1
|
Jing F, Mu J, Liu J, Hu C, Wu F, Gao Q. Senescent vascular endothelial cells promote oral squamous cell carcinoma progression through complement C3 activation. Arch Oral Biol 2025; 174:106242. [PMID: 40158302 DOI: 10.1016/j.archoralbio.2025.106242] [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: 01/11/2025] [Revised: 03/09/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
OBJECTIVE The tumour microenvironment (TME) plays a critical role in therapeutic response and clinical outcomes in cancer. Senescent stromal cells have been shown to promote tumour progression; however, the role of senescent vascular endothelial cells (VECs) in oral squamous cell carcinoma (OSCC) remains largely unknown. In this study, we aimed to explore the effects and potential mechanisms of senescent VECs in OSCC progression. DESIGN Cisplatin was used to induce senescence in two endothelial cell lines. Senescence-associated β-galactosidase (SA-β-gal) staining, immunoblotting, cell cycle and proliferation assays, and migration and invasion assays were performed to access senescence development and biological behavior. Additionally, RNA sequencing analysis, multiplex immunohistochemical staining, immunoblotting, and xenograft mouse models were used to investigate the senescence-associated secretory phenotype of senescent VECs during OSCC progression and its potential molecular mechanisms. RESULTS Cisplatin-induced senescent VECs exhibited senescence-related changes, including positive SA-β-gal expression and upregulation of p16, p21, and p53, along with attenuated proliferation and migration. Notably, cisplatin-induced VEC senescence promoted OSCC cell proliferation, migration, and invasion by activating complement C3. Increased gene and protein levels of C3 were observed in cisplatin-treated senescent VECs. Inhibition of C3 in vitro and in vivo reduced OSCC cell proliferation and invasion. CONCLUSION Senescent VECs induced by cisplatin promote OSCC proliferation and invasion through complement C3 activation. Targeting complement C3 in senescent VECs may offer a novel therapeutic strategy for OSCC treatment.
Collapse
Affiliation(s)
- Fangqi Jing
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingtian Mu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Junjiang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Can Hu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Fanglong Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Qinghong Gao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
2
|
Harel M, Dahan N, Lahav C, Jacob E, Elon Y, Puzanov I, Kelly RJ, Shaked Y, Leibowitz R, Carbone DP, Gandara DR, Dicker AP. Decoding resistance to immune checkpoint inhibitors in non-small cell lung cancer: a comprehensive analysis of plasma proteomics and therapeutic implications. J Immunother Cancer 2025; 13:e011427. [PMID: 40404205 PMCID: PMC12097049 DOI: 10.1136/jitc-2024-011427] [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: 01/13/2025] [Accepted: 05/05/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have shown substantial benefit for patients with advanced non-small cell lung cancer (NSCLC). However, resistance to ICIs remains a major clinical challenge. Here, we perform a comprehensive bioinformatic analysis of plasma proteomic profiles to explore the underlying biology of treatment resistance in NSCLC. METHODS The analysis was performed on 388 "resistance-associated proteins" (RAPs) that were previously described as pretreatment plasma proteomic predictors within the PROphet computational model designed to predict ICI clinical benefit in NSCLC. Putative tissue origins of the RAPs were explored using publicly available datasets. Enrichment analyses were performed to investigate RAP-related biological processes. Plasma proteomic data from 50 healthy subjects and 272 patients with NSCLC were compared, where patients were classified as displaying clinical benefit (CB; n=76) or no CB (NCB; n=196). Therapeutic agents targeting RAPs were identified in drug and clinical trial databases. RESULTS The RAP set was significantly enriched with proteins associated with lung cancer, liver tissue, cell proliferation, extracellular matrix, invasion, and metastasis. Comparison of RAP expression in healthy subjects and patients with NSCLC revealed five distinct RAP subsets that provide mechanistic insights. The RAP subset displaying a pattern of high expression in the healthy population relative to the NSCLC population included multiple proteins associated with antitumor activities, while the subset displaying a pattern of highest expression in the NCB population included proteins associated with various hallmarks of treatment resistance. Analysis of patient-specific RAP profiles revealed inter-patient diversity of potential resistance mechanisms, suggesting that RAPs may aid in developing personalized therapeutic strategies. Furthermore, examination of drug and clinical trial databases revealed that 17.5% of the RAPs are drug targets, highlighting the RAP set as a valuable resource for drug development. CONCLUSIONS The study provides insight into the underlying biology of ICI resistance in NSCLC and highlights the potential clinical value of RAP profiles for developing personalized therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- The Roswell Park Comprehensive Cancer Center Data Bank and BioRepository, Buffalo, New York, USA
| | - Ronan J Kelly
- Department of Hematology and Oncology, Baylor University Medical Center at Dallas, Dallas, Texas, USA
| | - Yuval Shaked
- Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | | | | | - David R Gandara
- Division of Hematology/Oncology, UC Davis Comprehensive Cancer Center, Sacramento, California, USA
| | - Adam P Dicker
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
3
|
Zhang N, Li J, Ren Y, Xu Y. Comprehensive pan-cancer analysis of PPP1R3G reveals its diagnostic, prognostic, and immunotherapeutic implications. Discov Oncol 2025; 16:530. [PMID: 40232629 PMCID: PMC12000506 DOI: 10.1007/s12672-025-02361-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/10/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND PPP1R3G, a regulatory subunit of protein phosphatase 1, plays a critical role in glycogen metabolism and has been implicated in various cancers. This study provides a comprehensive pan-cancer analysis of PPP1R3G, evaluating its expression, diagnostic and prognostic significance, and potential as a therapeutic target. METHODS We performed an extensive pan-cancer analysis of PPP1R3G using several databases to assess its expression and investigate its correlations with clinical outcomes. Our investigation included assessing PPP1R3G's impact on survival, its correlation with immune checkpoints and tumor stemness scores, and its prognostic significance. We also explored its relationship with immunomodulators, genomic profiles, and immunological characteristics, as well as its response to immunotherapy and involvement in various biological pathways. RESULTS PPP1R3G expression varied significantly across different cancers and correlated with both diagnostic and prognostic outcomes. Moreover, PPP1R3G was significantly linked to immune checkpoints, immunomodulators, prognosis, immunoregulatory genes, tumor stemness, cellular function, and immune infiltration across numerous cancer types. Further analysis of PPP1R3G-related gene enrichment, mutation profiles, RNA modifications, and genomic heterogeneity revealed that missense mutations were the predominant alteration affecting PPP1R3G. CONCLUSIONS Overall, the expression of PPP1R3G is closely associated with various cancers and may serve as a potential biomarker for cancer detection.
Collapse
Affiliation(s)
- Nie Zhang
- Graduate School of Anhui Medical University, Hefei, China
- Key Laboratory of Gametes and Abnormal Reproductive Tract of National Health Commission, Anhui Medical University, Hefei, China
| | - Jiaoyu Li
- Graduate School of Anhui Medical University, Hefei, China
- Key Laboratory of Gametes and Abnormal Reproductive Tract of National Health Commission, Anhui Medical University, Hefei, China
| | - Yanzhi Ren
- Department of Cardiology, Shizhong District People's Hospital, Zaozhuang, China
| | - Yahui Xu
- Graduate School of Anhui Medical University, Hefei, China.
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
4
|
Garlanda C, Dambra M, Magrini E. Interplay between the complement system and other immune pathways in the tumor microenvironment. Semin Immunol 2025; 78:101951. [PMID: 40209638 DOI: 10.1016/j.smim.2025.101951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025]
Abstract
Tumor growth and spread are sustained by the tumor microenvironment. Inflammatory cells and pathways have a fundamental role in the tumor microenvironment, driving or conditioning the functional activation of other leukocyte subsets and favoring evasion of anti-tumor immunity. One of the inflammatory pathways contributing to cancer-related inflammation is the complement system. Complement has long been considered an immune mechanism associated with immunosurveillance. More recently it emerged as a tumor promoting pathway, due to direct effects on cancer cells or indirect effects via immunosuppression driven by myeloid cells. The role of complement in cancer is complex and ambiguous, and depends on the tumor type and stage, as well as other factors including oncogenic drivers, leukocyte infiltration, interactions with other tumor microenvironment components or tumor cells. Other factors of complexity include the source of complement molecules, its canonical or non-canonical extracellular functions, its potential intracellular activation, and the interaction with other systems, such as the coagulation or the microbiome. Preclinical studies generally demonstrate the involvement of complement activation in smouldering inflammation in cancer and promotion of an immunosuppressive environment. These studies paved the way for clinical trials aimed at enhancing the potential of immunotherapy, in particular by targeting complement-dependent myeloid-sustained immunosuppression. However, the complex role of complement in cancer and the multiplicity of complement players may represent stumbling blocks and account for failures of clinical trials, and suggest that further studies are required to identify patient subsets who may benefit from specific complement molecule targeting in combination with conventional therapies or immunotherapy. Here, we will discuss the anti- or pro-tumor role of complement activation in cancer, focusing on the interactions of complement with immune cells within the tumor microenvironment, in particular the myeloid compartment. Furthermore, we will examine the potential of complement targeting in cancer treatment, particularly in the context of macrophage reprogramming.
Collapse
Affiliation(s)
- Cecilia Garlanda
- Department of Biomedical Sciences, Humanitas University, Milan, Pieve Emanuele 20072, Italy; IRCCS, Humanitas Research Hospital, Milan, Rozzano 20089, Italy.
| | - Monica Dambra
- IRCCS, Humanitas Research Hospital, Milan, Rozzano 20089, Italy
| | - Elena Magrini
- IRCCS, Humanitas Research Hospital, Milan, Rozzano 20089, Italy
| |
Collapse
|
5
|
Hong Y, Liu Y, Shen H, Li B, Li Q. A strategy for synergistic enhancement of immune circulation in head and neck squamous cell carcinoma by novel nucleic acid drug therapy and immunotherapy. J Transl Med 2025; 23:354. [PMID: 40114181 PMCID: PMC11927285 DOI: 10.1186/s12967-025-06344-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/01/2025] [Indexed: 03/22/2025] Open
Abstract
Studies have shown that in the pathogenesis of head and neck squamous cell carcinoma, immune circulation obstruction caused by various factors including metabolic abnormalities, gene mutations, and matrix barrier, is a critical factor for the induction of tumor development and progression. Therefore, the immunotherapy strategy of killing head and neck squamous cell carcinoma cells by an enhanced immune circulation mechanism has attracted much attention. In addition, the rapid development of new nucleic acid drug therapy, such as mRNA, oligonucleotide and small guide RNA (sgRNA), has taken immunotherapy of head and neck squamous cell carcinoma (immune checkpoint inhibitors, tumor vaccines, cellular immunotherapy, cytokines and adjuvants, etc.) to a new level. The combination of nucleic acid therapy with immunotherapy developed for its therapeutic properties has brought a new direction for the diagnosis and treatment of head and neck squamous cell carcinoma, and the combination of the two has had considerable curative effect to patients with refractory/recurrent head and neck squamous cell carcinoma. In this review, we summarized the latest progress of nucleic acid therapy applied to conventional immunotherapy for head and neck squamous cell carcinoma, discussed its mechanism of action and efficacy, and looked into the future development trend.
Collapse
Affiliation(s)
- Yangjian Hong
- Key Laboratory of Head & Neck Cancer Translation Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Yanyang Liu
- Key Laboratory of Head & Neck Cancer Translation Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Huize Shen
- Key Laboratory of Head & Neck Cancer Translation Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Bowen Li
- Key Laboratory of Head & Neck Cancer Translation Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China.
- Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| | - Qinglin Li
- Key Laboratory of Head & Neck Cancer Translation Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Senent Y, Remírez A, Repáraz D, Llopiz D, Celias DP, Sainz C, Entrialgo-Cadierno R, Suarez L, Rouzaut A, Alignani D, Tavira B, Lambris JD, Woodruff TM, de Andrea CE, Ruffell B, Sarobe P, Ajona D, Pio R. The C5a/C5aR1 Axis Promotes Migration of Tolerogenic Dendritic Cells to Lymph Nodes, Impairing the Anticancer Immune Response. Cancer Immunol Res 2025; 13:384-399. [PMID: 39666368 DOI: 10.1158/2326-6066.cir-24-0250] [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: 03/15/2024] [Revised: 07/30/2024] [Accepted: 12/10/2024] [Indexed: 12/13/2024]
Abstract
The precise mechanisms by which the complement system contributes to the establishment of an immunosuppressive tumor microenvironment and promotes tumor progression remain unclear. In this study, we investigated the expression and function of complement C5a receptor 1 (C5aR1) in human and mouse cancer-associated dendritic cells (DC). First, we observed an overexpression of C5aR1 in tumor-infiltrating DCs, compared with DCs from the blood or spleen. C5aR1 expression was restricted to type 2 conventional DCs and monocyte-derived DCs, which displayed a tolerogenic phenotype capable of inhibiting T-cell activation and promoting tumor growth. C5aR1 engagement in DCs drove their migration from tumors to tumor-draining lymph nodes, where C5a levels were higher. We used this knowledge to optimize an anticancer therapy aimed at enhancing DC activity. In three syngeneic tumor models, C5aR1 inhibition significantly enhanced the efficacy of poly I:C, a Toll-like receptor 3 agonist, in combination with PD-1/PD-L1 blockade. The contribution of C5aR1 inhibition to the antitumor activity of the combination treatment relied on type 1 conventional DCs and antigen-specific CD8+ T cells, required lymphocyte egress from secondary lymphoid organs, and was associated with an increase in IFNγ signaling. In conclusion, our study highlights the importance of the C5a/C5aR1 axis in the biology of cancer-associated DCs and provides compelling evidence for the therapeutic potential of modulating the complement system to enhance DC-mediated immune responses against tumors.
Collapse
Affiliation(s)
- Yaiza Senent
- Cancer Division, Program in Solid Tumors, Cancer Center Clínica Universidad de Navarra (CCUN), Cima Universidad de Navarra, Pamplona, Spain
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
| | - Ana Remírez
- Cancer Division, Program in Solid Tumors, Cancer Center Clínica Universidad de Navarra (CCUN), Cima Universidad de Navarra, Pamplona, Spain
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - David Repáraz
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Program in Immunology and Immunotherapy, CCUN, Cima Universidad de Navarra, Pamplona, Spain
| | - Diana Llopiz
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Program in Immunology and Immunotherapy, CCUN, Cima Universidad de Navarra, Pamplona, Spain
| | - Daiana P Celias
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Cristina Sainz
- Cancer Division, Program in Solid Tumors, Cancer Center Clínica Universidad de Navarra (CCUN), Cima Universidad de Navarra, Pamplona, Spain
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Rodrigo Entrialgo-Cadierno
- Cancer Division, Program in Solid Tumors, Cancer Center Clínica Universidad de Navarra (CCUN), Cima Universidad de Navarra, Pamplona, Spain
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Lucia Suarez
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain
| | - Ana Rouzaut
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
| | - Diego Alignani
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
- Cytometry Unit, Cima Universidad de Navarra, Pamplona, Spain
| | - Beatriz Tavira
- Cancer Division, Program in Solid Tumors, Cancer Center Clínica Universidad de Navarra (CCUN), Cima Universidad de Navarra, Pamplona, Spain
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Queensland, Australia
| | | | - Brian Ruffell
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Pablo Sarobe
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Program in Immunology and Immunotherapy, CCUN, Cima Universidad de Navarra, Pamplona, Spain
| | - Daniel Ajona
- Cancer Division, Program in Solid Tumors, Cancer Center Clínica Universidad de Navarra (CCUN), Cima Universidad de Navarra, Pamplona, Spain
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Ruben Pio
- Cancer Division, Program in Solid Tumors, Cancer Center Clínica Universidad de Navarra (CCUN), Cima Universidad de Navarra, Pamplona, Spain
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|
7
|
Zhang X, Lan R, Liu Y, Pillarisetty VG, Li D, Zhao CL, Sarkar SA, Liu W, Hanna I, Gupta M, Hajdu C, Melamed J, Shusterman M, Widmer J, Allendorf J, Liu YZ. Complement activation in tumor microenvironment after neoadjuvant therapy and its impact on pancreatic cancer outcomes. NPJ Precis Oncol 2025; 9:58. [PMID: 40032924 PMCID: PMC11876354 DOI: 10.1038/s41698-025-00848-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 02/24/2025] [Indexed: 03/05/2025] Open
Abstract
Neoadjuvant therapy (NAT) is increasingly being used for pancreatic ductal adenocarcinoma (PDAC). This study investigates how NAT differentially impacts PDAC's carcinoma cells and the tumor microenvironment (TME). Spatial transcriptomics was used to compare gene expression profiles in carcinoma cells and the TME of 23 NAT-treated versus 13 NAT-naïve PDACs. Findings were validated by single-nucleus RNA sequencing (snRNA-seq) analysis. NAT induces apoptosis and inhibits proliferation of carcinoma cells and coordinately upregulates multiple complement genes (C1R, C1S, C3, C4B and C7) within the TME. Higher TME complement expression following NAT is associated with increased immunomodulatory and neurotrophic cancer-associated fibroblasts (CAFs); more CD4+ T cells; reduced immune exhaustion gene expression, and improved overall survival. snRNA-seq analysis demonstrates C3 complement is mainly upregulated in CAFs. These findings suggest that local complement dynamics could serve as a novel biomarker for prognosis, evaluating treatment response, and guiding therapeutic strategies in NAT-treated PDAC patients.
Collapse
Affiliation(s)
- Xiaofei Zhang
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA.
| | - Ruoxin Lan
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Yongjun Liu
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Venu G Pillarisetty
- Department of Surgery, University of Washington School of Medicine, Seattle, WA, USA
| | - Danting Li
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Chaohui L Zhao
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Suparna A Sarkar
- Department of Pathology and Laboratory Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Weiguo Liu
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Iman Hanna
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Mala Gupta
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Cristina Hajdu
- Department of Pathology and Laboratory Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Jonathan Melamed
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Michael Shusterman
- Department of Oncology, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Jessica Widmer
- Department of Gastroenterology, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - John Allendorf
- Department of Surgery, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Yao-Zhong Liu
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA.
| |
Collapse
|
8
|
Ovcinnikovs V, Dijkman K, Zom GG, Beurskens FJ, Trouw LA. Enhancing complement activation by therapeutic anti-tumor antibodies: Mechanisms, strategies, and engineering approaches. Semin Immunol 2025; 77:101922. [PMID: 39742715 DOI: 10.1016/j.smim.2024.101922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/06/2024] [Accepted: 12/07/2024] [Indexed: 01/04/2025]
Abstract
The complement system plays an integral role in both innate and adaptive immune responses. Beyond its protective function against infections, complement is also known to influence tumor immunity, where its activation can either promote tumor progression or mediate tumor cell destruction, depending on the context. One such context can be provided by antibodies, with their inherent capacity to activate the classical complement pathway. In recent years, our understanding of the mechanisms governing complement activation by IgG and IgM antibodies has expanded significantly. At the same time, preclinical and clinical studies on antibodies such as rituximab, ofatumumab, and daratumumab have provided evidence for the role of complement in therapeutic success, encouraging strategies to further enhance its activity. In this review we examine the main determinants of antibody-mediated complement activation, highlighting the importance of antibody subclass, affinity, valency, and geometry of antigen engagement. We summarize the evidence for complement involvement in anti-tumor activity and challenges of accurately estimating the extent of its contribution to therapeutic efficacy. Furthermore, we explore several engineering approaches designed to enhance complement activation, including increased Fc oligomerization and C1q affinity, bispecific C1q-recruiting antibodies, IgG subclass chimeras, as well as antibody and paratope combinations. Strategies targeting membrane-bound complement regulatory proteins to overcome tumor-associated complement inhibition are also discussed as a method to boost therapeutic efficacy. Finally, we highlight the potential of complement-dependent cellular cytotoxicity (CDCC) and complement-dependent cellular phagocytosis (CDCP) as effector mechanisms that warrant deeper investigation. By integrating advances in antibody and complement biology with insights from efforts to enhance complement activation in therapeutic antibodies, this review aims to provide a comprehensive framework of antibody design and engineering strategies that optimize complement activity for improved anti-tumor efficacy.
Collapse
Affiliation(s)
| | - Karin Dijkman
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | | | | - Leendert A Trouw
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
9
|
Zhou L, Fan S, Zhang W, Gong Z, Wang D, Tang D. The battle within: cell death by phagocytosis in cancer. Clin Transl Oncol 2025; 27:871-886. [PMID: 39167272 DOI: 10.1007/s12094-024-03650-x] [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: 06/06/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
The process by which living cells are phagocytosed and digested to death is called cell death by phagocytosis, a term that has just recently been generalized and redefined. It is characterized by the phagocytosis of living cells and the cessation of cell death by phagocytosis. Phagocytosis of dead cells is a widely discussed issue in cancer, cell death by phagocytosis can stimulate phagocytosis and stimulate adaptive immunity in tumors, and at the same time, do not-eat-me signaling is an important site for cancer cells to evade recognition by phagocytes. Therefore, we discuss in this review cell death by phagocytosis occurring in cancer tissues and emphasize the difference between this new concept and the phagocytosis of dead tumor cells. Immediately thereafter, we describe the mechanisms by which cell death by phagocytosis occurs and how tumors escape phagocytosis. Finally, we summarize the potential clinical uses of cell death by phagocytosis in tumor therapy and strive to provide ideas for tumor therapy.
Collapse
Affiliation(s)
- Lujia Zhou
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Shiying Fan
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Wenjie Zhang
- School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Zhiyuan Gong
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225000, China.
| |
Collapse
|
10
|
Ricciuti J, Liu Q, Khan ANMNH, Joseph JM, Veuskens BR, Giridharan T, Suzuki S, Emmons TR, Yaffe MB, Kuijpers TW, Jongerius I, Brouwer MC, Pouw RB, Odunsi K, Frederick P, Mager KL, Lele S, Gaulin N, Hakim C, Edwards RP, Olawaiye AB, Sukumanovich P, Taylor S, Elishaev E, Zsiros E, Modugno F, Moysich KB, Segal BH. Prognostic significance of serum complement activation, neutrophil extracellular traps and extracellular DNA in newly diagnosed epithelial ovarian cancer. Gynecol Oncol 2025; 193:49-57. [PMID: 39764857 PMCID: PMC11871990 DOI: 10.1016/j.ygyno.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/25/2024] [Accepted: 12/05/2024] [Indexed: 02/02/2025]
Abstract
PURPOSE We observed that the tumor microenvironment (TME) in metastatic epithelial ovarian cancer (EOC) and in other solid tumors can reprogram normal neutrophils to acquire a complement-dependent suppressor phenotype characterized by inhibition of stimulated T cell activation. This study aims to evaluate whether serum markers of neutrophil activation and complement at diagnosis of EOC would be associated with clinical outcomes. EXPERIMENTAL DESIGN We conducted a two-center prospective study of patients with newly diagnosed EOC (N = 188). Blood and ascites fluid were collected at diagnosis for biomarker analysis. Patients were evaluated for progression-free survival (PFS) and overall survival (OS). RESULTS The median OS was 47 months (95 % CI: 34-58) and the median PFS was 12 months (95 % CI: 11-15). Pre-treatment serum levels of genomic DNA (gDNA), markers of neutrophil degranulation (myeloperoxidase [MPO]) and neutrophil extracellular traps (NETs) (citrullinated histone H3 [CitH3]), and complement activation (C3b/c) were each associated with worse OS in univariate analysis. In multivariate analyses controlling for age, stage, and optimal debulking, serum gDNA, MPO, and CitH3 remained associated with worse OS, while C3b/c levels were not. In an exploratory analysis, the largest magnitude of difference in 2-year OS occurred in patients with low C3b/c and low CitH3 compared to all other patients (87 % vs 46 % survival, respectively). In ascites fluid, increased factor H, a negative regulator of complement activation, was associated with improved OS in univariate analysis. CONCLUSIONS These results point to serum gDNA, NETs, and complement activation as potential prognostic biomarkers in patients with newly diagnosed EOC.
Collapse
Affiliation(s)
- Jason Ricciuti
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
- Dr. Ricciuti is currently in the Division of Gynecologic Oncology, Saint Louis University School of Medicine, Saint Louis, MO
| | - Qian Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - ANM Nazmul H. Khan
- Department of Internal Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Janine M. Joseph
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Bert R.J. Veuskens
- Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Institute for Immunology and Infection Diseases, Amsterdam, the Netherlands
| | | | - Sora Suzuki
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Tiffany R. Emmons
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Michael B. Yaffe
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Division of Acute Care Surgery, Trauma and Surgical Critical Care, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Taco W. Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Amsterdam, the Netherlands
- Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Institute for Immunology and Infection Diseases, Amsterdam, the Netherlands
| | - Ilse Jongerius
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Amsterdam, the Netherlands
- Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Institute for Immunology and Infection Diseases, Amsterdam, the Netherlands
- Current affiliation Genmab, Utrecht, the Netherland
| | - Mieke C. Brouwer
- Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Institute for Immunology and Infection Diseases, Amsterdam, the Netherlands
| | - Richard B. Pouw
- Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Institute for Immunology and Infection Diseases, Amsterdam, the Netherlands
- Sanquin Diagnostic Services, Amsterdam, the Netherlands
| | - Kunle Odunsi
- University of Chicago Medicine Comprehensive Cancer Center, Chicago, IL
| | - Peter Frederick
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Katherine LaVigne Mager
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
- Dr. Ricciuti is currently in the Division of Gynecologic Oncology, Saint Louis University School of Medicine, Saint Louis, MO
| | - Shashikant Lele
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
- Dr. Ricciuti is currently in the Division of Gynecologic Oncology, Saint Louis University School of Medicine, Saint Louis, MO
| | - Nicole Gaulin
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
- Dr. Ricciuti is currently in the Division of Gynecologic Oncology, Saint Louis University School of Medicine, Saint Louis, MO
| | - Christiane Hakim
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Robert P. Edwards
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Women’s Cancer Research Center, Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA
| | - Alexander B. Olawaiye
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Paniti Sukumanovich
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Sarah Taylor
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Women’s Cancer Research Center, Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA
| | - Esther Elishaev
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Women’s Cancer Research Center, Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA
| | - Emese Zsiros
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
- Dr. Ricciuti is currently in the Division of Gynecologic Oncology, Saint Louis University School of Medicine, Saint Louis, MO
| | - Francesmary Modugno
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health
- Women’s Cancer Research Center, Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA
| | - Kirsten B. Moysich
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Brahm H. Segal
- Departments of Internal Medicine and Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
- Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| |
Collapse
|
11
|
Wang M, Yu F, Zhang Y. Present and future of cancer nano-immunotherapy: opportunities, obstacles and challenges. Mol Cancer 2025; 24:26. [PMID: 39827147 PMCID: PMC11748575 DOI: 10.1186/s12943-024-02214-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/25/2024] [Indexed: 01/22/2025] Open
Abstract
Clinically, multimodal therapies are adopted worldwide for the management of cancer, which continues to be a leading cause of death. In recent years, immunotherapy has firmly established itself as a new paradigm in cancer care that activates the body's immune defense to cope with cancer. Immunotherapy has resulted in significant breakthroughs in the treatment of stubborn tumors, dramatically improving the clinical outcome of cancer patients. Multiple forms of cancer immunotherapy, including immune checkpoint inhibitors (ICIs), adoptive cell therapy and cancer vaccines, have become widely available. However, the effectiveness of these immunotherapies is not much satisfying. Many cancer patients do not respond to immunotherapy, and disease recurrence appears to be unavoidable because of the rapidly evolving resistance. Moreover, immunotherapies can give rise to severe off-target immune-related adverse events. Strategies to remove these hindrances mainly focus on the development of combinatorial therapies or the exploitation of novel immunotherapeutic mediations. Nanomaterials carrying anticancer agents to the target site are considered as practical approaches for cancer treatment. Nanomedicine combined with immunotherapies offers the possibility to potentiate systemic antitumor immunity and to facilitate selective cytotoxicity against cancer cells in an effective and safe manner. A myriad of nano-enabled cancer immunotherapies are currently under clinical investigation. Owing to gaps between preclinical and clinical studies, nano-immunotherapy faces multiple challenges, including the biosafety of nanomaterials and clinical trial design. In this review, we provide an overview of cancer immunotherapy and summarize the evidence indicating how nanomedicine-based approaches increase the efficacy of immunotherapies. We also discuss the key challenges that have emerged in the era of nanotechnology-based cancer immunotherapy. Taken together, combination nano-immunotherapy is drawing increasing attention, and it is anticipated that the combined treatment will achieve the desired success in clinical cancer therapy.
Collapse
Affiliation(s)
- Man Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 38 Dengzhou Road, Qingdao, 266021, China.
| | - Fei Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 38 Dengzhou Road, Qingdao, 266021, China
| | - Yuan Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 38 Dengzhou Road, Qingdao, 266021, China
| |
Collapse
|
12
|
Paredes-Villa AA, Aguilar-Arce IE, Meneses-Morales I, Cervantes-Roldán R, Valadéz-Graham V, León-Del-Río A. NHERF2 regulatory function in signal transduction pathways and control of gene expression: Implications for cellular homeostasis and breast cancer. Arch Med Res 2025; 56:103179. [PMID: 39813852 DOI: 10.1016/j.arcmed.2024.103179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/20/2024] [Accepted: 12/27/2024] [Indexed: 01/18/2025]
Abstract
Na⁺/H⁺ exchanger regulatory factor 2 (NHERF2) is a nucleocytoplasmic protein initially identified as a regulator of membrane-bound sodium-hydrogen exchanger 3 (NHE3). In the cytoplasm, NHERF2 regulates the activity of G protein-coupled receptors (GPCRs), including beta-2 adrenergic receptor (2β-AR), lysophosphatidic acid receptor 2, and parathyroid hormone type 1 receptor. In the nucleus, NHERF2 acts as a coregulator of transcription factors such as sex-determining region Y protein (SRY), involved in male sex determination, and estrogen receptor alpha (ERα). ERα is a ligand-dependent transcription factor that controls mammary gland growth and differentiation during puberty and pregnancy and plays a major role in the development and progression of breast cancer tumors. Altogether, the regulatory functions of NHERF2 on ion channels, GPCRs, and nuclear transcription factors have a modulatory effect on signal transduction pathways, metabolic homeostasis, cell proliferation and differentiation, neurotransmission, muscle contraction, and renal electrolyte balance. This work highlights NHERF2 functions in the cytoplasm and nucleus and underscores the nuclear mechanisms through which NHERF2 participates in the regulation of gene expression and tumor growth and progression in breast cancer.
Collapse
Affiliation(s)
- Adrián-Alejandro Paredes-Villa
- Programa de Investigación de Cancer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico; Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico; Posgrado en Ciencias Biológicas, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Isaac Esaú Aguilar-Arce
- Programa de Investigación de Cancer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico; Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico; Posgrado en Ciencias Biomédicas, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Iván Meneses-Morales
- Facultad de Ciencias Químicas, Universidad Juárez del Estado de Durango, Durango, Mexico
| | - Rafael Cervantes-Roldán
- Programa de Investigación de Cancer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico; Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Viviana Valadéz-Graham
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Alfonso León-Del-Río
- Programa de Investigación de Cancer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico; Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
13
|
Ni Q, Yang H, Rao H, Zhang L, Xiong M, Han X, Deng B, Wang L, Chen J, Shi Y. The role of the C5a-C5aR pathway in iron metabolism and gastric cancer progression. Front Immunol 2025; 15:1522181. [PMID: 39850877 PMCID: PMC11754390 DOI: 10.3389/fimmu.2024.1522181] [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: 11/04/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
Gastric cancer continues to be a leading global health concern, with current therapeutic approaches requiring significant improvement. While the disruption of iron metabolism in the advancement of gastric cancer has been well-documented, the underlying regulatory mechanisms remain largely unexplored. Additionally, the complement C5a-C5aR pathway has been identified as a crucial factor in gastric cancer development. The impact of the complement system on iron metabolism and its role in gastric cancer progression is an area warranting further investigation. Our research demonstrates that the C5a-C5aR pathway promotes gastric cancer progression by enhancing iron acquisition in tumor cells through two mechanisms. First, it drives macrophage polarization toward the M2 phenotype, which has a strong iron-release capability. Second, it increases the expression of LCN2, a high-affinity iron-binding protein critical for iron export from tumor-associated macrophages, by activating endoplasmic reticulum stress in these cells. Both mechanisms facilitate the transfer of iron from macrophages to cancer cells, thereby promoting tumor cell proliferation. This study aims to elucidate the connection between the complement C5a-C5aR pathway and iron metabolism within the tumor microenvironment. Our data suggest a pivotal role of the C5a-C5aR pathway in tumor iron management, indicating that targeting its regulatory mechanisms may pave the way for future iron-targeted therapeutic approaches in cancer treatment.
Collapse
Affiliation(s)
- Qinxue Ni
- The First Affiliated Hospital of Army Military Medical University, Department of General Surgery, Chongqing, China
| | - Hong Yang
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Hang Rao
- The First Affiliated Hospital of Army Military Medical University, Department of General Surgery, Chongqing, China
| | - Liyong Zhang
- The First Affiliated Hospital of Army Military Medical University, Department of General Surgery, Chongqing, China
| | - Mengyuan Xiong
- The First Affiliated Hospital of Army Military Medical University, Department of General Surgery, Chongqing, China
| | - Xiao Han
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Boshao Deng
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lulu Wang
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jian Chen
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yan Shi
- The First Affiliated Hospital of Army Military Medical University, Department of General Surgery, Chongqing, China
| |
Collapse
|
14
|
Sharma M, Alessandro P, Cheriyamundath S, Lopus M. Therapeutic and diagnostic applications of carbon nanotubes in cancer: recent advances and challenges. J Drug Target 2024; 32:287-299. [PMID: 38252035 DOI: 10.1080/1061186x.2024.2309575] [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: 10/07/2023] [Accepted: 01/11/2024] [Indexed: 01/23/2024]
Abstract
Carbon nanotubes (CNTs) are allotropes of carbon, composed of carbon atoms forming a tube-like structure. Their high surface area, chemical stability, and rich electronic polyaromatic structure facilitate their drug-carrying capacity. Therefore, CNTs have been intensively explored for several biomedical applications, including as a potential treatment option for cancer. By incorporating smart fabrication strategies, CNTs can be designed to specifically target cancer cells. This targeted drug delivery approach not only maximizes the therapeutic utility of CNTs but also minimizes any potential side effects of free drug molecules. CNTs can also be utilised for photothermal therapy (PTT) which uses photosensitizers to generate reactive oxygen species (ROS) to kill cancer cells, and in immunotherapeutic applications. Regarding the latter, for example, CNT-based formulations can preferentially target intra-tumoural regulatory T-cells. CNTs also act as efficient antigen presenters. With their capabilities for photoacoustic, fluorescent and Raman imaging, CNTs are excellent diagnostic tools as well. Further, metallic nanoparticles, such as gold or silver nanoparticles, are combined with CNTs to create nanobiosensors to measure biological reactions. This review focuses on current knowledge about the theranostic potential of CNT, challenges associated with their large-scale production, their possible side effects and important parameters to consider when exploring their clinical usage.
Collapse
Affiliation(s)
- Muskan Sharma
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari, Mumbai, India
| | - Parodi Alessandro
- Department of Translational Medicine, Sirius University of Science and Technology, Sirius, Russia
| | - Sanith Cheriyamundath
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari, Mumbai, India
| | - Manu Lopus
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari, Mumbai, India
| |
Collapse
|
15
|
Prado LG, Nagy LE. Role of Complement in Liver Diseases. Semin Liver Dis 2024; 44:510-522. [PMID: 39608405 DOI: 10.1055/s-0044-1795143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
This review aims to summarize recent research using animal models, cell models, and human data regarding the role of complement in liver disease. Complement is part of the innate immune system and was initially characterized for its role in control of pathogens. However, evidence now indicates that complement also plays an important role in the response to cellular injury that is independent of pathogens. The liver is the main organ responsible for producing circulating complement. In response to liver injury, complement is activated and likely plays a dual role, both contributing to and protecting from injury. In uncontrolled complement activation, cell injury and liver inflammation occur, contributing to progression of liver disease. Complement activation is implicated in the pathogenesis of multiple liver diseases, including alcohol-associated liver disease, metabolic dysfunction-associated steatotic liver disease, fibrosis and cirrhosis, hepatocellular carcinoma, and autoimmune hepatitis. However, the mechanisms by which complement is overactivated in liver diseases are still being unraveled.
Collapse
Affiliation(s)
- Luan G Prado
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio
| | - Laura E Nagy
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
16
|
Yang H, Li G, Zhang J, Zhao J, Zhao Y, Wu Y, Sun Z, Song S, Zou Y, Zou Z, Han X, Deng B, Wang L, Rao H, Xu G, Wang S, Guo S, Ding H, Shi Y, Wu Y, Chen J. A novel hollow iron nanoparticle system loading PEG-Fe 3O 4 with C5a receptor antagonist for breast cancer treatment. Front Immunol 2024; 15:1466180. [PMID: 39483473 PMCID: PMC11524822 DOI: 10.3389/fimmu.2024.1466180] [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: 07/17/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
Breast cancer is the most diagnosed malignancy and major cause of cancer death among women population in the worldwide. Ferroptosis is a recently discovered iron-dependent regulated cell death involved in tumor progression and therapeutic response. Moreover, increasing studies have implied that ferroptosis is a promising approach to eliminating cancer cells like developing iron nanoparticles as a therapeutic agent. However, resistance to ferroptosis is a vital distinctive hallmark of cancer. Therefore, further investigation of the mechanism of ferroptosis resistance to enhance its tumor sensitivity is essential for ferroptosis-target breast cancer therapy. Our results revealed that the activation of C5a/C5aR pathway can drive resistance to ferroptosis and reshaping breast cancer immune microenvironment. Accordingly, loading PEG-Fe3O4 with C5aRA significantly improved the anti-tumor effect of PEG- Fe3O4 by inhibiting ferroptosis resistance and increasing macrophage polarization toward M1 phenotype. Our findings presented a novel cancer therapy strategy that combined cancer cell metal metabolism regulation and immunotherapy. The study also provided support for further evaluation of PEG- Fe3O4@C5aRA as a novel therapeutic strategy for breast cancer in clinical trials.
Collapse
Affiliation(s)
- Hong Yang
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Guiqing Li
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ji Zhang
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jing Zhao
- Biomedical Analysis Center, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yunpei Zhao
- Department of Cardio-renal, Chinese People’s Liberation Army 74th Group Military Hospital, Guangzhou, China
| | - Yufei Wu
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zihan Sun
- Breast Disease Center, Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shuangshuang Song
- The First Affiliated Hospital of Army Military Medical University, Department of General Practice, Chongqing, China
| | - Ying Zou
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhihao Zou
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiao Han
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Boshao Deng
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lulu Wang
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Hang Rao
- Department of General Surgery, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Guilian Xu
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shufeng Wang
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Sheng Guo
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Huanyu Ding
- Institute of Medical Technology, Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Yan Shi
- Department of General Surgery, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yuzhang Wu
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jian Chen
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
17
|
Chen C, Yuan M, Xia L, Wu X, Zhong X, Zhang H, Zhang L, Liu X, Wang Z, Sun C. Expression of CREBBP and EP300 Associated With Tumor Volume in Patients With Grade-3 Glioma: A Retrospective Analysis. Clin Med Insights Oncol 2024; 18:11795549241287777. [PMID: 39429683 PMCID: PMC11490948 DOI: 10.1177/11795549241287777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 09/11/2024] [Indexed: 10/22/2024] Open
Abstract
Background Reliable predictive data are crucial for making accurate treatment decisions in glioma patients, but it can be challenging to obtain due to limited information in many cases. Numerous research studies have indicated the involvement of cyclic adenosine monophosphate (cAMP)-response element binding protein (CREBBP) and E1A binding protein p300 (EP300) in tumorigenesis and tumor progression across various types. Methods The messenger RNA (mRNA) expression levels of CREBBP and EP300 were retrospectively analyzed in 17 grade-3 glioma patients. The SYBR Green real-time polymerase chain reaction (RT-PCR) technique was employed for mRNA expression analysis, with the glyceraldehyde-3-phosphate dehydrogenase gene (GAPDH) used as a reference gene for data normalization. In addition, the relationship between CREBBP, EP300 expression and patients' clinical information, imaging features, histologic features, immune factors, and overall survival was assessed through univariate analyses. Results The analysis of the data unveiled a statistically significant upregulation of CREBBP and EP300 mRNA expression levels in large gliomas as compared with their smaller counterparts (P < .05). Histological examination using hematoxylin and eosin (H&E) staining exhibited marked cellular heterogeneity, with heightened cell density observed specifically within tumors displaying elevated CREBBP expression levels. In contrast, there was a substantial downregulation of complement 3 and complement 4 within larger tumor volumes when compared with smaller ones (P < .05). However, these findings do not serve as clinically relevant prognostic indicators for glioma. Conclusions It is suggested that higher expression levels of CREBBP and EP300 are positively associated with increased tumor volume. Inhibition of CREBBP and EP300 enhances local immunogenicity, leading to the recruitment of immune cells and release of cytokines for effective tumor eradication, ultimately resulting in the inhibition of tumor growth.
Collapse
Affiliation(s)
- Cuiwei Chen
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Meiqin Yuan
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Liang Xia
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Xin Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xingguang Zhong
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Huangjie Zhang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Lidan Zhang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Xuan Liu
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Zeng Wang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Integrated Traditional Chinese and Western Medicine Oncology Laboratory, Key Laboratory of Traditional Chinese Medicine of Zhejiang Province, Hangzhou, China
| | - Caixing Sun
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
18
|
Zuo A, Lv J, Jia W, Ba Y, Liu S, Zhang Y, Weng S, Xu H, Liu L, Wang L, Han X, Liu Z. High ratio of resident to exhausted CD4 + T cells predicts favorable prognosis and potentially better immunotherapeutic efficacy in hepatocellular carcinoma. BMC Cancer 2024; 24:1152. [PMID: 39289669 PMCID: PMC11409587 DOI: 10.1186/s12885-024-12916-0] [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: 06/20/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Tumor-infiltrating lymphocytes (TILs) are significantly implicated in regulating the tumor immune microenvironment (TIME) and immunotherapeutic response. However, little is known about the impact of the resident and exhausted status of TILs in hepatocellular carcinoma (HCC). METHODS Single-cell RNA sequencing data was applied to discover resident and exhausted signatures of TILs. Survival outcomes, biological function, immune infiltration, genomic variation, immunotherapeutic efficacy, and sorafenib response were further explored the clinical significance and molecular association of TILs in HCC. Moreover, a candidate gene with predictive capability for the dismal subtype was identified through univariate Cox regression analysis, survival analysis, and the BEST website. RESULTS Single-cell analysis revealed that CD8 + T, CD4 + T, and NK cells were strongly associated with resident and exhausted patterns. Specific resident and exhausted signatures for each subpopulation were extracted in HCC. Further multivariate Cox analysis revealed that the ratio of resident to exhausted CD4 + T cells in TIME was an independent prognostic factor. After incorporating tumor purity with the ratio of resident to exhausted CD4 + T cells, we stratified HCC patients into three subtypes and found that (i) CD4 residencyhighexhaustionlow subtype was endowed with favorable prognosis, immune activation, and sensitivity to immunotherapy; (ii) CD4 exhaustionhighresidencylow subtype was characterized by genome instability and sensitivity to sorafenib; (iii) Immune-desert subtype was associated with malignant-related pathways and poor prognosis. Furthermore, spindle assembly abnormal protein 6 homolog (SASS6) was identified as a key gene, which accurately predicted the immune-desert subtype. Prognostic analysis as well as in vitro and in vivo experiments further demonstrated that SASS6 was closely associated with tumor prognosis, proliferation, and migration. CONCLUSIONS The ratio of resident to exhausted CD4 + T cells shows promise as a potential biomarker for HCC prognosis and immunotherapy response and SASS6 may serve as a biomarker and therapeutic target for prognostic assessment of HCC.
Collapse
Affiliation(s)
- Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China
| | - Jinxiang Lv
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Wenlong Jia
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shutong Liu
- School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Long Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, 710049, China
| | - Libo Wang
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China.
| |
Collapse
|
19
|
Du Z, Sui D, Xin D, Tang X, Li M, Liu X, Deng Y, Song Y. Sialic acid-modified doxorubicin liposomes target tumor-related immune cells to relieve multiple inhibitions of CD8 + T cells. J Liposome Res 2024; 34:464-474. [PMID: 38196168 DOI: 10.1080/08982104.2023.2298901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/18/2023] [Indexed: 01/11/2024]
Abstract
In different types of cancer treatments, cancer-specific T cells are required for effective anticancer immunity, which has a central role in cancer immunotherapy. However, due to the multiple inhibitions of CD8+ T cells by tumor-related immune cells, CD8+ T-cell mediated antitumor immunotherapy has not achieved breakthrough progress in the treatment of solid tumors. Receptors for sialic acid (SA) are highly expressed in tumor-associated immune cells, so SA-modified nanoparticles are a drug delivery nanoplatform using tumor-associated immune cells as vehicles. To relieve the multiple inhibitions of CD8+ T cells by tumor-associated immune cells, we prepared SA-modified doxorubicin liposomes (SL-DOX, Scheme 1A). In our study, free SA decreased the toxicity of SL-DOX to tumor-associated immune cells. Compared with common liposomes, SL-DOX could inhibit tumor growth more effectively. It is worth noting that SL-DOX could not only kill tumor-related neutrophils and monocytes to relieve the multiple inhibitions of CD8+ T cells but also induce immunogenic death of tumor cells to promote the infiltration and differentiation of CD8+ T cells (Scheme 1B). Therefore, SL-DOX has potential value for the clinical therapeutic effect of CD8+ T cells mediating anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Zhouchunxiao Du
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Dezhi Sui
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Dongzhe Xin
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xueying Tang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Mingze Li
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xinrong Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
20
|
Liu C, Liu L. Identification and immunoassay of prognostic genes associated with the complement system in acute myeloid leukemia. J Formos Med Assoc 2024; 123:904-915. [PMID: 38341328 DOI: 10.1016/j.jfma.2024.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/12/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Studies have associated the development of pulmonary leukemia with the activation of the complement system. However, the roles and mechanisms of complement system-related genes (CSRGs) in acute myeloid leukemia (AML) have not been investigated extensively. This study used The Cancer Genome Atlas (TCGA)-AML and GSE37642 datasets. Differentially expressed CSRGs (CSRDEGs) were identified by overlapping genes differentially expressed between the high and low CSRG score groups and key module genes identified in a weighted gene co-expression network analysis. Univariate and multivariate Cox analyses identified CSRG-related biomarkers, which were used to build a prognostic model. After gene set enrichment analysis (GSEA), immune-related and drug-sensitivity analyses were performed in the high- and low-risk groups. Four prognosis-related biomarkers were identified and used to develop a prognostic model: MEOX2, IGFBP5, CH25H, and RAB3B. The model's performance was verified in a test cohort (a subset of samples from the TCGA-AML dataset) and a validation cohort (GSE37642). The GSEA revealed that the high-risk group was mainly enriched for Golgi organization and cytokine-cytokine receptor interactions, and the low-risk group was mainly enriched in the hedgehog signaling pathway and spliceosome. Lastly, two immune cells were found to show differential infiltration between risk groups, which correlated with the risk scores. M1 macrophage infiltration was significantly positively correlated with RAB3B expression. Sensitivity to 36 drugs differed significantly between risk groups. This study screened four CSRG-related biomarkers (MEOX2, IGFBP5, CH25H, and RAB3B) to provide a basis for predicting AML prognosis.
Collapse
Affiliation(s)
- Chen Liu
- Department of Hematology, First Affiliated Hospital of Chongqing Medical University, ChongQing, 400016, China.
| | - Lin Liu
- Department of Hematology, First Affiliated Hospital of Chongqing Medical University, ChongQing, 400016, China.
| |
Collapse
|
21
|
Cao X, Hu Z, Sheng X, Sun Z, Yang L, Shu H, Liu X, Yan G, Zhang L, Liu C, Zhang Y, Wang H, Lu H. Glyco-signatures in patients with advanced lung cancer during anti-PD-1/PD-L1 immunotherapy. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1099-1107. [PMID: 38952341 PMCID: PMC11464919 DOI: 10.3724/abbs.2024110] [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: 01/23/2024] [Accepted: 03/25/2024] [Indexed: 07/03/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) targeting programmed cell death 1/programmed cell death ligand-1 (PD-1/PD-L1) have significantly prolonged the survival of advanced/metastatic patients with lung cancer. However, only a small proportion of patients can benefit from ICIs, and clinical management of the treatment process remains challenging. Glycosylation has added a new dimension to advance our understanding of tumor immunity and immunotherapy. To systematically characterize anti-PD-1/PD-L1 immunotherapy-related changes in serum glycoproteins, a series of serum samples from 12 patients with metastatic lung squamous cell carcinoma (SCC) and lung adenocarcinoma (ADC), collected before and during ICIs treatment, are firstly analyzed with mass-spectrometry-based label-free quantification method. Second, a stratification analysis is performed among anti-PD-1/PD-L1 responders and non-responders, with serum levels of glycopeptides correlated with treatment response. In addition, in an independent validation cohort, a large-scale site-specific profiling strategy based on chemical labeling is employed to confirm the unusual characteristics of IgG N-glycosylation associated with anti-PD-1/PD-L1 treatment. Unbiased label-free quantitative glycoproteomics reveals serum levels' alterations related to anti-PD-1/PD-L1 treatment in 27 out of 337 quantified glycopeptides. The intact glycopeptide EEQFN 177STYR (H3N4) corresponding to IgG4 is significantly increased during anti-PD-1/PD-L1 treatment (FC=2.65, P=0.0083) and has the highest increase in anti-PD-1/PD-L1 responders (FC=5.84, P=0.0190). Quantitative glycoproteomics based on protein purification and chemical labeling confirms this observation. Furthermore, obvious associations between the two intact glycopeptides (EEQFN 177STYR (H3N4) of IgG4, EEQYN 227STFR (H3N4F1) of IgG3) and response to treatment are observed, which may play a guiding role in cancer immunotherapy. Our findings could benefit future clinical disease management.
Collapse
Affiliation(s)
- Xinyi Cao
- Institutes of Biomedical Sciences and Shanghai Cancer CenterFudan UniversityShanghai200032China
- Department of Laboratory MedicineHuashan HospitalFudan UniversityShanghai200040China
| | - Zhihuang Hu
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | | | - Zhenyu Sun
- Institutes of Biomedical Sciences and Shanghai Cancer CenterFudan UniversityShanghai200032China
| | - Lijun Yang
- Department of ChemistryFudan UniversityShanghai200433China
| | - Hong Shu
- Department of Clinical LaboratoryGuangxi Medical University Cancer HospitalNanning530021China
| | - Xiaojing Liu
- Department of ChemistryFudan UniversityShanghai200433China
| | - Guoquan Yan
- Institutes of Biomedical Sciences and Shanghai Cancer CenterFudan UniversityShanghai200032China
| | - Lei Zhang
- Institutes of Biomedical Sciences and Shanghai Cancer CenterFudan UniversityShanghai200032China
| | - Chao Liu
- Beijing Advanced Innovation Center for Precision MedicineBeihang UniversityBeijing100083China
| | - Ying Zhang
- Institutes of Biomedical Sciences and Shanghai Cancer CenterFudan UniversityShanghai200032China
- Department of ChemistryFudan UniversityShanghai200433China
- NHC Key Laboratory of Glycoconjugates ResearchFudan UniversityShanghai200032China
| | - Huijie Wang
- Institutes of Biomedical Sciences and Shanghai Cancer CenterFudan UniversityShanghai200032China
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Haojie Lu
- Institutes of Biomedical Sciences and Shanghai Cancer CenterFudan UniversityShanghai200032China
- Department of ChemistryFudan UniversityShanghai200433China
- NHC Key Laboratory of Glycoconjugates ResearchFudan UniversityShanghai200032China
| |
Collapse
|
22
|
Du YJ, Jiang Y, Hou YM, Shi YB. Complement factor I knockdown inhibits colon cancer development by affecting Wnt/β-catenin/c-Myc signaling pathway and glycolysis. World J Gastrointest Oncol 2024; 16:2634-2650. [DOI: 10.4251/wjgo.v16.i6.2634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/24/2024] [Accepted: 03/27/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Colon cancer (CC) occurrence and progression are considerably influenced by the tumor microenvironment. However, the exact underlying regulatory mechanisms remain unclear.
AIM To investigate immune infiltration-related differentially expressed genes (DEGs) in CC and specifically explored the role and potential molecular mechanisms of complement factor I (CFI).
METHODS Immune infiltration-associated DEGs were screened for CC using bioinformatics. Quantitative reverse transcription polymerase chain reaction was used to examine hub DEGs expression in the CC cell lines. Stable CFI-knockdown HT29 and HCT116 cell lines were constructed, and the diverse roles of CFI in vitro were assessed using CCK-8, 5-ethynyl-2’-deoxyuridine, wound healing, and transwell assays. Hematoxylin and eosin staining and immunohistochemistry staining were employed to evaluate the influence of CFI on the tumorigenesis of CC xenograft models constructed using BALB/c male nude mice. Key proteins associated with glycolysis and the Wnt pathway were measured using western blotting.
RESULTS Six key immune infiltration-related DEGs were screened, among which the expression of CFI, complement factor B, lymphoid enhancer binding factor 1, and SRY-related high-mobility-group box 4 was upregulated, whereas that of fatty acid-binding protein 1, and bone morphogenic protein-2 was downregulated. Furthermore, CFI could be used as a diagnostic biomarker for CC. Functionally, CFI silencing inhibited CC cell proliferation, migration, invasion, and tumor growth. Mechanistically, CFI knockdown downregulated the expression of key glycolysis-related proteins (glucose transporter type 1, hexokinase 2, lactate dehydrogenase A, and pyruvate kinase M2) and the Wnt pathway-related proteins (β-catenin and c-Myc). Further investigation indicated that CFI knockdown inhibited glycolysis in CC by blocking the Wnt/β-catenin/c-Myc pathway.
CONCLUSION The findings of the present study demonstrate that CFI plays a crucial role in CC development by influencing glycolysis and the Wnt/β-catenin/c-Myc pathway, indicating that it could serve as a promising target for therapeutic intervention in CC.
Collapse
Affiliation(s)
- Yong-Jun Du
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
| | - Yue Jiang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Yan-Mei Hou
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
| | - Yong-Bo Shi
- Department of Proctology, Zigong Hospital of Traditional Chinese Medicine, Zigong 643000, Sichuan Province, China
| |
Collapse
|
23
|
Du YJ, Jiang Y, Hou YM, Shi YB. Complement factor I knockdown inhibits colon cancer development by affecting Wnt/β-catenin/c-Myc signaling pathway and glycolysis. World J Gastrointest Oncol 2024; 16:2646-2662. [PMID: 38994157 PMCID: PMC11236223 DOI: 10.4251/wjgo.v16.i6.2646] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/24/2024] [Accepted: 03/27/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Colon cancer (CC) occurrence and progression are considerably influenced by the tumor microenvironment. However, the exact underlying regulatory mechanisms remain unclear. AIM To investigate immune infiltration-related differentially expressed genes (DEGs) in CC and specifically explored the role and potential molecular mechanisms of complement factor I (CFI). METHODS Immune infiltration-associated DEGs were screened for CC using bioinformatics. Quantitative reverse transcription polymerase chain reaction was used to examine hub DEGs expression in the CC cell lines. Stable CFI-knockdown HT29 and HCT116 cell lines were constructed, and the diverse roles of CFI in vitro were assessed using CCK-8, 5-ethynyl-2'-deoxyuridine, wound healing, and transwell assays. Hematoxylin and eosin staining and immunohistochemistry staining were employed to evaluate the influence of CFI on the tumorigenesis of CC xenograft models constructed using BALB/c male nude mice. Key proteins associated with glycolysis and the Wnt pathway were measured using western blotting. RESULTS Six key immune infiltration-related DEGs were screened, among which the expression of CFI, complement factor B, lymphoid enhancer binding factor 1, and SRY-related high-mobility-group box 4 was upregulated, whereas that of fatty acid-binding protein 1, and bone morphogenic protein-2 was downregulated. Furthermore, CFI could be used as a diagnostic biomarker for CC. Functionally, CFI silencing inhibited CC cell proliferation, migration, invasion, and tumor growth. Mechanistically, CFI knockdown downregulated the expression of key glycolysis-related proteins (glucose transporter type 1, hexokinase 2, lactate dehydrogenase A, and pyruvate kinase M2) and the Wnt pathway-related proteins (β-catenin and c-Myc). Further investigation indicated that CFI knockdown inhibited glycolysis in CC by blocking the Wnt/β-catenin/c-Myc pathway. CONCLUSION The findings of the present study demonstrate that CFI plays a crucial role in CC development by influencing glycolysis and the Wnt/β-catenin/c-Myc pathway, indicating that it could serve as a promising target for therapeutic intervention in CC.
Collapse
Affiliation(s)
- Yong-Jun Du
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
| | - Yue Jiang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Yan-Mei Hou
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
| | - Yong-Bo Shi
- Department of Proctology, Zigong Hospital of Traditional Chinese Medicine, Zigong 643000, Sichuan Province, China
| |
Collapse
|
24
|
Zhao ZX, Li S, Liu LX. Thymoquinone affects hypoxia-inducible factor-1α expression in pancreatic cancer cells via HSP90 and PI3K/AKT/mTOR pathways. World J Gastroenterol 2024; 30:2793-2816. [PMID: 38899332 PMCID: PMC11185293 DOI: 10.3748/wjg.v30.i21.2793] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 04/14/2024] [Accepted: 05/08/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is associated with some of the worst prognoses of all major cancers. Thymoquinone (TQ) has a long history in traditional medical practice and is known for its anti-cancer, anti-inflammatory, anti-fibrosis and antioxidant pharmacological activities. Recent studies on hypoxia-inducible factor-1α (HIF-1α) and PC have shown that HIF-1α affects the occurrence and development of PC in many aspects. In addition, TQ could inhibit the development of renal cancer by decreasing the expression of HIF-1α. Therefore, we speculate whether TQ affects HIF-1α expression in PC cells and explore the mechanism. AIM To elucidate the effect of TQ in PC cells and the regulatory mechanism of HIF-1α expression. METHODS Cell counting kit-8 assay, Transwell assay and flow cytometry were performed to detect the effects of TQ on the proliferative activity, migration and invasion ability and apoptosis of PANC-1 cells and normal pancreatic duct epithelial (hTERT-HPNE) cells. Quantitative real-time polymerase chain reaction and western blot assay were performed to detect the expression of HIF-1α mRNA and protein in PC cells. The effects of TQ on the HIF-1α protein initial expression pathway and ubiquitination degradation in PANC-1 cells were examined by western blot assay and co-immunoprecipitation. RESULTS TQ significantly inhibited proliferative activity, migration, and invasion ability and promoted apoptosis of PANC-1 cells; however, no significant effects on hTERT-HPNE cells were observed. TQ significantly reduced the mRNA and protein expression levels of HIF-1α in PANC-1, AsPC-1, and BxPC-3 cells. TQ significantly inhibited the expression of the HIF-1α initial expression pathway (PI3K/AKT/mTOR) related proteins, and promoted the ubiquitination degradation of the HIF-1α protein in PANC-1 cells. TQ had no effect on the hydroxylation and von Hippel Lindau protein mediated ubiquitination degradation of the HIF-1α protein but affected the stability of the HIF-1α protein by inhibiting the interaction between HIF-1α and HSP90, thus promoting its ubiquitination degradation. CONCLUSION The regulatory mechanism of TQ on HIF-1α protein expression in PC cells was mainly to promote the ubiquitination degradation of the HIF-1α protein by inhibiting the interaction between HIF-1α and HSP90; Secondly, TQ reduced the initial expression of HIF-1α protein by inhibiting the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Zhan-Xue Zhao
- Department of General Surgery, Qinghai Provincial People's Hospital, Xining 810007, Qinghai Province, China
| | - Shuai Li
- Department of Clinical Pharmacy, The Affiliated Hospital of Qinghai University, Xining 810001, Qinghai Province, China
| | - Lin-Xun Liu
- Department of General Surgery, Qinghai Provincial People's Hospital, Xining 810007, Qinghai Province, China
| |
Collapse
|
25
|
Huang Z, Wang Z, Xu C, Yan Y, Cao X, Zhang F, Shen B. FGL2 as a predictive biomarker for prognosis and immunotherapy in bladder cancer. Int J Med Sci 2024; 21:1447-1460. [PMID: 38903931 PMCID: PMC11186416 DOI: 10.7150/ijms.91874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 05/04/2024] [Indexed: 06/22/2024] Open
Abstract
Background: Metastasis and immunosuppression result in unfavorable prognosis in bladder cancer (BLCA). FGL1 and FGL2 are two members of the fibrinogen-related proteins family, but their potential effects on BLCA remain elusive. Methods: The expression profile of FGL1 and FGL2 in BLCA was analyzed in multiple databases. Furthermore, the expression of FGL2 was validated in BLCA tissues. The predictive capability of FGL2 was evaluated by Kaplan-Meier analysis, univariate analysis, and multivariate Cox regression. A nomogram model was constructed based on FGL2 expression and clinicopathological parameters for clinical practice. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analyses (GSEA) were performed to investigate enrichment in the biological processes. In addition, the correlation between FGL2 and immunological characteristics in the BLCA tumor microenvironment (TME), including tumor-infiltrating immune cells (TICs), cancer-immunity cycles, immune checkpoint molecules (ICPs), immunophenoscores (IPS), and response to anti-PD-L1 immunotherapy was further analyzed. Results: FGL2 was found to be downregulated in BLCA due to hypermethylation of the FGL2 promoter region, which was associated with an unfavorable prognosis. Moreover, BLCA patients with high FGL2 expression exhibited better response to immunotherapy. Conclusions: Our research revealed that FGL2 was downregulated in BLCA and was negatively correlated with DNA methylation. High FGL2 expression was confirmed as an independent risk for prognosis. Moreover, FGL2 is a promising indicator for the response to immunotherapy in patients with BLCA.
Collapse
Affiliation(s)
- Zhengnan Huang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Zeyi Wang
- Department of Urology, Huadong Hospital, Fudan University, Shanghai, China. 200040, China
| | - Chengdang Xu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Yilin Yan
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Xiangqian Cao
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Fang Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Bing Shen
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| |
Collapse
|
26
|
Zhang X, Lan R, Liu Y, Pillarisetty VG, Li D, Zhao CL, Sarkar SA, Liu W, Hanna I, Gupta M, Hajdu C, Melamed J, Shusterman M, Widmer J, Allendorf J, Liu YZ. Enhanced Complement Expression in the Tumor Microenvironment Following Neoadjuvant Therapy: Implications for Immunomodulation and Survival in Pancreatic Ductal Adenocarcinoma. RESEARCH SQUARE 2024:rs.3.rs-4104258. [PMID: 38798691 PMCID: PMC11118688 DOI: 10.21203/rs.3.rs-4104258/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Background Neoadjuvant therapy (NAT) is increasingly being used for pancreatic ductal adenocarcinoma (PDAC) treatment. However, its specific effects on carcinoma cells and the tumor microenvironment (TME) are not fully understood. This study aims to investigate how NAT differentially impacts PDAC's carcinoma cells and TME. Methods Spatial transcriptomics was used to compare gene expression profiles in carcinoma cells and the TME between 23 NAT-treated and 13 NAT-naïve PDAC patients, correlating with their clinicopathologic features. Analysis of an online single-nucleus RNA sequencing (snRNA-seq) dataset was performed for validation of the specific cell types responsible for NAT-induced gene expression alterations. Results NAT not only induces apoptosis and inhibits proliferation in carcinoma cells but also significantly remodels the TME. Notably, NAT induces a coordinated upregulation of multiple key complement genes (C3, C1S, C1R, C4B and C7) in the TME, making the complement pathway one of the most significantly affected pathways by NAT. Patients with higher TME complement expression following NAT exhibit improved overall survival. These patients also exhibit increased immunomodulatory and neurotrophic cancer-associated fibroblasts (CAFs); more CD4+ T cells, monocytes, and mast cells; and reduced immune exhaustion gene expression. snRNA-seq analysis demonstrates C3 complement was specifically upregulated in CAFs but not in other stroma cell types. Conclusions NAT can enhance complement production and signaling within the TME, which is associated with reduced immunosuppression in PDAC. These findings suggest that local complement dynamics could serve as a novel biomarker for prognosis, evaluating treatment response and resistance, and guiding therapeutic strategies in NAT-treated PDAC patients.
Collapse
Affiliation(s)
- Xiaofei Zhang
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Long Island, NY
| | - Ruoxin Lan
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | - Yongjun Liu
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA
| | - Venu G Pillarisetty
- Department of Surgery, University of Washington School of Medicine, Seattle, WA
| | - Danting Li
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | - Chaohui L Zhao
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Long Island, NY
| | - Suparna A. Sarkar
- Department of Pathology and Laboratory Medicine, New York University Grossman School of Medicine, New York, NY
| | - Weiguo Liu
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Long Island, NY
| | - Iman Hanna
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Long Island, NY
| | - Mala Gupta
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Long Island, NY
| | - Cristina Hajdu
- Department of Pathology and Laboratory Medicine, New York University Grossman School of Medicine, New York, NY
| | - Jonathan Melamed
- Department of Pathology and Laboratory Medicine, New York University Grossman Long Island School of Medicine, Long Island, NY
| | - Michael Shusterman
- Department of Oncology, New York University Grossman Long Island School of Medicine, Long Island, NY
| | - Jessica Widmer
- Department of Gastroenterology, New York University Grossman Long Island School of Medicine, Long Island, NY
| | - John Allendorf
- Department of Surgery, New York University Grossman Long Island School of Medicine, Long Island, NY
| | - Yao-Zhong Liu
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| |
Collapse
|
27
|
Zhong Z, Deng W, Wu J, Shang H, Tong Y, He Y, Huang Q, Ba X, Chen Z, Tang K. Cell membrane coated nanoparticles as a biomimetic drug delivery platform for enhancing cancer immunotherapy. NANOSCALE 2024; 16:8708-8738. [PMID: 38634521 DOI: 10.1039/d4nr00284a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Cancer immunotherapy, a burgeoning modality for cancer treatment, operates by activating the autoimmune system to impede the growth of malignant cells. Although numerous immunotherapy strategies have been employed in clinical cancer therapy, the resistance of cancer cells to immunotherapeutic medications and other apprehensions impede the attainment of sustained advantages for most patients. Recent advancements in nanotechnology for drug delivery hold promise in augmenting the efficacy of immunotherapy. However, the efficacy is currently constrained by the inadequate specificity of delivery, low rate of response, and the intricate immunosuppressive tumor microenvironment. In this context, the investigation of cell membrane coated nanoparticles (CMNPs) has revealed their ability to perform targeted delivery, immune evasion, controlled release, and immunomodulation. By combining the advantageous features of natural cell membranes and nanoparticles, CMNPs have demonstrated their unique potential in the realm of cancer immunotherapy. This review aims to emphasize recent research progress and elucidate the underlying mechanisms of CMNPs as an innovative drug delivery platform for enhancing cancer immunotherapy. Additionally, it provides a comprehensive overview of the current immunotherapeutic strategies involving different cell membrane types of CMNPs, with the intention of further exploration and optimization.
Collapse
Affiliation(s)
- Zichen Zhong
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Wen Deng
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Jian Wu
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Haojie Shang
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Yonghua Tong
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Qiu Huang
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| |
Collapse
|
28
|
Santiso A, Heinemann A, Kargl J. Prostaglandin E2 in the Tumor Microenvironment, a Convoluted Affair Mediated by EP Receptors 2 and 4. Pharmacol Rev 2024; 76:388-413. [PMID: 38697857 DOI: 10.1124/pharmrev.123.000901] [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: 04/13/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 05/05/2024] Open
Abstract
The involvement of the prostaglandin E2 (PGE2) system in cancer progression has long been recognized. PGE2 functions as an autocrine and paracrine signaling molecule with pleiotropic effects in the human body. High levels of intratumoral PGE2 and overexpression of the key metabolic enzymes of PGE2 have been observed and suggested to contribute to tumor progression. This has been claimed for different types of solid tumors, including, but not limited to, lung, breast, and colon cancer. PGE2 has direct effects on tumor cells and angiogenesis that are known to promote tumor development. However, one of the main mechanisms behind PGE2 driving cancerogenesis is currently thought to be anchored in suppressed antitumor immunity, thus providing possible therapeutic targets to be used in cancer immunotherapies. EP2 and EP4, two receptors for PGE2, are emerging as being the most relevant for this purpose. This review aims to summarize the known roles of PGE2 in the immune system and its functions within the tumor microenvironment. SIGNIFICANCE STATEMENT: Prostaglandin E2 (PGE2) has long been known to be a signaling molecule in cancer. Its presence in tumors has been repeatedly associated with disease progression. Elucidation of its effects on immunological components of the tumor microenvironment has highlighted the potential of PGE2 receptor antagonists in cancer treatment, particularly in combination with immune checkpoint inhibitor therapeutics. Adjuvant treatment could increase the response rates and the efficacy of immune-based therapies.
Collapse
Affiliation(s)
- Ana Santiso
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Akos Heinemann
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Julia Kargl
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| |
Collapse
|
29
|
Xiao H, Li X, Liang S, Yang S, Han S, Huang J, Shuai X, Ren J. Dual-Responsive Nanomedicine Activates Programmed Antitumor Immunity through Targeting Lymphatic System. ACS NANO 2024; 18:11070-11083. [PMID: 38639726 DOI: 10.1021/acsnano.3c11464] [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: 04/20/2024]
Abstract
Effective antitumor immunotherapy depends on evoking a cascade of cancer-immune cycles with lymph nodes (LNs) as the initial sites for activating antitumor immunity, making drug administration through the lymphatic system highly attractive. Here, we describe a nanomedicine with dual responsiveness to pH and enzyme for a programmed activation of antitumor immune through the lymphatic system. The proposed nanomedicine can release the STING agonist diABZI-C2-NH2 in the LNs' acidic environment to activate dendritic cells (DCs) and T cells. Then, the remaining nanomedicine hitchhikes on the activated T cells (PD-1+ T cells) through binding to PD-1, resulting in an effective delivery into tumor tissues owing to the tumor-homing capacity of PD-1+ T cells. The enzyme matrix metalloproteinase-2 (MMP-2) being enriched in tumor tissue triggers the release of PD-1 antibody (aPD-1) which exerts immune checkpoint blockade (ICB) therapy. Eventually, the nanomedicine delivers a DNA methylation inhibitor GSK-3484862 (GSK) into tumor cells, and then the latter combines with granzyme B (GZMB) to trigger tumor cell pyroptosis. Consequently, the pyroptotic tumor cells induce robust immunogenic cell death (ICD) enhancing the DCs maturation and initiating the cascading antitumor immune response. Study on a 4T1 breast tumor mouse model demonstrates the prominent antitumor therapeutic outcome of this nanomedicine through creating a positive feedback loop of cancer-immunity cycles including immune activation in LNs, T cell-mediated drug delivery, ICB therapy, and tumor cell pyroptosis-featured ICD.
Collapse
Affiliation(s)
- Hong Xiao
- Department of Medical Ultrasonic, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Xiaoxia Li
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Simin Liang
- Department of Medical Ultrasonic, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Shuguang Yang
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Shisong Han
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Jinsheng Huang
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Jie Ren
- Department of Medical Ultrasonic, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
30
|
Sheikhlary S, Lopez DH, Moghimi S, Sun B. Recent Findings on Therapeutic Cancer Vaccines: An Updated Review. Biomolecules 2024; 14:503. [PMID: 38672519 PMCID: PMC11048403 DOI: 10.3390/biom14040503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/06/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Cancer remains one of the global leading causes of death and various vaccines have been developed over the years against it, including cell-based, nucleic acid-based, and viral-based cancer vaccines. Although many vaccines have been effective in in vivo and clinical studies and some have been FDA-approved, there are major limitations to overcome: (1) developing one universal vaccine for a specific cancer is difficult, as tumors with different antigens are different for different individuals, (2) the tumor antigens may be similar to the body's own antigens, and (3) there is the possibility of cancer recurrence. Therefore, developing personalized cancer vaccines with the ability to distinguish between the tumor and the body's antigens is indispensable. This paper provides a comprehensive review of different types of cancer vaccines and highlights important factors necessary for developing efficient cancer vaccines. Moreover, the application of other technologies in cancer therapy is discussed. Finally, several insights and conclusions are presented, such as the possibility of using cold plasma and cancer stem cells in developing future cancer vaccines, to tackle the major limitations in the cancer vaccine developmental process.
Collapse
Affiliation(s)
- Sara Sheikhlary
- Department of Biomedical Engineering, College of Engineering, The University of Arizona, Tucson, AZ 85721, USA
| | - David Humberto Lopez
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| | - Sophia Moghimi
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| | - Bo Sun
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| |
Collapse
|
31
|
Xing A, Lv D, Wu C, Zhou K, Zhao T, Zhao L, Wang H, Feng H. Tertiary Lymphoid Structures Gene Signature Predicts Prognosis and Immune Infiltration Analysis in Head and Neck Squamous Cell Carcinoma. Curr Genomics 2024; 25:88-104. [PMID: 38751598 PMCID: PMC11092909 DOI: 10.2174/0113892029278082240118053857] [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: 10/26/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 05/18/2024] Open
Abstract
Objectives This study aims to assess the prognostic implications of gene signature of the tertiary lymphoid structures (TLSs) in head and neck squamous cell carcinoma (HNSCC) and scrutinize the influence of TLS on immune infiltration. Methods Patients with HNSCC from the Cancer Genome Atlas were categorized into high/low TLS signature groups based on the predetermined TLS signature threshold. The association of the TLS signature with the immune microenvironment, driver gene mutation status, and tumor mutational load was systematically analyzed. Validation was conducted using independent datasets (GSE41613 and GSE102349). Results Patients with a high TLS signature score exhibited better prognosis compared to those with a low TLS signature score. The group with a high TLS signature score had significantly higher immune cell subpopulations compared to the group with a low TLS signature score. Moreover, the major immune cell subpopulations and immune circulation characteristics in the tumor immune microenvironment were positively correlated with the TLS signature. Mutational differences in driver genes were observed between the TLS signature high/low groups, primarily in the cell cycle and NRF2 signaling pathways. Patients with TP53 mutations and high TLS signature scores demonstrated a better prognosis compared to those with TP53 wild-type. In the independent cohort, the relationship between TLS signatures and patient prognosis and immune infiltration was also confirmed. Additionally, immune-related biological processes and signaling pathways were activated with elevated TLS signature. Conclusion High TLS signature is a promising independent prognostic factor for HNSCC patients. Immunological analysis indicated a correlation between TLS and immune cell infiltration in HNSCC. These findings provide a theoretical basis for future applications of TLS signature in HNSCC prognosis and immunotherapy.
Collapse
Affiliation(s)
- Aiyan Xing
- Department of Pathology, Shandong University Qilu Hospital, Jinan, Shandong, 250012, China
| | - Dongxiao Lv
- Cancer Center, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
- Cancer Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Changshun Wu
- Department of Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
- Department of Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Kai Zhou
- Cancer Center, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
- Cancer Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Tianhui Zhao
- Department of Translational Medicine, Genecast Biotechnology Co., Ltd, Wuxi, Jiangsu, 214104, China
| | - Lihua Zhao
- Department of Translational Medicine, Genecast Biotechnology Co., Ltd, Wuxi, Jiangsu, 214104, China
| | - Huaqing Wang
- Department of Medical Oncology, Tianjin Union Medical Center, The Affiliated Hospital of Nankai University, Tianjin, 300000, China
| | - Hong Feng
- Cancer Center, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
- Cancer Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| |
Collapse
|
32
|
Chen R, Wu J, Che Y, Jiao Y, Sun H, Zhao Y, Chen P, Meng L, Zhao T. Machine learning-driven prognostic analysis of cuproptosis and disulfidptosis-related lncRNAs in clear cell renal cell carcinoma: a step towards precision oncology. Eur J Med Res 2024; 29:176. [PMID: 38491523 PMCID: PMC10943875 DOI: 10.1186/s40001-024-01763-1] [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: 02/01/2024] [Accepted: 03/01/2024] [Indexed: 03/18/2024] Open
Abstract
Cuproptosis and disulfidptosis, recently discovered mechanisms of cell death, have demonstrated that differential expression of key genes and long non-coding RNAs (lncRNAs) profoundly influences tumor development and affects their drug sensitivity. Clear cell renal cell carcinoma (ccRCC), the most common subtype of kidney cancer, presently lacks research utilizing cuproptosis and disulfidptosis-related lncRNAs (CDRLRs) as prognostic markers. In this study, we analyzed RNA-seq data, clinical information, and mutation data from The Cancer Genome Atlas (TCGA) on ccRCC and cross-referenced it with known cuproptosis and disulfidptosis-related genes (CDRGs). Using the LASSO machine learning algorithm, we identified four CDRLRs-ACVR2B-AS1, AC095055.1, AL161782.1, and MANEA-DT-that are strongly associated with prognosis and used them to construct a prognostic risk model. To verify the model's reliability and validate these four CDRLRs as significant prognostic factors, we performed dataset grouping validation, followed by RT-qPCR and external database validation for differential expression and prognosis of CDRLRs in ccRCC. Gene function and pathway analysis were conducted using Gene Ontology (GO) and Gene Set Enrichment Analysis (GSEA) for high- and low-risk groups. Additionally, we have analyzed the tumor mutation burden (TMB) and the immune microenvironment (TME), employing the oncoPredict and Immunophenoscore (IPS) algorithms to assess the sensitivity of diverse risk categories to targeted therapeutics and immunosuppressants. Our predominant objective is to refine prognostic predictions for patients with ccRCC and inform treatment decisions by conducting an exhaustive study on cuproptosis and disulfidptosis.
Collapse
Affiliation(s)
- Ronghui Chen
- School of Clinical Medicine, Shandong Second Medical University, Weifang, 261053, China
- Department of Oncology, People's Hospital of Rizhao, Rizhao, 276826, China
| | - Jun Wu
- Department of Oncology, People's Hospital of Rizhao, Rizhao, 276826, China
| | - Yinwei Che
- Department of Central Laboratory, Shandong Provincial Key Medical and Health Laboratory, Rizhao Key Laboratory of Basic Research on Anesthesia and Respiratory Intensive Care, The People's Hospital of Rizhao, Rizhao, 276826, Shandong, China
| | - Yuzhuo Jiao
- Department of Central Laboratory, Shandong Provincial Key Medical and Health Laboratory, Rizhao Key Laboratory of Basic Research on Anesthesia and Respiratory Intensive Care, The People's Hospital of Rizhao, Rizhao, 276826, Shandong, China
| | - Huashan Sun
- Department of Central Laboratory, Shandong Provincial Key Medical and Health Laboratory, Rizhao Key Laboratory of Basic Research on Anesthesia and Respiratory Intensive Care, The People's Hospital of Rizhao, Rizhao, 276826, Shandong, China
| | - Yinuo Zhao
- Department of Pathology, People's Hospital of Rizhao, Rizhao, 276826, China
| | - Pingping Chen
- Department of Pathology, People's Hospital of Rizhao, Rizhao, 276826, China
| | - Lingxin Meng
- Department of Oncology, People's Hospital of Rizhao, Rizhao, 276826, China.
| | - Tao Zhao
- Department of Central Laboratory, Shandong Provincial Key Medical and Health Laboratory, Rizhao Key Laboratory of Basic Research on Anesthesia and Respiratory Intensive Care, The People's Hospital of Rizhao, Rizhao, 276826, Shandong, China
| |
Collapse
|
33
|
Krizova L, Benesova I, Zemanova P, Spacek J, Strizova Z, Humlova Z, Mikulova V, Petruzelka L, Vocka M. Immunophenotyping of peripheral blood in NSCLC patients discriminates responders to immune checkpoint inhibitors. J Cancer Res Clin Oncol 2024; 150:99. [PMID: 38383923 PMCID: PMC10881622 DOI: 10.1007/s00432-024-05628-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024]
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) dramatically changed the prognosis of patients with NSCLC. Unfortunately, a reliable predictive biomarker is still missing. Commonly used biomarkers, such as PD-L1, MSI, or TMB, are not quite accurate in predicting ICI efficacy. METHODS In this prospective observational cohort study, we investigated the predictive role of erythrocytes, thrombocytes, innate and adaptive immune cells, complement proteins (C3, C4), and cytokines from peripheral blood of 224 patients with stage III/IV NSCLC treated with ICI alone (pembrolizumab, nivolumab, and atezolizumab) or in combination (nivolumab + ipilimumab) with chemotherapy. These values were analyzed for associations with the response to the treatment and survival endpoints. RESULTS Higher baseline Tregs, MPV, hemoglobin, and lower monocyte levels were associated with favorable PFS and OS. Moreover, increased baseline basophils and lower levels of C3 predicted significantly improved PFS. The levels of the baseline immature granulocytes, C3, and monocytes were significantly associated with the occurrence of partial regression at the first restaging. Multiple studied parameters (n = 9) were related to PFS benefit at the time of first restaging as compared to baseline values. In addition, PFS nonbenefit group showed a decrease in lymphocyte count after three months of therapy. The OS benefit was associated with higher levels of lymphocytes, erythrocytes, hemoglobin, MCV, and MPV, and a lower value of NLR after three months of treatment. CONCLUSION Our work suggests that parameters from peripheral venous blood may be potential biomarkers in NSCLC patients on ICI. The baseline values of Tregs, C3, monocytes, and MPV are especially recommended for further investigation.
Collapse
Affiliation(s)
- Ludmila Krizova
- Department of Oncology, General University Hospital in Prague and First Faculty of Medicine, Charles University, U Nemocnice 499/2, 128 00, Prague 2, Czech Republic
| | - Iva Benesova
- Department of Immunology, Second Faculty of Medicine, Charles University in Prague and University Hospital in Motol, Prague, Czech Republic
| | - Petra Zemanova
- Department of Oncology, General University Hospital in Prague and First Faculty of Medicine, Charles University, U Nemocnice 499/2, 128 00, Prague 2, Czech Republic
| | - Jan Spacek
- Department of Oncology, General University Hospital in Prague and First Faculty of Medicine, Charles University, U Nemocnice 499/2, 128 00, Prague 2, Czech Republic
| | - Zuzana Strizova
- Department of Immunology, Second Faculty of Medicine, Charles University in Prague and University Hospital in Motol, Prague, Czech Republic
| | - Zuzana Humlova
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Veronika Mikulova
- Institute of Medical Biochemistry and Laboratory Diagnostics, Laboratory of Clinical Immunology and Allergology, General University Hospital in Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Lubos Petruzelka
- Department of Oncology, General University Hospital in Prague and First Faculty of Medicine, Charles University, U Nemocnice 499/2, 128 00, Prague 2, Czech Republic
| | - Michal Vocka
- Department of Oncology, General University Hospital in Prague and First Faculty of Medicine, Charles University, U Nemocnice 499/2, 128 00, Prague 2, Czech Republic.
| |
Collapse
|
34
|
Mastellos DC, Hajishengallis G, Lambris JD. A guide to complement biology, pathology and therapeutic opportunity. Nat Rev Immunol 2024; 24:118-141. [PMID: 37670180 DOI: 10.1038/s41577-023-00926-1] [Citation(s) in RCA: 89] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2023] [Indexed: 09/07/2023]
Abstract
Complement has long been considered a key innate immune effector system that mediates host defence and tissue homeostasis. Yet, growing evidence has illuminated a broader involvement of complement in fundamental biological processes extending far beyond its traditional realm in innate immunity. Complement engages in intricate crosstalk with multiple pattern-recognition and signalling pathways both in the extracellular and intracellular space. Besides modulating host-pathogen interactions, this crosstalk guides early developmental processes and distinct cell trajectories, shaping tissue immunometabolic and regenerative programmes in different physiological systems. This Review provides a guide to the system-wide functions of complement. It highlights illustrative paradigm shifts that have reshaped our understanding of complement pathobiology, drawing examples from evolution, development of the central nervous system, tissue regeneration and cancer immunity. Despite its tight spatiotemporal regulation, complement activation can be derailed, fuelling inflammatory tissue pathology. The pervasive contribution of complement to disease pathophysiology has inspired a resurgence of complement therapeutics with major clinical developments, some of which have challenged long-held dogmas. We thus highlight major therapeutic concepts and milestones in clinical complement intervention.
Collapse
Affiliation(s)
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
35
|
Nicolini A, Rossi G, Ferrari P. Experimental and clinical evidence in favour of an effective immune stimulation in ER-positive, endocrine-dependent metastatic breast cancer. Front Immunol 2024; 14:1225175. [PMID: 38332913 PMCID: PMC10850262 DOI: 10.3389/fimmu.2023.1225175] [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: 05/18/2023] [Accepted: 09/04/2023] [Indexed: 02/10/2024] Open
Abstract
In ER+ breast cancer, usually seen as the low immunogenic type, the main mechanisms favouring the immune response or tumour growth and immune evasion in the tumour microenvironment (TME) have been examined. The principal implications of targeting the oestrogen-mediated pathways were also considered. Recent experimental findings point out that anti-oestrogens contribute to the reversion of the immunosuppressive TME. Moreover, some preliminary clinical data with the hormone-immunotherapy association in a metastatic setting support the notion that the reversion of immune suppression in TME is likely favoured by the G0-G1 state induced by anti-oestrogens. Following immune stimulation, the reverted immune suppression allows the boosting of the effector cells of the innate and adaptive immune response. This suggests that ER+ breast cancer is a molecular subtype where a successful active immune manipulation can be attained. If this is confirmed by a prospective multicentre trial, which is expected in light of the provided evidence, the proposed hormone immunotherapy can also be tested in the adjuvant setting. Furthermore, the different rationale suggests a synergistic activity of our proposed immunotherapy with the currently recommended regimen consisting of antioestrogens combined with cyclin kinase inhibitors. Overall, this lays the foundation for a shift in clinical practice within this most prevalent molecular subtype of breast cancer.
Collapse
Affiliation(s)
- Andrea Nicolini
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Giuseppe Rossi
- Epidemiology and Biostatistics Unit, Institute of Clinical Physiology, National Research Council and Gabriele Monasterio Foundation, Pisa, Italy
| | - Paola Ferrari
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
36
|
Yu B, Geng C, Wu Z, Zhang Z, Zhang A, Yang Z, Huang J, Xiong Y, Yang H, Chen Z. A CIC-related-epigenetic factors-based model associated with prediction, the tumor microenvironment and drug sensitivity in osteosarcoma. Sci Rep 2024; 14:1308. [PMID: 38225273 PMCID: PMC10789798 DOI: 10.1038/s41598-023-49770-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 12/12/2023] [Indexed: 01/17/2024] Open
Abstract
Osteosarcoma is generally considered a cold tumor and is characterized by epigenetic alterations. Although tumor cells are surrounded by many immune cells such as macrophages, T cells may be suppressed, be inactivated, or not be presented due to various mechanisms, which usually results in poor prognosis and insensitivity to immunotherapy. Immunotherapy is considered a promising anti-cancer therapy in osteosarcoma but requires more research, but osteosarcoma does not currently respond well to this therapy. The cancer immunity cycle (CIC) is essential for anti-tumor immunity, and is epigenetically regulated. Therefore, it is possible to modulate the immune microenvironment of osteosarcoma by targeting epigenetic factors. In this study, we explored the correlation between epigenetic modulation and CIC in osteosarcoma through bioinformatic methods. Based on the RNA data from TARGET and GSE21257 cohorts, we identified epigenetic related subtypes by NMF clustering and constructed a clinical prognostic model by the LASSO algorithm. ESTIMATE, Cibersort, and xCell algorithms were applied to analyze the tumor microenvironment. Based on eight epigenetic biomarkers (SFMBT2, SP140, CBX5, HMGN2, SMARCA4, PSIP1, ACTR6, and CHD2), two subtypes were identified, and they are mainly distinguished by immune response and cell cycle regulation. After excluding ACTR6 by LASSO regression, the prognostic model was established and it exhibited good predictive efficacy. The risk score showed a strong correlation with the tumor microenvironment, drug sensitivity and many immune checkpoints. In summary, our study sheds a new light on the CIC-related epigenetic modulation mechanism of osteosarcoma and helps search for potential drugs for osteosarcoma treatment.
Collapse
Affiliation(s)
- Bin Yu
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan Province, China
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Chengkui Geng
- Department of Orthopedics of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan Province, China
| | - Zhongxiong Wu
- Department of Orthopedics of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan Province, China
| | - Zhongzi Zhang
- Department of Orthopedics of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan Province, China
| | - Aili Zhang
- Department of Orthopedics of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan Province, China
| | - Ze Yang
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan Province, China
| | - Jiazheng Huang
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan Province, China
| | - Ying Xiong
- Department of Orthopedics of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan Province, China
| | - Huiqin Yang
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan Province, China.
| | - Zhuoyuan Chen
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan Province, China.
| |
Collapse
|
37
|
Musella M, Manduca N, Maccafeo E, Ruggiero E, Sistigu A. In Vitro Evaluation of Cancer Cell Immunogenicity and Antigen-Specific T-Cell Cytotoxicity by Flow Cytometry. Methods Mol Biol 2024; 2748:13-28. [PMID: 38070104 DOI: 10.1007/978-1-0716-3593-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
A cardinal principle of oncoimmunology is that cancer cells can be eliminated by tumor-infiltrating cytotoxic CD8 T lymphocytes. This has been widely demonstrated during the last 20 years and also recently harnessed for therapy. However, emerging evidence indicates that even neoplasms showing striking initial responses to conventional and targeted (immuno)therapies often acquire resistance, resulting in tumor relapse, increased aggressiveness, and metastatization. Indeed, tumors are complex ecosystems whose malignant and nonmalignant cells, constituting the tumor microenvironment, constantly interact and evolve in space and time. Together with patient's own genetic factors, such environmental interplays may curtail antitumor immune responses leading to cancer immune evasion and natural/acquired (immuno)therapy resistance. In this context, cancer stem cells (CSCs) are thought to be the roots of therapy failure. Flow cytometry is a powerful technology that finds extensive applications in cancer biology. It offers several unique advantages as it allows the rapid, quantitative, and multiparametric analysis of cell populations or functions at the single-cell level. In this chapter, we discuss a two-color flow cytometric protocol to evaluate cancer cell immunogenicity by analyzing the proliferative and tumor-killing potential of ovalbumin (OVA)-specific CD8 OT-1 T cells exposed to OVA-expressing MCA205 sarcoma cells and their CSC counterparts.
Collapse
Affiliation(s)
- Martina Musella
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Nicoletta Manduca
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ester Maccafeo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Eliana Ruggiero
- Experimental Hematology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antonella Sistigu
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy.
| |
Collapse
|
38
|
Liu Z, Yang M, Shu H, Zhou J. A novel prognostic and therapeutic target biomarker based on complement-related gene signature in gastric cancer. Transl Cancer Res 2023; 12:3565-3580. [PMID: 38192986 PMCID: PMC10774048 DOI: 10.21037/tcr-23-628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 10/18/2023] [Indexed: 01/10/2024]
Abstract
Background Gastric cancer (GC) is one of the most prevalent cancer types that reduce human life expectancy. The current tumor-node-metastasis (TNM) staging system is inadequate in identifying higher or lower risk of GC patients because of tumor heterogeneity. Research shows that complement plays a dual role in the tumor development and progression of GC. Methods We downloaded GC data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). A complement-related risk signature was constructed through bioinformatics analysis. Subsequently, the predictive ability of this signature was validated with GSE84437 dataset, and a nomogram integrating risk score and common clinical factors was established. Besides, we evaluated the association of risk score with the immune and stromal cell infiltration in TCGA. Furthermore, immunotherapy response prediction and drug susceptibility analysis were conducted to access the ability of the risk signature in predicting the therapeutic effect. Results A complement-related gene (CRG) signature, based on six genes (SPLG, C9, ITIH1, ZFPM2, CD36, and SERPINE1), was established. In both the training and validation sets, the overall survival of GC patients in the high-risk group was lower than that of the low-risk group, and the nomogram to predict the 1-, 2-, and 3-year survival rates of GC patients was developed. In addition, CIBERSORT algorithm showed the high-risk patients had higher levels of immune cell infiltration than low-risk patients, and the ESTIMATE results implied that the high-risk group had more stromal component in tumor microenvironment. Besides, compared to the low-risk group, there were higher expressions of most immune checkpoint genes and HLA genes in the high-risk group, and the high-risk patients showed higher sensitivity to the chemotherapy and targeted drugs (axitinib, dasatinib, pazopanib, saracatinib, sunitinib and temsirolimus). Conclusions The novel CRG signature may act as a reliable, efficient tool for prognostic prediction and treatment guidance in future clinical practice.
Collapse
Affiliation(s)
- Zuming Liu
- Digestive Department, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, China
| | - Mingwei Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hang Shu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jianmei Zhou
- Digestive Department, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, China
- Department of Infectious Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
39
|
Abizanda-Campo S, Virumbrales-Muñoz M, Humayun M, Marmol I, Beebe DJ, Ochoa I, Oliván S, Ayuso JM. Microphysiological systems for solid tumor immunotherapy: opportunities and challenges. MICROSYSTEMS & NANOENGINEERING 2023; 9:154. [PMID: 38106674 PMCID: PMC10724276 DOI: 10.1038/s41378-023-00616-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/29/2023] [Accepted: 09/20/2023] [Indexed: 12/19/2023]
Abstract
Immunotherapy remains more effective for hematologic tumors than for solid tumors. One of the main challenges to immunotherapy of solid tumors is the immunosuppressive microenvironment these tumors generate, which limits the cytotoxic capabilities of immune effector cells (e.g., cytotoxic T and natural killer cells). This microenvironment is characterized by hypoxia, nutrient starvation, accumulated waste products, and acidic pH. Tumor-hijacked cells, such as fibroblasts, macrophages, and T regulatory cells, also contribute to this inhospitable microenvironment for immune cells by secreting immunosuppressive cytokines that suppress the antitumor immune response and lead to immune evasion. Thus, there is a strong interest in developing new drugs and cell formulations that modulate the tumor microenvironment and reduce tumor cell immune evasion. Microphysiological systems (MPSs) are versatile tools that may accelerate the development and evaluation of these therapies, although specific examples showcasing the potential of MPSs remain rare. Advances in microtechnologies have led to the development of sophisticated microfluidic devices used to recapitulate tumor complexity. The resulting models, also known as microphysiological systems (MPSs), are versatile tools with which to decipher the molecular mechanisms driving immune cell antitumor cytotoxicity, immune cell exhaustion, and immune cell exclusion and to evaluate new targeted immunotherapies. Here, we review existing microphysiological platforms to study immuno-oncological applications and discuss challenges and opportunities in the field.
Collapse
Affiliation(s)
- Sara Abizanda-Campo
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI USA
- University of Wisconsin Carbone Cancer Center, Madison, WI USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI USA
- Tissue Microenvironment Lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, Spain
- Centro Investigación Biomédica en Red. Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, Spain
| | - María Virumbrales-Muñoz
- University of Wisconsin Carbone Cancer Center, Madison, WI USA
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI USA
| | - Mouhita Humayun
- Department of Biological Engineering, Massachusetts Institute of Technology Cambridge, Cambridge, MA USA
| | - Ines Marmol
- Tissue Microenvironment Lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, Spain
| | - David J Beebe
- University of Wisconsin Carbone Cancer Center, Madison, WI USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI USA
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI USA
| | - Ignacio Ochoa
- Tissue Microenvironment Lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, Spain
- Centro Investigación Biomédica en Red. Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, Spain
| | - Sara Oliván
- Tissue Microenvironment Lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, Spain
| | - Jose M Ayuso
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI USA
- University of Wisconsin Carbone Cancer Center, Madison, WI USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI USA
| |
Collapse
|
40
|
Yang X, Ren H, Li Z, Peng X, Fu J. Combinations of radiotherapy with immunotherapy in nasopharyngeal carcinoma. Int Immunopharmacol 2023; 125:111094. [PMID: 37871379 DOI: 10.1016/j.intimp.2023.111094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023]
Abstract
BACKGROUND The treatment of nasopharyngeal carcinoma (NPC) is currently based on concurrent chemoradiotherapy. The prognosis of early NPC is better, while the prognosis of advanced NPC is poor. Immunotherapy is becoming increasingly commonly employed in clinical practice as a new strategy for treating malignant tumors. It has shown promising results in the treatment of certain malignant tumors, making it a current clinical research hotspot. METHODS This review summarizes the current immunotherapy on NPC, highlighting the application of immunotherapy and radiotherapy in the treatment of NPC. RESULTS X-rays can either increase or suppress anti-tumor immune responses through various pathways and mechanisms. Immune checkpoint inhibitors can usually enhance X-ray-induced anti-tumor immune responses. Detecting the immune checkpoint markers and tumor mutation markers, and the functional status of effector cells in patients can aid in the development of individualized treatment that improves the treatment efficacy with reducing drug resistance and adverse reactions. The development of a multivalent vaccine for NPC will help improve the efficacy of the vaccine. Combining techniques that increase the tumor antigens release, such as radiotherapy and oncolytic virus vaccines, may enhance the ability of the immune response. CONCLUSIONS To shed further light on the application of immunotherapy in NPC, large pooled studies must accumulate sufficient cases with detailed exposure data.
Collapse
Affiliation(s)
- Xiaojing Yang
- Department of Radiation Oncology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hanru Ren
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, Shanghai, China
| | - Zhen Li
- Department of Radiation Oncology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue Peng
- Department of Breast Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Fu
- Department of Radiation Oncology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
41
|
James F, Lorger M. Immunotherapy in the context of immune-specialized environment of brain metastases. DISCOVERY IMMUNOLOGY 2023; 2:kyad023. [PMID: 38567052 PMCID: PMC10917168 DOI: 10.1093/discim/kyad023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/18/2023] [Accepted: 11/15/2023] [Indexed: 04/04/2024]
Abstract
Brain metastases (BrM) develop in 20-40% of patients with advanced cancer. They mainly originate from lung cancer, melanoma, breast cancer, and renal cell carcinoma, and are associated with a poor prognosis. While patients with BrM traditionally lack effective treatment options, immunotherapy is increasingly gaining in importance in this group of patients, with clinical trials in the past decade demonstrating the efficacy and safety of immune checkpoint blockade in BrM originating from specific tumor types, foremost melanoma. The brain is an immune-specialized environment with several unique molecular, cellular, and anatomical features that affect immune responses, including those against tumors. In this review we discuss the potential role that some of these unique characteristics may play in the efficacy of immunotherapy, mainly focusing on the lymphatic drainage in the brain and the role of systemic anti-tumor immunity that develops due to the presence of concurrent extracranial disease in addition to BrM.
Collapse
Affiliation(s)
- Fiona James
- School of Medicine, University of Leeds, Leeds, UK
| | | |
Collapse
|
42
|
Zhang L, Guan M, Zhang X, Yu F, Lai F. Machine-learning and combined analysis of single-cell and bulk-RNA sequencing identified a DC gene signature to predict prognosis and immunotherapy response for patients with lung adenocarcinoma. J Cancer Res Clin Oncol 2023; 149:13553-13574. [PMID: 37507593 PMCID: PMC10590321 DOI: 10.1007/s00432-023-05151-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 07/09/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND Innate immune effectors, dendritic cells (DCs), influence cancer prognosis and immunotherapy significantly. As such, dendritic cells are important in killing tumors and influencing tumor microenvironment, whereas their roles in lung adenocarcinoma (LUAD) are largely unknown. METHODS In this study, 1658 LUAD patients from different cohorts were included. In addition, 724 cancer patients who received immunotherapy were also included. To identify DC marker genes in LUAD, we used single-cell RNAsequencing data for analysis and determined 83 genes as DC marker genes. Following that, integrative machine learning procedure was developed to construct a signature for DC marker genes. RESULTS Using TCGA bulk-RNA sequencing data as the training set, we developed a signature consisting of seven genes and classified patients by their risk status. Another six independent cohorts demonstrated the signature' s prognostic power, and multivariate analysis demonstrated it was an independent prognostic factor. LUAD patients in the high-risk group displayed more advanced features, discriminatory immune-cell infiltrations and immunosuppressive states. Cell-cell communication analysis indicates that tumor cells with lower risk scores communicate more actively with the tumor microenvironment. Eight independent immunotherapy cohorts revealed that patients with low-risk had better immunotherapy responses. Drug sensitivity analysis indicated that targeted therapy agents exhibited greater sensitivity to low-risk patients, while chemotherapy agents displayed greater sensitivity to high-risk patients. In vitro experiments confirmed that CTSH is a novel protective factor for LUAD. CONCLUSIONS An unique signature based on DC marker genes that is highly predictive of LUAD patients' prognosis and response to immunotherapy. CTSH is a new biomarker for LUAD.
Collapse
Affiliation(s)
- Liangyu Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Maohao Guan
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xun Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Fengqiang Yu
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| | - Fancai Lai
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
43
|
Hu Z, Jin X, Hong W, Sui Q, Zhao M, Huang Y, Li M, Wang Q, Zhan C, Chen Z. Dissecting the single-cell transcriptome network of macrophage and identifies a signature to predict prognosis in lung adenocarcinoma. Cell Oncol (Dordr) 2023; 46:1351-1368. [PMID: 37079186 PMCID: PMC10116118 DOI: 10.1007/s13402-023-00816-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2023] [Indexed: 04/21/2023] Open
Abstract
PURPOSE The tumor immune microenvironment (TME) plays a vital role in tumorigenesis, progression, and treatment. Macrophages, as an important component of the tumor microenvironment, play an essential role in antitumor immunity and TME remodeling. In this study, we aimed to explore the different functions of different origins macrophages in TME and their value as potential predictive markers of prognosis and treatment. METHODS We performed single-cell analysis using 21 lung adenocarcinoma (LUAD), 12 normal, and four peripheral blood samples from our data and public databases. A prognostic prediction model was then constructed using 502 TCGA patients and explored the potential factors affecting prognosis. The model was validated using data from 4 different GEO datasets with 544 patients after integration. RESULTS According to the source of macrophages, we classified macrophages into alveolar macrophages (AMs) and interstitial macrophages (IMs). AMs mainly infiltrated in normal lung tissue and expressed proliferative, antigen-presenting, scavenger receptors genes, while IMs occupied the majority in TME and expressed anti-inflammatory, lipid metabolism-related genes. Trajectory analysis revealed that AMs rely on self-renew, whereas IMs originated from monocytes in the blood. Cell-to-cell communication showed that AMs interacted mainly with T cells through the MHC I/II signaling pathway, while IMs mostly interacted with tumor-associated fibrocytes and tumor cells. We then constructed a risk model based on macrophage infiltration and showed an excellent predictive power. We further revealed the possible reasons for its potential prognosis prediction by differential genes, immune cell infiltration, and mutational differences. CONCLUSION In conclusion, we investigated the composition, expression differences, and phenotypic changes of macrophages from different origins in lung adenocarcinoma. In addition, we developed a prognostic prediction model based on different macrophage subtype infiltration, which can be used as a valid prognostic biomarker. New insights were provided into the role of macrophages in the prognosis and potential treatment of LUAD patients.
Collapse
Affiliation(s)
- Zhengyang Hu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Xing Jin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Weifeng Hong
- Department of Radiotherapy, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Qihai Sui
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Mengnan Zhao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Yiwei Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Ming Li
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China.
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China.
| | - Zhencong Chen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180, Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
44
|
Wu Z, Yang Y, Zha Y. Radiomics Features on Magnetic Resonance Images Can Predict C5aR1 Expression Levels and Prognosis in High-Grade Glioma. Cancers (Basel) 2023; 15:4661. [PMID: 37760630 PMCID: PMC10527364 DOI: 10.3390/cancers15184661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND The complement component C5a receptor 1 (C5aR1) regulates cancer immunity. This retrospective study aimed to assess its prognostic value in high-grade glioma (HGG) and predict C5aR1 expression using a radiomics approach. METHODS Among 298 patients with HGG, 182 with MRI data were randomly divided into training and test groups for radiomics analysis. We examined the association between C5aR1 expression and prognosis through Kaplan-Meier and Cox regression analyses. We used maximum relevance-minimum redundancy and recursive feature elimination algorithms for radiomics feature selection. We then built a support vector machine (SVM) and a logistic regression model, investigating their performances using receiver operating characteristic, calibration curves, and decision curves. RESULTS C5aR1 expression was elevated in HGG and was an independent prognostic factor (hazard ratio = 3.984, 95% CI: 2.834-5.607). Both models presented with >0.8 area under the curve values in the training and test datasets, indicating efficient discriminatory ability, with SVM performing marginally better. The radiomics score calculated using the SVM model correlated significantly with overall survival (p < 0.01). CONCLUSIONS Our results highlight C5aR1's role in HGG development and prognosis, supporting its potential as a prognostic biomarker. Our radiomics model can noninvasively and effectively predict C5aR1 expression and patient prognosis in HGG.
Collapse
Affiliation(s)
| | | | - Yunfei Zha
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan 430000, China; (Z.W.); (Y.Y.)
| |
Collapse
|
45
|
Phung CD, Nguyen BL, Jeong J, Chang J, Jin SG, Choi H, Ku SK, Kim JO. Shaping the "hot" immunogenic tumor microenvironment by nanoparticles co-delivering oncolytic peptide and TGF-β1 siRNA for boosting checkpoint blockade therapy. Bioeng Transl Med 2023; 8:e10392. [PMID: 37693065 PMCID: PMC10487304 DOI: 10.1002/btm2.10392] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/24/2022] [Accepted: 07/16/2022] [Indexed: 09/12/2023] Open
Abstract
Induction of potent immune responses toward tumors remains challenging in cancer immunotherapy, in which it only showed benefits in a minority of patients with "hot" tumors, which possess pre-existing effector immune cells within the tumor. In this study, we proposed a nanoparticle-based strategy to fire up the "cold" tumor by upregulating the components associated with T and NK cell recruitment and activation and suppressing TGF-β1 secretion by tumor cells. Specifically, LTX-315, a first-in-class oncolytic cationic peptide, and TGF-β1 siRNA were co-entrapped in a polymer-lipid hybrid nanoparticle comprising PLGA, DSPE-mPEG, and DSPE-PEG-conjugated with cRGD peptide (LTX/siR-NPs). The LTX/siR-NPs showed significant inhibition of TGF-β1 expression, induction of type I interferon release, and triggering immunogenic cell death (ICD) in treated tumor cells, indicated via the increased levels of danger molecules, an in vitro setting. The in vivo data showed that the LTX/siR-NPs could effectively protect the LTX-315 peptide from degradation in serum, which highly accumulated in tumor tissue. Consequently, the LTX/siR-NPs robustly suppressed TGF-β1 production by tumor cells and created an immunologically active tumor with high infiltration of antitumor effector immune cells. As a result, the combination of LTX/siR-NP treatment with NKG2A checkpoint inhibitor therapy remarkably increased numbers of CD8+NKG2D+ and NK1.1+NKG2D+ within tumor masses, and importantly, inhibited the tumor growth and prolonged survival rate of treated mice. Taken together, this study suggests the potential of the LTX/siR-NPs for inflaming the "cold" tumor for potentiating the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Cao Dai Phung
- College of PharmacyYeungnam UniversityGyeongsanRepublic of Korea
| | - Bao Loc Nguyen
- College of PharmacyYeungnam UniversityGyeongsanRepublic of Korea
| | - Jee‐Heon Jeong
- Department of Precision Medicine, School of MedicineSungkyunkwan UniversitySuwonRepublic of Korea
| | - Jae‐Hoon Chang
- College of PharmacyYeungnam UniversityGyeongsanRepublic of Korea
| | - Sung Giu Jin
- Department of Pharmaceutical EngineeringDankook UniversityCheonanRepublic of Korea
| | - Han‐Gon Choi
- College of Pharmacy & Institute of Pharmaceutical Science and TechnologyHanyang UniversityAnsanRepublic of Korea
| | - Sae Kwang Ku
- College of Korean MedicineDaegu Haany UniversityGyeongsanRepublic of Korea
| | - Jong Oh Kim
- College of PharmacyYeungnam UniversityGyeongsanRepublic of Korea
| |
Collapse
|
46
|
Yao M, Liu X, Qian Z, Fan D, Sun X, Zhong L, Wu P. Research progress of nanovaccine in anti-tumor immunotherapy. Front Oncol 2023; 13:1211262. [PMID: 37692854 PMCID: PMC10484753 DOI: 10.3389/fonc.2023.1211262] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Tumor vaccines aim to activate dormant or unresponsive tumor-specific T lymphocytes by using tumor-specific or tumor-associated antigens, thus enhancing the body's natural defense against cancer. However, the effectiveness of tumor vaccines is limited by the presence of tumor heterogeneity, low immunogenicity, and immune evasion mechanisms. Fortunately, multifunctional nanoparticles offer a unique chance to address these issues. With the advantages of their small size, high stability, efficient drug delivery, and controlled surface chemistry, nanomaterials can precisely target tumor sites, improve the delivery of tumor antigens and immune adjuvants, reshape the immunosuppressive tumor microenvironment, and enhance the body's anti-tumor immune response, resulting in improved efficacy and reduced side effects. Nanovaccine, a type of vaccine that uses nanotechnology to deliver antigens and adjuvants to immune cells, has emerged as a promising strategy for cancer immunotherapy due to its ability to stimulate immune responses and induce tumor-specific immunity. In this review, we discussed the compositions and types of nanovaccine, and the mechanisms behind their anti-tumor effects based on the latest research. We hope that this will provide a more scientific basis for designing tumor vaccines and enhancing the effectiveness of tumor immunotherapy.
Collapse
Affiliation(s)
- Min Yao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiyu Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
| | - Zhangbo Qian
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
| | - Dianfa Fan
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
| | - Xinjun Sun
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
| | - Liping Zhong
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
| | - Pan Wu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
47
|
Parry PI, Lefringhausen A, Turni C, Neil CJ, Cosford R, Hudson NJ, Gillespie J. 'Spikeopathy': COVID-19 Spike Protein Is Pathogenic, from Both Virus and Vaccine mRNA. Biomedicines 2023; 11:2287. [PMID: 37626783 PMCID: PMC10452662 DOI: 10.3390/biomedicines11082287] [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: 06/17/2023] [Revised: 07/17/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
The COVID-19 pandemic caused much illness, many deaths, and profound disruption to society. The production of 'safe and effective' vaccines was a key public health target. Sadly, unprecedented high rates of adverse events have overshadowed the benefits. This two-part narrative review presents evidence for the widespread harms of novel product COVID-19 mRNA and adenovectorDNA vaccines and is novel in attempting to provide a thorough overview of harms arising from the new technology in vaccines that relied on human cells producing a foreign antigen that has evidence of pathogenicity. This first paper explores peer-reviewed data counter to the 'safe and effective' narrative attached to these new technologies. Spike protein pathogenicity, termed 'spikeopathy', whether from the SARS-CoV-2 virus or produced by vaccine gene codes, akin to a 'synthetic virus', is increasingly understood in terms of molecular biology and pathophysiology. Pharmacokinetic transfection through body tissues distant from the injection site by lipid-nanoparticles or viral-vector carriers means that 'spikeopathy' can affect many organs. The inflammatory properties of the nanoparticles used to ferry mRNA; N1-methylpseudouridine employed to prolong synthetic mRNA function; the widespread biodistribution of the mRNA and DNA codes and translated spike proteins, and autoimmunity via human production of foreign proteins, contribute to harmful effects. This paper reviews autoimmune, cardiovascular, neurological, potential oncological effects, and autopsy evidence for spikeopathy. With many gene-based therapeutic technologies planned, a re-evaluation is necessary and timely.
Collapse
Affiliation(s)
- Peter I. Parry
- Children’s Health Research Clinical Unit, Faculty of Medicine, The University of Queensland, South Brisbane, QLD 4101, Australia
- Department of Psychiatry, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Astrid Lefringhausen
- Children’s Health Defence (Australia Chapter), Huskisson, NSW 2540, Australia; (A.L.); (R.C.); (J.G.)
| | - Conny Turni
- Microbiology Research, QAAFI (Queensland Alliance for Agriculture and Food Innovation), The University of Queensland, St. Lucia, QLD 4072, Australia;
| | - Christopher J. Neil
- Department of Medicine, University of Melbourne, Melbourne, VIC 3010, Australia;
| | - Robyn Cosford
- Children’s Health Defence (Australia Chapter), Huskisson, NSW 2540, Australia; (A.L.); (R.C.); (J.G.)
| | - Nicholas J. Hudson
- School of Agriculture and Food Science, The University of Queensland, Brisbane, QLD 4072, Australia;
| | - Julian Gillespie
- Children’s Health Defence (Australia Chapter), Huskisson, NSW 2540, Australia; (A.L.); (R.C.); (J.G.)
| |
Collapse
|
48
|
Fang D, He Y, Yi Y, Mei J, Liu C. Hub gene associated with prognosis in bladder cancer is a novel therapeutic target. PeerJ 2023; 11:e15670. [PMID: 37601252 PMCID: PMC10439716 DOI: 10.7717/peerj.15670] [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: 03/27/2023] [Accepted: 06/09/2023] [Indexed: 08/22/2023] Open
Abstract
Objective Bladder cancer is a clinical and social conundrum due to its high incidence and recurrence rate. It is urgent to find new targets for the diagnosis and treatment of bladder cancer and improve the prognosis and survival rate of bladder cancer patients. We sought a prognosis-related gene, built related models of evaluated bladder cancer and identified the function of the hub gene in bladder cancer. Methods We downloaded the data of bladder cancer patients from the TCGA database, and used differentially expressed genes (DEGs), copy number variation (CNV) and survival analysis to scan the hub genes associated with prognosis in bladder cancer. Then, multi-factor cox regression was used to obtain the bladder cancer prognosis correlation model. Then, we analyzed the relationship between the expression of hub gene and immune microenvironment of bladder cancer. The relationship between the expression of hub gene and prognosis in bladder cancer patients was verified by immunohistochemistry. Cell proliferation assay and drug sensitivity test in vivo were used to verify the inhibition of bladder cancer by targeted inhibitors. Results In bladder cancer, we screened seven hub genes (ACLY, CNP, NKIRAS2, P3H4, PDIA6, VPS25 and XPO1) associated with survival. Moreover, the multifactor regression model constructed with hub gene can well distinguish the prognosis of bladder cancer. Hub gene is mostly associated with immune microenvironment. Immunohistochemical results basically confirmed the importance of XPO1 in bladder cancer. Selinexor (an inhibitor of XPO1) could effectively inhibit the proliferation of bladder cancer in the cell proliferation experiments by CCK-8 assays and it could suppress the growth of bladder cancer in mouse bladder cancer model. Conclusions In this study, a prognostic model with seven hub genes has provided great help for the prognosis prediction of bladder cancer patients. And XPO1 is an important target affecting the prognosis of bladder cancer, and inhibition of XPO1 can effectively inhibit bladder cancer proliferation and growth.
Collapse
Affiliation(s)
- Dengpan Fang
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Urology, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Yuanqiao He
- Center of Laboratory Animal Science, Nanchang University,, Nanchang, China
- Jiangxi Province Key Laboratory of Laboratory Animal, Nanchang, China
- Nanchang Royo Biotechnology, Nanchang, China
| | - Yun Yi
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiaqi Mei
- The First Clinical Medical College, Nanchang University, Nanchang, China
| | - Cundong Liu
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
49
|
Wu L, Yan J, Bai Y, Chen F, Zou X, Xu J, Huang A, Hou L, Zhong Y, Jing Z, Yu Q, Zhou X, Jiang Z, Wang C, Cheng M, Ji Y, Hou Y, Luo R, Li Q, Wu L, Cheng J, Wang P, Guo D, Huang W, Lei J, Liu S, Yan Y, Chen Y, Liao S, Li Y, Sun H, Yao N, Zhang X, Zhang S, Chen X, Yu Y, Li Y, Liu F, Wang Z, Zhou S, Yang H, Yang S, Xu X, Liu L, Gao Q, Tang Z, Wang X, Wang J, Fan J, Liu S, Yang X, Chen A, Zhou J. An invasive zone in human liver cancer identified by Stereo-seq promotes hepatocyte-tumor cell crosstalk, local immunosuppression and tumor progression. Cell Res 2023; 33:585-603. [PMID: 37337030 PMCID: PMC10397313 DOI: 10.1038/s41422-023-00831-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/22/2023] [Indexed: 06/21/2023] Open
Abstract
Dissecting and understanding the cancer ecosystem, especially that around the tumor margins, which have strong implications for tumor cell infiltration and invasion, are essential for exploring the mechanisms of tumor metastasis and developing effective new treatments. Using a novel tumor border scanning and digitization model enabled by nanoscale resolution-SpaTial Enhanced REsolution Omics-sequencing (Stereo-seq), we identified a 500 µm-wide zone centered around the tumor border in patients with liver cancer, referred to as "the invasive zone". We detected strong immunosuppression, metabolic reprogramming, and severely damaged hepatocytes in this zone. We also identified a subpopulation of damaged hepatocytes with increased expression of serum amyloid A1 and A2 (referred to collectively as SAAs) located close to the border on the paratumor side. Overexpression of CXCL6 in adjacent malignant cells could induce activation of the JAK-STAT3 pathway in nearby hepatocytes, which subsequently caused SAAs' overexpression in these hepatocytes. Furthermore, overexpression and secretion of SAAs by hepatocytes in the invasive zone could lead to the recruitment of macrophages and M2 polarization, further promoting local immunosuppression, potentially resulting in tumor progression. Clinical association analysis in additional five independent cohorts of patients with primary and secondary liver cancer (n = 423) showed that patients with overexpression of SAAs in the invasive zone had a worse prognosis. Further in vivo experiments using mouse liver tumor models in situ confirmed that the knockdown of genes encoding SAAs in hepatocytes decreased macrophage accumulation around the tumor border and delayed tumor growth. The identification and characterization of a novel invasive zone in human cancer patients not only add an important layer of understanding regarding the mechanisms of tumor invasion and metastasis, but may also pave the way for developing novel therapeutic strategies for advanced liver cancer and other solid tumors.
Collapse
Affiliation(s)
- Liang Wu
- Zhongshan-BGI Precision Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China
- BGI-Southwest, BGI-Shenzhen, Chongqing, China
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
| | - Jiayan Yan
- Zhongshan-BGI Precision Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Yinqi Bai
- Zhongshan-BGI Precision Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
- BGI-Hangzhou, Hangzhou, Zhejiang, China
| | - Feiyu Chen
- Zhongshan-BGI Precision Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Xuanxuan Zou
- BGI-Southwest, BGI-Shenzhen, Chongqing, China
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jiangshan Xu
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ao Huang
- Zhongshan-BGI Precision Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Liangzhen Hou
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yu Zhong
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
| | - Zehua Jing
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qichao Yu
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xiaorui Zhou
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Zhifeng Jiang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Chunqing Wang
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Mengnan Cheng
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yuan Ji
- Zhongshan-BGI Precision Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rongkui Luo
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qinqin Li
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Wu
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jianwen Cheng
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Pengxiang Wang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Dezhen Guo
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Waidong Huang
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Junjie Lei
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shang Liu
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
| | - Yizhen Yan
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
| | - Yiling Chen
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
| | - Sha Liao
- BGI-Southwest, BGI-Shenzhen, Chongqing, China
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
| | - Yuxiang Li
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
| | - Haixiang Sun
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Na Yao
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Xiangyu Zhang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Shiyu Zhang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Xi Chen
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
| | - Yang Yu
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Yao Li
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Fengming Liu
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Zheng Wang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Shaolai Zhou
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Huanming Yang
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
| | - Shuang Yang
- Zhongshan-BGI Precision Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xun Xu
- Zhongshan-BGI Precision Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, Shenzhen, Guangdong, China
| | - Longqi Liu
- Zhongshan-BGI Precision Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China
- BGI-Hangzhou, Hangzhou, Zhejiang, China
| | - Qiang Gao
- Zhongshan-BGI Precision Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Zhaoyou Tang
- Zhongshan-BGI Precision Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Xiangdong Wang
- Zhongshan-BGI Precision Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Wang
- Zhongshan-BGI Precision Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China
- James D. Watson Institute of Genome Science, Hangzhou, Zhejiang, China
| | - Jia Fan
- Zhongshan-BGI Precision Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Shiping Liu
- Zhongshan-BGI Precision Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China.
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China.
- Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen, Guangdong, China.
| | - Xinrong Yang
- Zhongshan-BGI Precision Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China.
| | - Ao Chen
- Zhongshan-BGI Precision Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China.
- BGI-Southwest, BGI-Shenzhen, Chongqing, China.
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, Guangdong, China.
- JFL-BGI STOmics Center, Jinfeng Laboratory, Chongqing, China.
| | - Jian Zhou
- Zhongshan-BGI Precision Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China.
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China.
| |
Collapse
|
50
|
Li F, Zhang XQ, Ho W, Tang M, Li Z, Bu L, Xu X. mRNA lipid nanoparticle-mediated pyroptosis sensitizes immunologically cold tumors to checkpoint immunotherapy. Nat Commun 2023; 14:4223. [PMID: 37454146 PMCID: PMC10349854 DOI: 10.1038/s41467-023-39938-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023] Open
Abstract
Synergistically improving T-cell responsiveness is promising for favorable therapeutic outcomes in immunologically cold tumors, yet current treatments often fail to induce a cascade of cancer-immunity cycle for effective antitumor immunity. Gasdermin-mediated pyroptosis is a newly discovered mechanism in cancer immunotherapy; however, cleavage in the N terminus is required to activate pyroptosis. Here, we report a single-agent mRNA nanomedicine-based strategy that utilizes mRNA lipid nanoparticles (LNPs) encoding only the N-terminus of gasdermin to trigger pyroptosis, eliciting robust antitumor immunity. In multiple female mouse models, we show that pyroptosis-triggering mRNA/LNPs turn cold tumors into hot ones and create a positive feedback loop to promote antitumor immunity. Additionally, mRNA/LNP-induced pyroptosis sensitizes tumors to anti-PD-1 immunotherapy, facilitating tumor growth inhibition. Antitumor activity extends beyond the treated lesions and suppresses the growth of distant tumors. We implement a strategy for inducing potent antitumor immunity, enhancing immunotherapy responses in immunologically cold tumors.
Collapse
Affiliation(s)
- Fengqiao Li
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Xue-Qing Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, PR China.
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, PR China.
| | - William Ho
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Maoping Tang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, PR China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, PR China
| | - Zhongyu Li
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Lei Bu
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Xiaoyang Xu
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA.
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA.
| |
Collapse
|