691:149334. [PMID: 38042034 DOI: 10.1016/j.bbrc.2023.149334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/17/2023] [Accepted: 11/23/2023] [Indexed: 12/04/2023]
Abstract
The combination of carbon ion radiotherapy and anti-PD-1 antibody represents a new approach to treating thoracic tumors. However, the lung damage caused by this combination therapy may limit its use, and the potential mechanisms for this are worthy of investigation. The objective of this research was to examine the potential involvement of repulsive guidance molecule b (RGMb) in lung damage promoted by the utilization of carbon ion irradiation combined with an anti-PD-1 antibody. The C57BL/6 mice have been randomly separated into four distinct groups: control, anti-PD-1, whole thorax carbon ion irradiation, and irradiation in combination with anti-PD-1 treatment groups (combination group). Detection of pathological changes in lung tissue using HE staining. Detection of pulmonary fibrosis by Masson staining and the hydroxyproline assay. ELISA to detect TNF-α, TGF-β, IL-6, and IL-1β expression levels within lung homogenates. The expression of RGMb, p38 MAPK, and Erk1/2 pathways was detected using a fully automated digital Western blotting system WES (ProteinSimple, USA). Flow cytometry was employed to analyze tissue-resident memory T cells (TRM) within the lung. Subsequently, the siRNA gene was employed to induce the downregulation of RGMb in mice in order to validate the involvement of RGMb in radiation-immune lung injury. The present study observed a significant increase in both inflammatory and fibrotic indicators within the mice group's lung tissue that received the combination treatment. The combination group exhibited elevated levels of TGF-β, TNF-α, IL-6, and IL-1β in lung homogenates. Anti-PD-1 antibody and carbon ion irradiation, upregulated RGMb, phospho-p38 MAPK and phospho-Erk1/2. The results obtained from the flow cytometry analysis indicated that the combination group was significantly higher in the number of clonal expansion TRMs, which were predominantly characterized by the expression of CD8+CD103+CD69-TRMs. The downregulate of RGMb via siRNA in mice resulted in a decrease in phospho-p38 MAPK and phospho-Erk1/2. The combination group exhibited a reduction in TNF-α, TGF-β, IL-6, and IL-1β in their lung tissues, and the number of CD8+CD103+CD69-TRM was significantly reduced. The combination group exhibited a significant improvement in inflammatory and fibrotic indicators within the lung tissues. Anti-PD-1 antibody and carbon ion irradiation synergistically regulate RGMb, leading to strong clonal expansion of lung TRM through the p38 MAPK and Erk1/2 pathways. The present study offers valuable insights into the treatment of lung injury due to the combined administration of carbon ion radiotherapy and anti-PD-1 antibody therapy.
Collapse
Affiliation(s)
- Shuangwu Feng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China.
| | - Hongtao Luo
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Chengcheng Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China.
| | - Yichao Geng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China.
| | - Zhen Yang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China; Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China.
| | - Xueshan Zhao
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China.
| | - Lina Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China.
| | - Ruifeng Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Yanying Zhang
- Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China.
| | - Tuanjie Che
- Key Laboratory of Functional Genomics and Molecular Diagnosis of Gansu Province, Lanzhou, Gansu, China.
| | - Qiuning Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Xiaohu Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China; Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China; University of Chinese Academy of Sciences, Beijing, China; Key Laboratory of Functional Genomics and Molecular Diagnosis of Gansu Province, Lanzhou, Gansu, China.
| |
Collapse
|
37
|
Luo W, Wen T, Qu X. Tumor immune microenvironment-based therapies in pancreatic ductal adenocarcinoma: time to update the concept. J Exp Clin Cancer Res 2024; 43:8. [PMID: 38167055 PMCID: PMC10759657 DOI: 10.1186/s13046-023-02935-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal solid tumors. The tumor immune microenvironment (TIME) formed by interactions among cancer cells, immune cells, cancer-associated fibroblasts (CAF), and extracellular matrix (ECM) components drives PDAC in a more immunosuppressive direction: this is a major cause of therapy resistance and poor prognosis. In recent years, research has advanced our understanding of the signaling mechanism by which TIME components interact with the tumor and the evolution of immunophenotyping. Through revolutionary technologies such as single-cell sequencing, we have gone from simply classifying PDACs as "cold" and "hot" to a more comprehensive approach of immunophenotyping that considers all the cells and matrix components. This is key to improving the clinical efficacy of PDAC treatments. In this review, we elaborate on various TIME components in PDAC, the signaling mechanisms underlying their interactions, and the latest research into PDAC immunophenotyping. A deep understanding of these network interactions will contribute to the effective combination of TIME-based therapeutic approaches, such as immune checkpoint inhibitors (ICI), adoptive cell therapy, therapies targeting myeloid cells, CAF reprogramming, and stromal normalization. By selecting the appropriate integrated therapies based on precise immunophenotyping, significant advances in the future treatment of PDAC are possible.
Collapse
Affiliation(s)
- Wenyu Luo
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, 110001, Liaoning, China
| | - Ti Wen
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, 110001, Liaoning, China.
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
38
|
Chen R, Coleborn E, Bhavsar C, Wang Y, Alim L, Wilkinson AN, Tran MA, Irgam G, Atluri S, Wong K, Shim JJ, Adityan S, Lee JS, Overwijk WW, Steptoe R, Yang D, Wu SY. miR-146a inhibits ovarian tumor growth in vivo via targeting immunosuppressive neutrophils and enhancing CD8 + T cell infiltration. Mol Ther Oncolytics 2023; 31:100725. [PMID: 37781339 PMCID: PMC10539880 DOI: 10.1016/j.omto.2023.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/08/2023] [Indexed: 10/03/2023] Open
Abstract
Immunotherapies have emerged as promising strategies for cancer treatment. However, existing immunotherapies have poor activity in high-grade serous ovarian cancer (HGSC) due to the immunosuppressive tumor microenvironment and the associated low tumoral CD8+ T cell (CTL) infiltration. Through multiple lines of evidence, including integrative analyses of human HGSC tumors, we have identified miR-146a as a master regulator of CTL infiltration in HGSC. Tumoral miR-146a expression is positively correlated with anti-cancer immune signatures in human HGSC tumors, and delivery of miR-146a to tumors resulted in significant reduction in tumor growth in both ID8-p53-/- and IG10 murine HGSC models. Increasing miR-146a expression in tumors improved anti-tumor immune responses by decreasing immune suppressive neutrophils and increasing CTL infiltration. Mechanistically, miR-146a targets IL-1 receptor-associated kinase 1 and tumor necrosis factor receptor-associated factor 6 adaptor molecules of the transcription factor nuclear factor κB signaling pathway in ID8-p53-/- cells and decreases production of the downstream neutrophil chemoattractant, C-X-C motif chemokine ligand 1. In addition to HGSC, tumoral miR-146a expression also correlates strongly with CTL infiltration in other cancer types including thyroid, prostate, breast, and adrenocortical cancers. Altogether, our findings highlight the ability of miR-146a to overcome immune suppression and improve CTL infiltration in tumors.
Collapse
Affiliation(s)
- Rui Chen
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Elaina Coleborn
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Chintan Bhavsar
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Yue Wang
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Louisa Alim
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Andrew N. Wilkinson
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | | | - Gowri Irgam
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Sharat Atluri
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kiefer Wong
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jae-Jun Shim
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Siddharth Adityan
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Ju-Seog Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Willem W. Overwijk
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Raymond Steptoe
- Frazer Institute, University of Queensland, Brisbane, QLD 4102, Australia
| | - Da Yang
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Sherry Y. Wu
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
39
|
Canel M, Sławińska AD, Lonergan DW, Kallor AA, Upstill-Goddard R, Davidson C, von Kriegsheim A, Biankin AV, Byron A, Alfaro J, Serrels A. FAK suppresses antigen processing and presentation to promote immune evasion in pancreatic cancer. Gut 2023; 73:131-155. [PMID: 36977556 PMCID: PMC10715489 DOI: 10.1136/gutjnl-2022-327927] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 03/19/2023] [Indexed: 03/30/2023]
Abstract
OBJECTIVE Immunotherapy for the treatment of pancreatic ductal adenocarcinoma (PDAC) has shown limited efficacy. Poor CD8 T-cell infiltration, low neoantigen load and a highly immunosuppressive tumour microenvironment contribute to this lack of response. Here, we aimed to further investigate the immunoregulatory function of focal adhesion kinase (FAK) in PDAC, with specific emphasis on regulation of the type-II interferon response that is critical in promoting T-cell tumour recognition and effective immunosurveillance. DESIGN We combined CRISPR, proteogenomics and transcriptomics with mechanistic experiments using a KrasG12Dp53R172H mouse model of pancreatic cancer and validated findings using proteomic analysis of human patient-derived PDAC cell lines and analysis of publicly available human PDAC transcriptomics datasets. RESULTS Loss of PDAC cell-intrinsic FAK signalling promotes expression of the immunoproteasome and Major Histocompatibility Complex class-I (MHC-I), resulting in increased antigen diversity and antigen presentation by FAK-/- PDAC cells. Regulation of the immunoproteasome by FAK is a critical determinant of this response, optimising the physicochemical properties of the peptide repertoire for high affinity binding to MHC-I. Expression of these pathways can be further amplified in a STAT1-dependent manner via co-depletion of FAK and STAT3, resulting in extensive infiltration of tumour-reactive CD8 T-cells and further restraint of tumour growth. FAK-dependent regulation of antigen processing and presentation is conserved between mouse and human PDAC, but is lost in cells/tumours with an extreme squamous phenotype. CONCLUSION Therapies aimed at FAK degradation may unlock additional therapeutic benefit for the treatment of PDAC through increasing antigen diversity and promoting antigen presentation.
Collapse
Affiliation(s)
- Marta Canel
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | | | - David W Lonergan
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Ashwin Adrian Kallor
- International Centre for Cancer Vaccine Science, University of Gdansk, Gdansk, Poland
| | - Rosie Upstill-Goddard
- The Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Catherine Davidson
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Alex von Kriegsheim
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Andrew V Biankin
- The Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Adam Byron
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Javier Alfaro
- International Centre for Cancer Vaccine Science, University of Gdansk, Gdansk, Poland
| | - Alan Serrels
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
40
|
Gong YT, Zhang LJ, Liu YC, Tang M, Lin JY, Chen XY, Chen YX, Yan Y, Zhang WD, Jin JM, Luan X. Neutrophils as potential therapeutic targets for breast cancer. Pharmacol Res 2023; 198:106996. [PMID: 37972723 DOI: 10.1016/j.phrs.2023.106996] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
Breast cancer (BC) remains the foremost cause of cancer mortality globally, with neutrophils playing a critical role in its pathogenesis. As an essential tumor microenvironment (TME) component, neutrophils are emerging as pivotal factors in BC progression. Growing evidence has proved that neutrophils play a Janus- role in BC by polarizing into the anti-tumor (N1) or pro-tumor (N2) phenotype. Clinical trials are evaluating neutrophil-targeted therapies, including Reparixin (NCT02370238) and Tigatuzumab (NCT01307891); however, their clinical efficacy remains suboptimal. This review summarizes the evidence regarding the close relationship between neutrophils and BC, emphasizing the critical roles of neutrophils in regulating metabolic and immune pathways. Additionally, we summarize the existing therapeutic approaches that target neutrophils, highlighting the challenges, and affirming the rationale for continuing to explore neutrophils as a viable therapeutic target in BC management.
Collapse
Affiliation(s)
- Yi-Ting Gong
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| | - Li-Jun Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yi-Chen Liu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Min Tang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jia-Yi Lin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xin-Yi Chen
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yi-Xu Chen
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yue Yan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wei-Dong Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Second Military Medical University, Shanghai 201203, China; Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China.
| | - Jin-Mei Jin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xin Luan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
41
|
Yi M, Li T, Niu M, Mei Q, Zhao B, Chu Q, Dai Z, Wu K. Exploiting innate immunity for cancer immunotherapy. Mol Cancer 2023; 22:187. [PMID: 38008741 PMCID: PMC10680233 DOI: 10.1186/s12943-023-01885-w] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/23/2023] [Indexed: 11/28/2023] Open
Abstract
Immunotherapies have revolutionized the treatment paradigms of various types of cancers. However, most of these immunomodulatory strategies focus on harnessing adaptive immunity, mainly by inhibiting immunosuppressive signaling with immune checkpoint blockade, or enhancing immunostimulatory signaling with bispecific T cell engager and chimeric antigen receptor (CAR)-T cell. Although these agents have already achieved great success, only a tiny percentage of patients could benefit from immunotherapies. Actually, immunotherapy efficacy is determined by multiple components in the tumor microenvironment beyond adaptive immunity. Cells from the innate arm of the immune system, such as macrophages, dendritic cells, myeloid-derived suppressor cells, neutrophils, natural killer cells, and unconventional T cells, also participate in cancer immune evasion and surveillance. Considering that the innate arm is the cornerstone of the antitumor immune response, utilizing innate immunity provides potential therapeutic options for cancer control. Up to now, strategies exploiting innate immunity, such as agonists of stimulator of interferon genes, CAR-macrophage or -natural killer cell therapies, metabolic regulators, and novel immune checkpoint blockade, have exhibited potent antitumor activities in preclinical and clinical studies. Here, we summarize the latest insights into the potential roles of innate cells in antitumor immunity and discuss the advances in innate arm-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Ming Yi
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, People's Republic of China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Qi Mei
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China
| | - Bin Zhao
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| | - Zhijun Dai
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China.
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
42
|
Fernandes R, Costa C, Fernandes R, Barros AN. Inflammation in Prostate Cancer: Exploring the Promising Role of Phenolic Compounds as an Innovative Therapeutic Approach. Biomedicines 2023; 11:3140. [PMID: 38137361 PMCID: PMC10740737 DOI: 10.3390/biomedicines11123140] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Prostate cancer (PCa) remains a significant global health concern, being a major cause of cancer morbidity and mortality worldwide. Furthermore, profound understanding of the disease is needed. Prostate inflammation caused by external or genetic factors is a central player in prostate carcinogenesis. However, the mechanisms underlying inflammation-driven PCa remain poorly understood. This review dissects the diagnosis methods for PCa and the pathophysiological mechanisms underlying the disease, clarifying the dynamic interplay between inflammation and leukocytes in promoting tumour development and spread. It provides updates on recent advances in elucidating and treating prostate carcinogenesis, and opens new insights for the use of bioactive compounds in PCa. Polyphenols, with their noteworthy antioxidant and anti-inflammatory properties, along with their synergistic potential when combined with conventional treatments, offer promising prospects for innovative therapeutic strategies. Evidence from the use of polyphenols and polyphenol-based nanoparticles in PCa revealed their positive effects in controlling tumour growth, proliferation, and metastasis. By consolidating the diverse features of PCa research, this review aims to contribute to increased understanding of the disease and stimulate further research into the role of polyphenols and polyphenol-based nanoparticles in its management.
Collapse
Affiliation(s)
- Raquel Fernandes
- Centre for Research and Technology of Agro-Environmental and Biological Sciences, CITAB, Inov4Agro, University of Trás-os-Montes and Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal;
| | - Cátia Costa
- Centre for Research and Technology of Agro-Environmental and Biological Sciences, CITAB, Inov4Agro, University of Trás-os-Montes and Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal;
| | - Rúben Fernandes
- FP-I3ID, Instituto de Investigação, Inovação e Desenvolvimento, FP-BHS, Biomedical and Health Sciences, Universidade Fernando Pessoa, 4249-004 Porto, Portugal;
- CECLIN, Centro de Estudos Clínicos, Hospital Fernando Pessoa, 4420-096 Gondomar, Portugal
- I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Ana Novo Barros
- Centre for Research and Technology of Agro-Environmental and Biological Sciences, CITAB, Inov4Agro, University of Trás-os-Montes and Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal;
| |
Collapse
|
43
|
Moinard-Butot F, Nannini S, Fischbach C, Abdallahoui S, Demarchi M, Petit T, Bender L, Schott R. Anaplastic Lymphoma Kinase Inhibitor-Induced Neutropenia: A Systematic Review. Cancers (Basel) 2023; 15:4940. [PMID: 37894307 PMCID: PMC10605921 DOI: 10.3390/cancers15204940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Lung cancers with ALK rearrangement represent less than 5% of all lung cancers. ALK inhibitors are currently used to treat first-line metastatic non-small cell lung cancer with ALK rearrangement. Compared to chemotherapy, ALK inhibitors have improved progression-free survival, overall survival, and quality of life for patients. The results of several phase 3 studies with a follow-up of over 6 years suggest that the life expectancy of these patients treated with targeted therapies is significantly higher than 5 years and could approach 10 years. Nevertheless, these treatments induce haematological toxicities, including neutropenia. Few data are available on neutropenia induced by ALK inhibitors and on the pathophysiological mechanism and therapeutic adaptations necessary to continue the treatment. Given the high efficacy of these treatments, managing side effects to avoid treatment interruptions is essential. Here, we have reviewed the data from published clinical studies and case reports to provide an overview of neutropenia induced by ALK inhibitors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Roland Schott
- Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe, 17 Rue Albert Calmette, 67033 Strasbourg, France; (F.M.-B.); (S.N.); (C.F.); (S.A.); (M.D.); (T.P.); (L.B.)
| |
Collapse
|
44
|
Guo J, Wang S, Gao Q. An integrated overview of the immunosuppression features in the tumor microenvironment of pancreatic cancer. Front Immunol 2023; 14:1258538. [PMID: 37771596 PMCID: PMC10523014 DOI: 10.3389/fimmu.2023.1258538] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 08/29/2023] [Indexed: 09/30/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies. It is characterized by a complex and immunosuppressive tumor microenvironment (TME), which is primarily composed of tumor cells, stromal cells, immune cells, and acellular components. The cross-interactions and -regulations among various cell types in the TME have been recognized to profoundly shape the immunosuppression features that meaningfully affect PDAC biology and treatment outcomes. In this review, we first summarize five cellular composition modules by integrating the cellular (sub)types, phenotypes, and functions in PDAC TME. Then we discuss an integrated overview of the cross-module regulations as a determinant of the immunosuppressive TME in PDAC. We also briefly highlight TME-targeted strategies that potentially improve PDAC therapy.
Collapse
Affiliation(s)
- Jinglong Guo
- Department of Cardiovascular Disease, the First Hospital of Jilin University, Changchun, China
| | - Siyue Wang
- Baylor College of Medicine, One Baylor Plaza, Houston, TX, United States
| | - Qi Gao
- Department of Cardiovascular Disease, the First Hospital of Jilin University, Changchun, China
| |
Collapse
|
45
|
Liu S, Wu W, Du Y, Yin H, Chen Q, Yu W, Wang W, Yu J, Liu L, Lou W, Pu N. The evolution and heterogeneity of neutrophils in cancers: origins, subsets, functions, orchestrations and clinical applications. Mol Cancer 2023; 22:148. [PMID: 37679744 PMCID: PMC10483725 DOI: 10.1186/s12943-023-01843-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/14/2023] [Indexed: 09/09/2023] Open
Abstract
Neutrophils, the most prevalent innate immune cells in humans, have garnered significant attention in recent years due to their involvement in cancer progression. This comprehensive review aimed to elucidate the important roles and underlying mechanisms of neutrophils in cancer from the perspective of their whole life cycle, tracking them from development in the bone marrow to circulation and finally to the tumor microenvironment (TME). Based on an understanding of their heterogeneity, we described the relationship between abnormal neutrophils and clinical manifestations in cancer. Specifically, we explored the function, origin, and polarization of neutrophils within the TME. Furthermore, we also undertook an extensive analysis of the intricate relationship between neutrophils and clinical management, including neutrophil-based clinical treatment strategies. In conclusion, we firmly assert that directing future research endeavors towards comprehending the remarkable heterogeneity exhibited by neutrophils is of paramount importance.
Collapse
Affiliation(s)
- Siyao Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P.R. China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenchuan Wu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P.R. China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yueshan Du
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P.R. China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hanlin Yin
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P.R. China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Qiangda Chen
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P.R. China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Weisheng Yu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P.R. China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenquan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P.R. China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jun Yu
- Departments of Medicine and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P.R. China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Wenhui Lou
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P.R. China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Ning Pu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P.R. China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
46
|
Zhou W, Cao X, Xu Q, Qu J, Sun Y. The double-edged role of neutrophil heterogeneity in inflammatory diseases and cancers. MedComm (Beijing) 2023; 4:e325. [PMID: 37492784 PMCID: PMC10363828 DOI: 10.1002/mco2.325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/31/2023] [Accepted: 06/09/2023] [Indexed: 07/27/2023] Open
Abstract
Neutrophils are important immune cells act as the body's first line of defense against infection and respond to diverse inflammatory cues. Many studies have demonstrated that neutrophils display plasticity in inflammatory diseases and cancers. Clarifying the role of neutrophil heterogeneity in inflammatory diseases and cancers will contribute to the development of novel treatment strategies. In this review, we have presented a review on the development of the understanding on neutrophil heterogeneity from the traditional perspective and a high-resolution viewpoint. A growing body of evidence has confirmed the double-edged role of neutrophils in inflammatory diseases and tumors. This may be due to a lack of precise understanding of the role of specific neutrophil subsets in the disease. Thus, elucidating specific neutrophil subsets involved in diseases would benefit the development of precision medicine. Thusly, we have summarized the relevance and actions of neutrophil heterogeneity in inflammatory diseases and cancers comprehensively. Meanwhile, we also discussed the potential intervention strategy for neutrophils. This review is intended to deepen our understanding of neutrophil heterogeneity in inflammatory diseases and cancers, while hold promise for precise treatment of neutrophil-related diseases.
Collapse
Affiliation(s)
- Wencheng Zhou
- Department of PharmacyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
| | - Xinran Cao
- State Key Laboratory of Pharmaceutical BiotechnologyDepartment of Biotechnology and Pharmaceutical SciencesSchool of Life ScienceNanjing UniversityNanjingChina
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical BiotechnologyDepartment of Biotechnology and Pharmaceutical SciencesSchool of Life ScienceNanjing UniversityNanjingChina
| | - Jiao Qu
- State Key Laboratory of Pharmaceutical BiotechnologyDepartment of Biotechnology and Pharmaceutical SciencesSchool of Life ScienceNanjing UniversityNanjingChina
| | - Yang Sun
- State Key Laboratory of Pharmaceutical BiotechnologyDepartment of Biotechnology and Pharmaceutical SciencesSchool of Life ScienceNanjing UniversityNanjingChina
| |
Collapse
|
47
|
Peng Z, Li M, Li H, Gao Q. PD-1/PD-L1 immune checkpoint blockade in ovarian cancer: dilemmas and opportunities. Drug Discov Today 2023:103666. [PMID: 37302543 DOI: 10.1016/j.drudis.2023.103666] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 05/23/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized treatment in oncology. Antibodies against PD-1/PD-L1 and ICI-based combinations are under clinical investigations in multiple cancers, including ovarian cancer. However, the success of ICIs has not materialized in ovarian cancer, which remains one of the few malignancies where ICIs exhibit modest efficacy as either monotherapy or combination therapy. In this review, we summarize completed and ongoing clinical trials of PD-1/PD-L1 blockade in ovarian cancer, categorize the underlying mechanisms of resistance emergence, and introduce candidate approaches to rewire the tumor microenvironment (TME) to potentiate anti-PD-1/PD-L1 antibodies. Teaser: The intrinsic resistance of ovarian cancer to PD-1/PD-L1 blockade could be overcome by advanced understanding of underlying mechanisms and discoveries of new actionable targets for combinatory treatment.
Collapse
Affiliation(s)
- Zikun Peng
- Department of Gynaecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Centre for Obstetrics and Gynaecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Li
- Department of Gynaecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Centre for Obstetrics and Gynaecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huayi Li
- Department of Gynaecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Centre for Obstetrics and Gynaecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinglei Gao
- Department of Gynaecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Centre for Obstetrics and Gynaecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
48
|
Hou Z, Lin J, Ma Y, Fang H, Wu Y, Chen Z, Lin X, Lu F, Wen S, Yu X, Huang H, Pan Y. Single-cell RNA sequencing revealed subclonal heterogeneity and gene signatures of gemcitabine sensitivity in pancreatic cancer. Front Pharmacol 2023; 14:1193791. [PMID: 37324492 PMCID: PMC10267405 DOI: 10.3389/fphar.2023.1193791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction: Resistance to gemcitabine is common and critically limits its therapeutic efficacy in pancreatic ductal adenocarcinoma (PDAC). Methods: We constructed 17 patient-derived xenograft (PDX) models from PDAC patient samples and identified the most notable responder to gemcitabine by screening the PDX sets in vivo. To analyze tumor evolution and microenvironmental changes pre- and post-chemotherapy, single-cell RNA sequencing (scRNA-seq) was performed. Results: ScRNA-seq revealed that gemcitabine promoted the expansion of subclones associated with drug resistance and recruited macrophages related to tumor progression and metastasis. We further investigated the particular drug-resistant subclone and established a gemcitabine sensitivity gene panel (GSGP) (SLC46A1, PCSK1N, KRT7, CAV2, and LDHA), dividing PDAC patients into two groups to predict the overall survival (OS) in The Cancer Genome Atlas (TCGA) training dataset. The signature was successfully validated in three independent datasets. We also found that 5-GSGP predicted the sensitivity to gemcitabine in PDAC patients in the TCGA training dataset who were treated with gemcitabine. Discussion and conclusion: Our study provides new insight into the natural selection of tumor cell subclones and remodeling of tumor microenvironment (TME) cells induced by gemcitabine. We revealed a specific drug resistance subclone, and based on the characteristics of this subclone, we constructed a GSGP that can robustly predict gemcitabine sensitivity and prognosis in pancreatic cancer, which provides a theoretical basis for individualized clinical treatment.
Collapse
Affiliation(s)
- Zelin Hou
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jiajing Lin
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yuan Ma
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Haizhong Fang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yuwei Wu
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhijiang Chen
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xianchao Lin
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Fengchun Lu
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Shi Wen
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | | | - Heguang Huang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yu Pan
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
49
|
Peng M, Ying Y, Zhang Z, Liu L, Wang W. Reshaping the Pancreatic Cancer Microenvironment at Different Stages with Chemotherapy. Cancers (Basel) 2023; 15:2448. [PMID: 37173915 PMCID: PMC10177210 DOI: 10.3390/cancers15092448] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/09/2023] [Accepted: 04/23/2023] [Indexed: 05/15/2023] Open
Abstract
The dynamic tumor microenvironment, especially the immune microenvironment, during the natural progression and/or chemotherapy treatment is a critical frontier in understanding the effects of chemotherapy on pancreatic cancer. Non-stratified pancreatic cancer patients always receive chemotherapeutic strategies, including neoadjuvant chemotherapy and adjuvant chemotherapy, predominantly according to their physical conditions and different disease stages. An increasing number of studies demonstrate that the pancreatic cancer tumor microenvironment could be reshaped by chemotherapy, an outcome caused by immunogenic cell death, selection and/or education of preponderant tumor clones, adaptive gene mutations, and induction of cytokines/chemokines. These outcomes could in turn impact the efficacy of chemotherapy, making it range from synergetic to resistant and even tumor-promoting. Under chemotherapeutic impact, the metastatic micro-structures in the primary tumor may be built to leak tumor cells into the lymph or blood vasculature, and micro-metastatic/recurrent niches rich in immunosuppressive cells may be recruited by cytokines and chemokines, which provide housing conditions for these circling tumor cells. An in-depth understanding of how chemotherapy reshapes the tumor microenvironment may lead to new therapeutic strategies to block its adverse tumor-promoting effects and prolong survival. In this review, reshaped pancreatic cancer tumor microenvironments due to chemotherapy were reflected mainly in immune cells, pancreatic cancer cells, and cancer-associated fibroblast cells, quantitatively, functionally, and spatially. Additionally, small molecule kinases and immune checkpoints participating in this remodeling process caused by chemotherapy are suggested to be blocked reasonably to synergize with chemotherapy.
Collapse
Affiliation(s)
- Maozhen Peng
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (M.P.); (Y.Y.); (Z.Z.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ying Ying
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (M.P.); (Y.Y.); (Z.Z.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zheng Zhang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (M.P.); (Y.Y.); (Z.Z.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (M.P.); (Y.Y.); (Z.Z.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wenquan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (M.P.); (Y.Y.); (Z.Z.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
50
|
Kou M, Lu W, Zhu M, Qu K, Wang L, Yu Y. Massively recruited sTLR9 + neutrophils in rapidly formed nodules at the site of tumor cell inoculation and their contribution to a pro-tumor microenvironment. Cancer Immunol Immunother 2023:10.1007/s00262-023-03451-1. [PMID: 37079065 DOI: 10.1007/s00262-023-03451-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/10/2023] [Indexed: 04/21/2023]
Abstract
Neutrophils exert either pro- or anti-tumor activities. However, few studies have focused on neutrophils at the tumor initiation stage. In this study, we unexpectedly found a subcutaneous nodule in the groin areas of mice inoculated with tumor cells. The nodule was developed 24 h after the inoculation, filled with tumor cells and massively recruited neutrophils, being designated as tumor nodules. 22% of the neutrophils in tumor nodules are surface TLR9 (sTLR9) expressing neutrophils (sTLR9+ neutrophils). With tumor progression, sTLR9+ neutrophils were sustainably increased in tumor nodules/tumor tissues, reaching to 90.8% on day 13 after inoculation, with increased expression of IL-10 and decreased or no expression of TNFα. In vivo administration of CpG 5805 significantly reduced sTLR9 expression of the sTLR9+ neutrophils. The reduction of sTLR9 on neutrophils in tumor nodules contributed to the induction of an anti-tumor microenvironment conductive to the inhibition of tumor growth. Overall, the study provides insights for understanding the role of sTLR9+ neutrophils in the tumor development, especially in the early stage.
Collapse
Affiliation(s)
- Mengyuan Kou
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Wenting Lu
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Mengru Zhu
- Department of Developmental-Behavioral Pediatrics, The First Hospital of Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Kuo Qu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Liying Wang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China.
| | - Yongli Yu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China.
| |
Collapse
|