1
|
Brambilla E, Brambilla DJF, Tregnago AC, Riva F, Pasqualotto FF, Soldera J. Exploring macrophage polarization as a prognostic indicator for colorectal cancer: Unveiling the impact of metalloproteinase mutations. World J Clin Cases 2025; 13:105011. [DOI: 10.12998/wjcc.v13.i23.105011] [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: 01/08/2025] [Revised: 03/24/2025] [Accepted: 05/07/2025] [Indexed: 06/04/2025] Open
Abstract
BACKGROUND Macrophages play a crucial role in the tumor microenvironment, displaying remarkable plasticity that allows them to either suppress or promote tumor progression. Their polarization into M1 or M2 phenotypes could have significant prognostic implications, and manipulating this polarization may offer a novel approach to controlling colorectal neoplasms.
AIM To evaluate the infiltration rates of M1 and M2 macrophages in colorectal neoplasia, specifically comparing cases with and without metalloproteinase mutations. Additionally, it sought to explore potential prognostic factors associated with the disease.
METHODS The study involved two cohorts of patients diagnosed with colorectal neoplasia: 33 patients with metalloproteinase mutations and 33 without. Macrophage quantity and polarization were assessed using markers indicative of M1 (iNOS) and M2 (CD163, CD206) macrophages. Prognostic factors and survival outcomes related to colorectal cancer (CRC) were also analyzed.
RESULTS Among the 61 patients, 28 (45.9%) exhibited metalloproteinase mutations, while 33 (54.1%) did not. Tumor staging revealed that 16.9% were in stage I, 34.2% in stage II, 42.4% in stage III, and 8.5% in stage IV. The study recorded 12 patient deaths (19.7%), with 21.2% from the control group and 17.9% from the mutation group. M2 macrophages, identified by CD163 and CD206 markers, had mean counts of 23 and 17, respectively, with standard deviations of 21 and 17. In contrast, M1 macrophages, identified by iNOS, had a mean count of five per site, with a standard deviation of 11.
CONCLUSION The study found no statistically significant differences in macrophage density between groups, irrespective of metalloproteinase mutation status, age, gender, tumor region, staging, TILS, tumor recurrence, or clinical outcomes. No association was observed between macrophage polarization and CRC prognosis or survival. However, patients with metalloproteinase mutations demonstrated a better survival rate, suggesting a potential protective role of this mutation in colorectal neoplasia.
Collapse
Affiliation(s)
- Eduardo Brambilla
- Clinical Gastroenterology, Universidade de Caxias do Sul, Caxias do Sul 95070-560, Rio Grande do Sul, Brazil
| | | | - Aline Caldart Tregnago
- Department of Pathology, Universidade de Caxias do Sul, Caxias do Sul 95070-560, Rio Grande do Sul, Brazil
| | - Floriano Riva
- Department of Pathology, Universidade de Caxias do Sul, Caxias do Sul 95070-560, Rio Grande do Sul, Brazil
| | - Fabio Firmbach Pasqualotto
- Department of Urology, Universidade de Caxias do Sul, Caxias do Sul 95070-560, Rio Grande do Sul, Brazil
| | - Jonathan Soldera
- Department of Gastroenterology and Acute Medicine, University of South Wales, Cardiff CF37 1DL, United Kingdom
| |
Collapse
|
2
|
Wan C, Xu Y, Zhu Y, Cao X, Wang P, Gu Y. NLRP3 inflammasome expression affects immune cell infiltration and clinical prognosis in Helicobacter pylori infection‑associated gastric cancer. Mol Med Rep 2025; 32:185. [PMID: 40314099 PMCID: PMC12059518 DOI: 10.3892/mmr.2025.13550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/26/2025] [Indexed: 05/03/2025] Open
Abstract
High Helicobacter pylori infection rates contribute to high gastric cancer (GC) incidence. While H. pylori infection is associated with GC development its mechanisms are still being studied. The aim of the present study was to examine the differences between H. pylori infection‑induced GC and non‑infected tissues, and to investigate the correlation between nucleotide‑binding oligomerization domain, leucine rich repeat and pyrin domain containing 3 (NLRP3) inflammasome expression and immune cell infiltration in GC, thus providing a theoretical basis for clinical prognosis and immunotherapy. High‑throughput RNA‑sequencing expression data from The Cancer Genome Atlas (TCGA) were analyzed. Additionally, TIMER2.0 and Kaplan‑Meier Plotter were used to analyze the differential expression of NLRP3 mRNA in various tumors, the effect of H. pylori infection on gene expression, and the association between NLRP3 and clinical prognosis among patients with GC. Immunohistochemistry (IHC) was used to assess the effects of NLRP3 protein expression on immune cell infiltration in clinical tissues with or without H. pylori infection. R software was used for data visualization and statistical analysis. TCGA data revealed that the expression levels of NLRP3 in GC tissues were increased compared with those in normal tissues (P<0.05), which was further validated in clinical samples. Furthermore, NLRP3 mRNA expression was significantly elevated in clinical GC tissues infected with H. pylori. Notably higher relative levels of NLRP3 mRNA were observed in tumor tissues with a tumor size ≥5 cm, lymph node metastasis, Tumor‑Node‑Metastasis stage III + IV or poor differentiation compared with the respective controls (P<0.05). IHC confirmed a significant increase in NLRP3 expression within H. pylori‑infected GC tissues compared with that in non‑infected tissues. In GC immune infiltration, NLRP3 expression was revealed to be associated with natural killer cell, whereas it was negatively correlated with regulatory T cells and CD8+ T cells. These findings indicated that NLRP3 may promote the polarization of tumor‑associated macrophages towards the M2 phenotype. High NLRP3 expression also promoted the infiltration of CD3+ and CD206+ cells, which significantly affected the survival rate of patients with GC. The immune infiltration of regulatory T lymphocytes was associated with better survival benefits for patients with GC; however, M2 macrophage infiltration was not conducive to the survival of patients with GC. Furthermore, survival analysis showed that high expression of NLRP3 was associated with a poorer 5‑year overall survival, progression‑free survival and post‑progression survival rates. In conclusion, elevated NLRP3 expression, which may be induced by H. pylori infection, could promote immune cell infiltration potentially by regulating cancer cell proliferation and migration, ultimately leading to an unfavorable prognosis and a notable reduction in the 5‑year survival rate.
Collapse
Affiliation(s)
- Chuandan Wan
- Central Laboratory, Changshu Medical Examination Institute, Changshu, Jiangsu 215500, P.R. China
| | - Yeqiong Xu
- Central Laboratory, Changshu Medical Examination Institute, Changshu, Jiangsu 215500, P.R. China
| | - Yanping Zhu
- Central Laboratory, Changshu Medical Examination Institute, Changshu, Jiangsu 215500, P.R. China
| | - Xuexian Cao
- Department of Oncology and Radiotherapy, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu 215500, P.R. China
| | - Ping Wang
- School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Yulan Gu
- Department of Oncology and Radiotherapy, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu 215500, P.R. China
| |
Collapse
|
3
|
Mauthner R, Marom T, Klein A, Mizrakli Y, Tamir SO. Pediatric acute mastoiditis severity - Can inflammatory biomarkers help us? Int J Pediatr Otorhinolaryngol 2025; 195:112405. [PMID: 40449298 DOI: 10.1016/j.ijporl.2025.112405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/18/2025] [Accepted: 05/26/2025] [Indexed: 06/03/2025]
Affiliation(s)
- Roye Mauthner
- Department of Otolaryngology Head and Neck Surgery, Assuta Ashdod Samson University Hospital, Ashdod, Israel; Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheba, Israel
| | - Tal Marom
- Department of Otolaryngology Head and Neck Surgery, Assuta Ashdod Samson University Hospital, Ashdod, Israel; Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheba, Israel
| | - Ayala Klein
- Department of Otolaryngology Head and Neck Surgery, Assuta Ashdod Samson University Hospital, Ashdod, Israel; Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheba, Israel
| | - Yuval Mizrakli
- Department of Otolaryngology-Head and Neck Surgery, Shamir Medical Center (formerly Assaf Harofeh Medical Center), Zerifin, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Sharon Ovnat Tamir
- Department of Otolaryngology Head and Neck Surgery, Assuta Ashdod Samson University Hospital, Ashdod, Israel; Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheba, Israel.
| |
Collapse
|
4
|
Zhong X, Han J, Li H, Shen X, Yu B, Chen T, Li H, Li J, Pang J, Qian L, Wu W, Tong X, Ding B. Glycosylated protein-related microenvironmental features in breast cancer are associated with patient prognosis. Mamm Genome 2025:10.1007/s00335-025-10137-9. [PMID: 40411577 DOI: 10.1007/s00335-025-10137-9] [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: 03/14/2025] [Accepted: 05/10/2025] [Indexed: 05/26/2025]
Abstract
The tumor microenvironment (TME) and aberrant glycosylation have been suggested to play key roles in cancer. This study integrated differentially expressed genes (DEGs) and weighted gene coexpression network analysis (WGCNA) to identify tumor microenvironment-related genes and construct a TME-risk prognostic signature (TMERS) through LASSO Cox regression. After batch effect removal, 44 TME-prognosis-related genes (TMEPGs) were identified and classified into three molecular subtypes via K-means clustering. The finalized 22-gene TMERS model demonstrated robust prognostic predictive capacity in GEO datasets. The results revealed distinct immune profiles and prognostic stratifications among genetic subtypes and risk groups, confirming that the TMERS is an independent prognostic indicator for breast cancer (BRCA). Glycosyltransferase genes (GTs) have potential therapeutic relevance through immune regulation, with TMEPG member killer cell lectin like receptor B1 (KLRB1) significantly correlated with BRCA prognosis. Cellular experiments demonstrated that KLRB1 overexpression suppressed BRCA cell proliferation and migration. This work establishes a novel prognostic model for BRCA while highlighting KLRB1 as a potential biomarker, providing new insights into TME-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoxiao Zhong
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China.
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China.
| | - Jiaxuan Han
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Huan Li
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Xiangyu Shen
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Bowen Yu
- Department of Gastrointestinal Surgery, Third XiangYa Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Ting Chen
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Haobing Li
- Department of Medical Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421200, Hunan, China
| | - Jun Li
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Jin Pang
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Liyuan Qian
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Wei Wu
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Xiaoliang Tong
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, 410000, China.
| | - Boni Ding
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China.
| |
Collapse
|
5
|
Wu Z, Zhang Z, Gu C. Prognostic and clinicopathological impact of systemic inflammation response index (SIRI) on patients with esophageal cancer: a meta-analysis. Syst Rev 2025; 14:104. [PMID: 40346701 PMCID: PMC12063246 DOI: 10.1186/s13643-025-02847-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/03/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Although the systemic inflammation response index (SIRI) is often associated with prognostic significance in esophageal cancer (EC) patients, the results continue to be conflicting. We focused on identifying SIRI's precise role in forecasting EC prognosis through performing this meta-analysis. METHODS This work searched PubMed, Web of Science, Embase, Cochrane Library, and CNKI till November 16, 2024, and determined pooled hazard ratios (HRs) and 95% confidence intervals (CIs) for evaluating EC prognosis forecasting efficiency of SIRI. The inclusion criteria: (1) pathologic confirmation of EC; (2) those reporting associations of SIRI with EC survival outcomes; (3) those reporting HRs and 95% CIs; (4) those with an available cut-off value of SIRI; and (5) no restriction in language. The exclusion criteria: (1) case reports, reviews, meeting abstracts, comments and letters; (2) those enrolling duplicate cases; and (3) animal studies. RESULTS We enrolled six studies comprising 2176 cases into the present work. Based on our combined findings, elevated SIRI showed significant relation to dismal overall survival (OS) (HR = 1.43, 95%CI = 1.20-1.71, p < 0.001; I2 = 48.8%, p = 0.098) and shortened progression-free survival (PFS) (HR = 2.00, 95%CI = 1.35-2.98, p = 0.001; I2 = 0, p = 0.409) in EC. Moreover, high SIRI exhibited obvious relation to male gender (OR = 1.86, 95%CI = 1.07-3.22, p = 0.027; I2 = 69.4%, p = 0.020), TNM stage of III-IV (OR = 1.52, 95%CI = 1.18-1.94, p = 0.001; I2 = 24.3%, p = 0.265), T3-T4 stage (OR = 1.73, 95%CI = 1.12-2.69, p = 0.014; I2 = 61.0%, p = 0.053), and lymph node metastasis (OR = 1.29, 95%CI = 1.02-1.64, p = 0.036; I2 = 42.7%, p = 0.155). However, SIRI was not markedly related to age, tumor location, tumor differentiation, or smoking history. CONCLUSION In summary, high SIRI is significantly related to dismal OS and shortened PFS of EC cases, together with advanced tumor stage, T3-T4 stage, and lymph node metastasis of EC. Due to some limitations, large prospective studies that utilize standardized threshold SIRI should be conducted to validate our results in the future.
Collapse
Affiliation(s)
- Zhong Wu
- Clinical Laboratory, Nanxun District Hospital of Traditional Chinese Medicine, Huzhou, Zhejiang, 313009, China
| | - Zongxin Zhang
- Clinical Laboratory, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, 313000, China
| | - Chao Gu
- Operating Room, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, 313000, Zhejiang, China.
| |
Collapse
|
6
|
Wang J, Tao X, Zhu J, Dai Z, Du Y, Xie Y, Chu X, Fu G, Lei Z. Tumor organoid-immune co-culture models: exploring a new perspective of tumor immunity. Cell Death Discov 2025; 11:195. [PMID: 40268893 PMCID: PMC12019369 DOI: 10.1038/s41420-025-02407-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 04/25/2025] Open
Abstract
Recent advancements in technology have significantly expanded the scope of tumor research, progressing from the study of individual cells to more intricate tissue and organ-level analyses. Tumor organoids have emerged as a highly realistic platform for investigating tumor growth, development, and their interactions with the surrounding microenvironment. However, a notable limitation of these organoids is their lack of the diverse cellular composition typically observed in actual tumors, which hinders their ability to fully replicate the complexity of the tumor microenvironment. Immune cells play a pivotal role, and tumor immunology has become a major research hotspot. Research in tumor immunology aims to elucidate how the immune system recognizes and attacks tumor cells, as well as how tumor cells evade immune surveillance. In recent years, there has been growing interest in co-culturing immune cells with tumor organoids, an approach that has yielded valuable insights into the intricate interactions between tumors and the immune system. The aim of this paper is to review and discuss the progress achieved in co-culturing tumor organoids with immune cells. By doing so, we hope to offer a new perspective and enhance our understanding of the complexity and diversity inherent in the tumor microenvironment.
Collapse
Affiliation(s)
- Jing Wang
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Xiaoyue Tao
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jialong Zhu
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhe Dai
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuanyang Du
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yiyang Xie
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoyuan Chu
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China.
| | - Gongbo Fu
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China.
| | - Zengjie Lei
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China.
| |
Collapse
|
7
|
Soleimani Mamalo A, Pashaei M, Valilo M, Ojarudi M. Cytokine-mediated regulation of immune cell metabolic pathways in the tumor microenvironment. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04133-8. [PMID: 40220026 DOI: 10.1007/s00210-025-04133-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025]
Abstract
Cancer, an important global health problem, is defined by aberrant cell proliferation and continues to be the main cause of death globally. The tumor microenvironment (TME) plays an essential role in the development of cancer, resistance to therapy, and regulation of the immune response. Some immune cells in the TME, like T cells, B cells, macrophages, dendritic cells, and natural killer cells, can either stop or help tumor growth, depending on how metabolic and cytokine changes happen. Cytokines function as essential signaling molecules that modulate immune cell metabolism, altering their functionality. This review focuses on how cytokine-mediated metabolic reprogramming affects the activity of immune cells inside the TME, which can either make the immune response stronger or weaker. New ways of treating cancer that focus on metabolic pathways and cytokine signaling, such as using IL (Interleukin) - 15, IL- 10, and IL- 4, show promise in boosting immune cell activity and making cancer treatments more effective. Finding these pathways could lead to new ways to treat cancer with immunotherapy that focus on metabolic competition and immune resistance in the TME.
Collapse
Affiliation(s)
| | - Mohammadreza Pashaei
- Department of Internal Medicine, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Valilo
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Masoud Ojarudi
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
8
|
Chen X, Chen B, Zhao H. Role of Neutrophils in Anti-Tumor Activity: Characteristics and Mechanisms of Action. Cancers (Basel) 2025; 17:1298. [PMID: 40282474 PMCID: PMC12025517 DOI: 10.3390/cancers17081298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
As one of the leading components in the immune system, neutrophils in the tumor microenvironment (TME) have received considerable attention in recent years. The tumor-killing effects of neutrophils in a variety of tumors have been reported. However, the functions of neutrophils in tumors remain to be completely elucidated, and both anti-tumor and tumor-promotion activities have been reported. This review focuses on the characteristics of neutrophils and their mechanisms of action in the TME, with an emphasis on their anti-tumor activity, including reactive oxygen species (ROS)-induced tumor killing, cytotoxic T lymphocytes (CTLs)-induced tumor killing, trogocytosis, cytotoxic enzymes, and trained immunity. Furthermore, the possible targets and methods of tumor treatment regimens for neutrophils are explored, with the aim of exploring the use of neutrophils in the future as a potential anti-tumor treatment strategy.
Collapse
Affiliation(s)
- Xin Chen
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi’an 710032, China;
| | - Bingdi Chen
- The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai 200092, China
| | - Huadong Zhao
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi’an 710032, China;
| |
Collapse
|
9
|
Wahnou H, El Kebbaj R, Hba S, Ouadghiri Z, El Faqer O, Pinon A, Liagre B, Limami Y, Duval RE. Neutrophils and Neutrophil-Based Drug Delivery Systems in Anti-Cancer Therapy. Cancers (Basel) 2025; 17:1232. [PMID: 40227814 PMCID: PMC11988188 DOI: 10.3390/cancers17071232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/15/2025] Open
Abstract
Neutrophils, the most abundant white blood cells, play a dual role in cancer progression. While they can promote tumor growth, metastasis, and immune suppression, they also exhibit anti-tumorigenic properties by attacking cancer cells and enhancing immune responses. This review explores the complex interplay between neutrophils and the tumor microenvironment (TME), highlighting their ability to switch between pro- and anti-tumor phenotypes based on external stimuli. Pro-tumorigenic neutrophils facilitate tumor growth through mechanisms such as neutrophil extracellular traps (NETs), secretion of pro-inflammatory cytokines, and immune evasion strategies. They contribute to angiogenesis, tumor invasion, and metastasis by releasing vascular endothelial growth factor (VEGF) and matrix metalloproteinases (MMPs). Conversely, anti-tumor neutrophils enhance cytotoxicity by generating reactive oxygen species (ROS), promoting antibody-dependent cell-mediated cytotoxicity (ADCC), and activating other immune cells such as cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells. Recent advances in neutrophil-based drug delivery systems have harnessed their tumor-homing capabilities to improve targeted therapy. Neutrophil-mimicking nanoparticles and membrane-coated drug carriers offer enhanced drug accumulation in tumors, reduced systemic toxicity, and improved therapeutic outcomes. Additionally, strategies to modulate neutrophil activity, such as inhibiting their immunosuppressive functions or reprogramming them towards an anti-tumor phenotype, are emerging as promising approaches in cancer immunotherapy. Understanding neutrophil plasticity and their interactions with the TME provides new avenues for therapeutic interventions. Targeting neutrophil-mediated mechanisms could enhance existing cancer treatments and lead to the development of novel immunotherapies, ultimately improving patient survival and clinical outcomes.
Collapse
Affiliation(s)
- Hicham Wahnou
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
| | - Riad El Kebbaj
- Sciences and Engineering of Biomedicals, Biophysics and Health Laboratory, Higher Institute of Health Sciences, Hassan First University, Settat 26000, Morocco;
| | - Soufyane Hba
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (A.P.); (B.L.)
| | - Zaynab Ouadghiri
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
| | - Othman El Faqer
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
| | - Aline Pinon
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (A.P.); (B.L.)
| | - Bertrand Liagre
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (A.P.); (B.L.)
| | - Youness Limami
- Sciences and Engineering of Biomedicals, Biophysics and Health Laboratory, Higher Institute of Health Sciences, Hassan First University, Settat 26000, Morocco;
| | | |
Collapse
|
10
|
Cui X, Cao C, Hao W, Pan X, Cao Y, Fu Y, Hao H, Jiao Y, Lin S, Cui S, Li R, Liu Y, Yan F. A Nanoplatform of Reversing Tumor Immunosuppressive Microenvironment Based on the NIR-II Gold Hollow Nanorod for the Treatment of Hepatocellular Carcinoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2500144. [PMID: 40130748 DOI: 10.1002/smll.202500144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 02/16/2025] [Indexed: 03/26/2025]
Abstract
Advanced hepatocellular carcinoma (HCC) presents a strongly immunosuppressive tumor microenvironment, which enables tumor cells to evade immune cell attacks and hinder effective drug killing, thereby hindering the achievement of the desired therapeutic effect. In response, a novel nanoplatform- AuHNR@γ-Fe2O3@Lenvatinib@β-Glucan (AFLG) with surface modified β-1,3-glucan is developed, which exhibits potent immunostimulatory effect and the capability of repolarizing macrophages, to counteract the immunosuppressive conditions present in the tumor microenvironment. Leveraging the hollow structure of gold nanorods, Lenvatinib is efficiently loaded, a first-line targeted drug for HCC, which effectively inhibits tumor angiogenesis. Additionally, through atomic layer deposition, γ-Fe2O3 is generated on the hollow gold nanorod surface, endowing it with chemodynamic therapy and magnetic resonance T2-weighted imaging capabilities while excellently maintaining the gold nanorod's superior photothermal therapy and photoacoustic imaging properties under 1064 nm excitation. These AFLG NPs feature dual-modal imaging and quadruple-modal synergistic therapy capabilities, along with their powerful potential in remodeling the immunosuppressive tumor microenvironment, offering an encouraging novel approach for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xinyuan Cui
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin 2nd Rd, Shanghai, 200025, P. R. China
| | - Cheng Cao
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, No. 800 Dongchuan Rd, Shanghai, 200240, P. R. China
| | - Wanting Hao
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin 2nd Rd, Shanghai, 200025, P. R. China
| | - Xinni Pan
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Rd, Shanghai, 200233, P. R. China
| | - Yu Cao
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, No. 800 Dongchuan Rd, Shanghai, 200240, P. R. China
| | - Yanfei Fu
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, No. 800 Dongchuan Rd, Shanghai, 200240, P. R. China
| | - Huifang Hao
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, No. 800 Dongchuan Rd, Shanghai, 200240, P. R. China
| | - Yingao Jiao
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, No. 800 Dongchuan Rd, Shanghai, 200240, P. R. China
| | - Shujing Lin
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, No. 800 Dongchuan Rd, Shanghai, 200240, P. R. China
| | - Shengsheng Cui
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, No. 800 Dongchuan Rd, Shanghai, 200240, P. R. China
| | - Ruokun Li
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin 2nd Rd, Shanghai, 200025, P. R. China
- Faculty of Medical Imaging Technology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, No. 150 Ruijin 2nd Rd, Shanghai, 200025, P. R. China
| | - Yanlei Liu
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, No. 800 Dongchuan Rd, Shanghai, 200240, P. R. China
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin 2nd Rd, Shanghai, 200025, P. R. China
- Faculty of Medical Imaging Technology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, No. 150 Ruijin 2nd Rd, Shanghai, 200025, P. R. China
| |
Collapse
|
11
|
Hu MM, Zhao Y, Zhang N, Gong FY, Zhang W, Dong CS, Dai JF, Wang J. Tumor Microenvironment: Obstacles and Opportunities for T Cell-Based Tumor Immunotherapies. Mol Cancer Res 2025; 23:277-287. [PMID: 39898773 DOI: 10.1158/1541-7786.mcr-24-0747] [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: 08/09/2024] [Revised: 11/20/2024] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
The complex composition and dynamic change of the tumor microenvironment (TME), mainly consisting of tumor cells, immune cells, stromal cells, and extracellular components, significantly impede the effector function of cytotoxic T lymphocytes (CTL), thus representing a major obstacle for tumor immunotherapies. In this review, we summarize and discuss the impacts and underlying mechanisms of major elements in the TME (different cell types, extracellular matrix, nutrients and metabolites, etc.) on the infiltration, survival, and effector functions of T cells, mainly CD8+ CTLs. Moreover, we also highlight recent advances that may potentiate endogenous antitumor immunity and improve the efficacy of T cell-based immunotherapies in patients with cancer by manipulating components inside/outside of the TME. A deeper understanding of the effects and action mechanisms of TME components on the tumor-eradicating ability of CTLs may pave the way for discovering new targets to augment endogenous antitumor immunity and for designing combinational therapeutic regimens to enhance the efficacy of tumor immunotherapies in the clinic.
Collapse
Affiliation(s)
- Miao-Miao Hu
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Ying Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Nan Zhang
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Fang-Yuan Gong
- Department of Immunology, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Wei Zhang
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Chun-Sheng Dong
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jian-Feng Dai
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jun Wang
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| |
Collapse
|
12
|
Akkız H, Şimşek H, Balcı D, Ülger Y, Onan E, Akçaer N, Delik A. Inflammation and cancer: molecular mechanisms and clinical consequences. Front Oncol 2025; 15:1564572. [PMID: 40165901 PMCID: PMC11955699 DOI: 10.3389/fonc.2025.1564572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Inflammation, a hallmark of cancer, has been associated with tumor progression, transition into malignant phenotype and efficacy of anticancer treatments in cancer. It affects all stages of cancer, from the initiation of carcinogenesis to metastasis. Chronic inflammation induces immunosup-pression, providing an environment conducive to carcinogenesis, whereas acute inflammation induces an antitumor immune response, leading to tumor suppression. Solid tumors have an inflammatory tumor microenvironment (TME) containing cancer cells, immune cells, stromal cells, and soluble molecules, which plays a key role in tumor progression and therapy response. Both cancer cells and stromal cells in the TME are highly plastic and constantly change their phenotypic and functional properties. Cancer-associated inflammation, the majority of which consists of innate immune cells, plays an important role in cancer cell plasticity, cancer progression and the development of anticancer drug resistance. Today, with the combined used of advanced technologies, such as single-cell RNA sequencing and spatial molecular imaging analysis, the pathways linking chronic inflammation to cancer have been largely elucidated. In this review article, we highlighted the molecular and cellular mechanisms involved in cancer-associated inflammation and its effects on cancer progression and treatment response. We also comprehensively review the mechanisms linking chronic inflammation to cancer in the setting of GI cancers.
Collapse
Affiliation(s)
- Hikmet Akkız
- Department of Gastroenterology, Medical Faculty, Bahçeşehir University, İstanbul, Türkiye
| | - Halis Şimşek
- Department of Gastroenterology, Medical Faculty, Hacettepe University, Ankara, Türkiye
| | - Deniz Balcı
- Department of Gastroenterology, Medical Faculty, Bahçeşehir University, İstanbul, Türkiye
| | - Yakup Ülger
- Department of Gastroenterology, Medical Faculty, Cukurova University, Adana, Türkiye
| | - Engin Onan
- Department of Nephrology, Medical Faculty, Baskent University, Adana, Türkiye
| | - Nevin Akçaer
- Department of Gastroenterology, Medical Faculty, Health Sciences University, Adana, Türkiye
| | - Anıl Delik
- Department of Gastroenterology, Medical Faculty, Cukurova University, Adana, Türkiye
- Department of Biology, Science and Literature Faculty, Cukurova University, Adana, Türkiye
| |
Collapse
|
13
|
Faur IF, Dobrescu A, Clim IA, Pasca P, Burta C, Tarta C, Brebu D, Neamtu AA, Braicu V, Duta C, Totolici B. Prognostic Significance of Peripheral Blood Parameters as Predictor of Neoadjuvant Chemotherapy Response in Breast Cancer. Int J Mol Sci 2025; 26:2541. [PMID: 40141182 PMCID: PMC11942583 DOI: 10.3390/ijms26062541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/28/2025] Open
Abstract
The standard treatment for breast cancer typically includes surgery, often followed by systemic therapy and individualized treatment regimens. However, there is growing interest in identifying pre-therapeutic biomarkers that can predict tumor response to neoadjuvant chemotherapy (NACT). This study systematically evaluated various analytical parameters, including age, TNM stage, histological type, molecular subtype, and several biomarker ratios, such as the platelet-to-lymphocyte ratio (PLR), neutrophil-to-lymphocyte ratio (NLR), lymphocyte-to-monocyte ratio (LMR), systemic immune-inflammatory index (SII), and prognostic nutritional index (PNI). We aimed to assess the predictive value of these parameters regarding the tumor's response rate to NACT. The analysis revealed a statistically significant association between the pathological complete response-pCR (absence of any detectable cancer cells in the tissue following neoadjuvant chemotherapy (NACT))-rate and NLR in the subgroup with values between 1 and 3 (p = 0.001). The optimal cut-off for PLR was determined to be 120.45, with 80.55% of patients achieving pCR showing PLR values below this threshold (p = 0.000). Similarly, the LMR cut-off was found to be 12.34, with 77.77% of patients with pCR having LMR values below this threshold (p = 0.002). Additionally, lower pre-therapeutic values of NLR (p < 0.001), PLR (p = 0.002), SII (p = 0.001), and LMR (p = 0.001) were significantly correlated with pCR compared to the non-pCR subgroup (p < 0.005). These findings highlight the predictive potential of these biomarkers for achieving pCR following NACT. Our study supports the hypothesis that pre-therapeutic values of NLR, PLR, SII, and LMR can serve as predictive biomarkers for pCR in breast cancer patients undergoing NACT. However, the PNI did not demonstrate predictive potential in relation to pCR. These biomarkers may provide valuable insights into patient prognosis and guide personalized treatment strategies.
Collapse
Affiliation(s)
- Ionut Flaviu Faur
- IInd Surgery Clinic, Timisoara Emergency County Hospital, 300723 Timisoara, Romania; (I.F.F.); (A.D.); (P.P.); (C.T.); (D.B.); (V.B.); (C.D.)
- X Department of General Surgery, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Multidisciplinary Doctoral School “Vasile Goldiș”, Western University of Arad, 310025 Arad, Romania
| | - Amadeus Dobrescu
- IInd Surgery Clinic, Timisoara Emergency County Hospital, 300723 Timisoara, Romania; (I.F.F.); (A.D.); (P.P.); (C.T.); (D.B.); (V.B.); (C.D.)
- X Department of General Surgery, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Ioana Adelina Clim
- Doctoral School of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Paul Pasca
- IInd Surgery Clinic, Timisoara Emergency County Hospital, 300723 Timisoara, Romania; (I.F.F.); (A.D.); (P.P.); (C.T.); (D.B.); (V.B.); (C.D.)
- X Department of General Surgery, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Cosmin Burta
- Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania;
| | - Cristi Tarta
- IInd Surgery Clinic, Timisoara Emergency County Hospital, 300723 Timisoara, Romania; (I.F.F.); (A.D.); (P.P.); (C.T.); (D.B.); (V.B.); (C.D.)
- X Department of General Surgery, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Dan Brebu
- IInd Surgery Clinic, Timisoara Emergency County Hospital, 300723 Timisoara, Romania; (I.F.F.); (A.D.); (P.P.); (C.T.); (D.B.); (V.B.); (C.D.)
- X Department of General Surgery, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Andreea-Adriana Neamtu
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania;
| | - Vlad Braicu
- IInd Surgery Clinic, Timisoara Emergency County Hospital, 300723 Timisoara, Romania; (I.F.F.); (A.D.); (P.P.); (C.T.); (D.B.); (V.B.); (C.D.)
- X Department of General Surgery, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Ciprian Duta
- IInd Surgery Clinic, Timisoara Emergency County Hospital, 300723 Timisoara, Romania; (I.F.F.); (A.D.); (P.P.); (C.T.); (D.B.); (V.B.); (C.D.)
- X Department of General Surgery, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Bogdan Totolici
- Ist Clinic of General Surgery, Arad County Emergency Clinical Hospital, 310158 Arad, Romania;
- Department of General Surgery, Faculty of Medicine, “Vasile Goldiș” Western University of Arad, 310025 Arad, Romania
| |
Collapse
|
14
|
Turlej E, Domaradzka A, Radzka J, Drulis-Fajdasz D, Kulbacka J, Gizak A. Cross-Talk Between Cancer and Its Cellular Environment-A Role in Cancer Progression. Cells 2025; 14:403. [PMID: 40136652 PMCID: PMC11940884 DOI: 10.3390/cells14060403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
The tumor microenvironment is a dynamic and complex three-dimensional network comprising the extracellular matrix and diverse non-cancerous cells, including fibroblasts, adipocytes, endothelial cells and various immune cells (lymphocytes T and B, NK cells, dendritic cells, monocytes/macrophages, myeloid-derived suppressor cells, and innate lymphoid cells). A constantly and rapidly growing number of studies highlight the critical role of these cells in shaping cancer survival, metastatic potential and therapy resistance. This review provides a synthesis of current knowledge on the modulating role of the cellular microenvironment in cancer progression and response to treatment.
Collapse
Affiliation(s)
- Eliza Turlej
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Aleksandra Domaradzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Justyna Radzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Dominika Drulis-Fajdasz
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Julita Kulbacka
- Departament of Molecular and Cellular Biology, Faculty of Pharmacy, Wrocław Medical University, Borowska 211A, 50-556 Wrocław, Poland;
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Agnieszka Gizak
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| |
Collapse
|
15
|
Wang Y, Tian W, Li R, Zhou D, Ding K, Feng S, Ge Y, Luo Y, Chen Z, Hou H. Platelet FcRγ inhibits tumor metastasis by preventing the colonization of circulating tumor cells. Eur J Pharmacol 2025; 990:177286. [PMID: 39848529 DOI: 10.1016/j.ejphar.2025.177286] [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: 05/24/2024] [Revised: 12/18/2024] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
Fc receptor γ subunit (FcRγ) activation plays a crucial role in cancer carcinogenesis. Here, we aimed to uncover the impact of FcRγ on circulating tumor cells (CTC) colonization and the underlying mechanism. FcRγ deficient (FcRγ-/-) mice were used to investigate the functional effects of FcRγ in cancer metastasis, and the results demonstrated that FcRγ deficiency significantly promotes metastasis. The tumor metastasis effect, antiplatelet, platelet or neutrophil infusion experiments were conducted with FcRγ deficient (FcRγ-/-) mice and wild type mice (WT), bearing B16F10 or LCC tumor cells. Blood routine test, flow cytometry, immunofluorescent staining and in vivo image were applied for analysis. Platelet adhesion and neutrophil chemotaxis were analyzed by flow cytometry and ELISA in vitro. Platelet adoptive model was used for mimicing early colonization stage. Our results indicated FcRγ deficiency significantly promoted tumor metastasis accompanied with increased number of platelet and neutrophil in the lung. Further investigation showed that FcRγ-/- platelet infusion, rather than FcRγ-/- neutrophils, promoted CTC colonization. While platelet inhibitor Aspirin abrogated the platelet-mediated infiltration of neutrophil in the lung. Mechanistically, platelet FcRγ deficiency facilitated the adhesion of platelets and cancer cells and increased secretion of CXCL5 and CXCL7 which triggered the platelet-induced neutrophil recruitment. In sum, our study indicates that FcRγ is a restrainer in controlling cancer metastasis through regulating the adhesion of platelets and cancer cells and recruiting more neutrophils, which provides a potential target for anti-metastatic therapies. The level of FcRγ expression in platelets could act as a novel biomarker for cancer metastasis.
Collapse
Affiliation(s)
- Yun Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China
| | - Wei Tian
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China; School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China
| | - Rui Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China
| | - Dewang Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China
| | - Kaiqiang Ding
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China
| | - Shuang Feng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China
| | - Yao Ge
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China
| | - Yan Luo
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China
| | - Zhen Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China
| | - Hui Hou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China.
| |
Collapse
|
16
|
Shi B, Du M, Chen Z. Advances in tumor immunotherapy targeting macrophages. Expert Rev Clin Immunol 2025; 21:259-276. [PMID: 39636579 DOI: 10.1080/1744666x.2024.2438721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 11/03/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION In recent years, immunotherapy has shown significant therapeutic potential in patients with advanced tumors. However, only a small number of individuals benefit, mainly due to the tumor microenvironment (TME), which provides conditions for the development of tumors. Macrophages in TME, known as tumor-associated macrophages (TAM), are mainly divided into M1 anti-tumor and M2 pro-tumor phenotypes, which play a regulatory role in various stages of tumorigenesis, promote tumorigenesis and metastasis, and cause immunotherapy resistance. AREAS COVERED This review focuses on research strategies and preclinical/clinical research progress in translating TAM into antitumor phenotype by referring to the PubMed database for five years. These include small molecule chemotherapy drug development, metabolic regulation, gene editing, physical stimulation, nanotechnology-mediated combination therapy strategies, and chimeric antigen receptor-based immunotherapy. EXPERT OPINION It is necessary to explore the surface-specific receptors and cell signaling pathways of TAM further to improve the specificity and targeting of drugs and to strengthen research in the field of probes that can monitor changes in TAM in real time. In addition, the physical stimulation polarization strategy has the advantages of being noninvasive, economical, and stable and will have excellent clinical transformation value in the future.
Collapse
Affiliation(s)
- Binrui Shi
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Medical imaging, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
- The Seventh Affiliated Hospital, Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Meng Du
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Seventh Affiliated Hospital, Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Zhiyi Chen
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
17
|
Zhu H, Wang F, Lu X, Wu H, Shi C, Matsas S, Peixoto RD, Aruquipa MPS, Tang C, Feng S. Low expression of PRTN3 regulates the progression of gastric cancer by inhibition of cell cycle and promotion of apoptosis. Transl Cancer Res 2025; 14:1415-1427. [PMID: 40104702 PMCID: PMC11912076 DOI: 10.21037/tcr-2025-153] [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: 01/16/2025] [Accepted: 02/13/2025] [Indexed: 03/20/2025]
Abstract
Background Proteinase 3 (PRTN3) has been linked to the progression of different cancer types. In this study, the expression and cell biological function of PRTN3 were investigated in gastric cancer (GC) to assess its role in GC progression. Methods The PRTN3 levels in 20 pairs of GC tissues were detected via quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) and Western blotting, while immunohistochemical staining was used to assess the PRTN3 levels in 47 GC tissue samples. The effects of stable lentivirus-mediated PRTN3 knockdown on GC cell proliferative, cell cycle, and apoptotic activity were evaluated using Cell Counting Kit-8 (CCK-8) and colony formation assays, nude mouse models, and flow cytometry. Results Elevated levels of PRTN3 messenger RNA (mRNA) and protein were noted in GC tissues, mostly in the cytosol. High PRTN3 levels were positively correlated with GC tumor N staging. in vitro knockdown of PRTN3 suppressed cell cycle progression, promoted apoptotic induction, and decreased the concentrations of cell cycle-associated proteins (cyclin D1, CDK4, and CDK6) and apoptosis-related Bcl-2 while inducing the upregulation of Bax. Downregulation of PRTN3 inhibited GC cell growth both in vitro and in mouse models. Conclusions Our study found that high expression of PRTN3 is associated with GC tumor N staging. And PRTN3 silencing could regulate GC progression by inhibiting the cell cycle and promoting apoptosis in GC cells, which could be a potential target for GC diagnosis and treatment.
Collapse
Affiliation(s)
- Haoyu Zhu
- Department of Gastrointestinal Surgery, Nantong First People Hospital, Medical School of Nantong University, Nantong, China
| | - Fei Wang
- Department of Gastrointestinal Surgery, Nantong First People Hospital, Nantong, China
| | - Xinran Lu
- Department of Gastrointestinal Surgery, Nantong First People Hospital, Medical School of Nantong University, Nantong, China
| | - Hao Wu
- Department of Gastrointestinal Surgery, Nantong First People Hospital, Nantong, China
| | - Chao Shi
- Department of Pathology, Nantong First People Hospital, Nantong, China
| | - Silvio Matsas
- Centro de Estudos e Pesquisas de Hematologia e Oncologia (CEPHO), Santo André, SP, Brazil
| | | | | | - Chong Tang
- Department of Gastrointestinal Surgery, Nantong First People Hospital, Nantong, China
| | - Shichun Feng
- Department of Gastrointestinal Surgery, Nantong First People Hospital, Nantong, China
| |
Collapse
|
18
|
Zheng Y, Shi J. EFNB1 drives glioma progression and shapes the immune microenvironment: a potential prognostic biomarker. Discov Oncol 2025; 16:249. [PMID: 40014231 PMCID: PMC11868007 DOI: 10.1007/s12672-025-01867-y] [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: 11/22/2024] [Accepted: 02/03/2025] [Indexed: 02/28/2025] Open
Abstract
Gliomas, a heterogeneous group of tumors affecting the brain and spinal cord, present a significant clinical challenge. Ephrin B1 (EFNB1) has been implicated in various malignancies. However, its role in gliomas remained poorly understood. Hence, this study aimed to elucidate the connection between EFNB1 and the progression of glioma. A retrospective RNA-seq analysis was conducted by utilizing the data from glioma patients in the TCGA and CGGA databases. Kaplan-Meier survival analysis and multivariate regression models were employed to evaluate the prognostic significance of EFNB1. RT-PCR was used to quantify EFNB1 expression in glioma tissues and cell lines. Meanwhile, in vitro assays were carried out to assess its functional roles in glioma cells. Our findings demonstrated that EFNB1 expression was significantly elevated in gliomas and other cancers. Moreover, high EFNB1 expression was closely correlated with advanced clinical stages and poor prognosis. Notably, multivariate analysis identified EFNB1 as an independent prognostic factor for overall survival. KEGG pathway analysis suggested that EFNB1 was involved in critical biological processes, including the cell cycle, protein processing in the endoplasmic reticulum, Epstein-Barr virus infection, and Salmonella infection. Furthermore, EFNB1 expression was associated with immune cell infiltration, particularly Th2 cells, macrophages, and plasmacytoid dendritic cells. In glioma cells, EFNB1 expression was markedly increased. Consequently, functional experiments demonstrated that EFNB1 knockdown inhibited glioma cell proliferation, invasion, and migration. These results highlighted EFNB1 as a novel independent prognostic biomarker and suggest its potential role in shaping the immunological microenvironment of gliomas.
Collapse
Affiliation(s)
- Yungui Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou, China
| | - Jiasong Shi
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou, China.
| |
Collapse
|
19
|
Sun Y, Cai J, Zhang Y, Bao S. A high concentration of neutrophil extracellular traps is observed in humans and mice suffering from endometriosis. J Reprod Immunol 2025; 167:104414. [PMID: 39657366 DOI: 10.1016/j.jri.2024.104414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/12/2024]
Abstract
We wished to ascertain if there is an association between neutrophil extracellular traps and endometriosis (EMS). We collected the lesional tissues and normal endometrium of 30 patients suffering from endometriosis. Samples were also taken from healthy controls. Blood from the peripheral circulation was collected to isolate serum and neutrophils. A mouse model of endometriosis was also created. Expression of citrullinated histone and the myeloperoxidase level in tissue were measured by immunofluorescence staining and western blotting. The myeloperoxidase level in peripheral blood serum was measured by enzyme-linked immunosorbent assay. Staining (Trypan Blue) and flow cytometry were used to measure the apoptosis of neutrophils in peripheral blood. BALB/C mice were modeled by allotransplantation, and the experimental parameters noted above quantified. The myeloperoxidase content in the peripheral blood of patients with endometriosis was increased compared with that in healthy controls. Flow cytometry showed that the percent apoptosis of neutrophils in patients with endometriosis was lower than that in healthy controls. Expression of citrullinated histone was higher in the endometriosis group in humans and mice compared with respective controls according to immunofluorescence staining and western blotting. Our data suggest that a high concentration of neutrophil extracellular traps was observed in humans and mice suffering from endometriosis.
Collapse
Affiliation(s)
- Yuting Sun
- Department of Gynecology and Obstetrics Mudanjiang Medical University Affliated Honggi Hospital, No.5 Tongxiang Road, Aimin District, Mudanjiang,Heilongjiang 157011, China
| | - Junhong Cai
- Medical Laboratory Center, Hainan General Hospital, Haikou 570102, China
| | - Yanan Zhang
- Department of Gynecology and Obstetrics Mudanjiang Medical University Affliated Honggi Hospital, No.5 Tongxiang Road, Aimin District, Mudanjiang,Heilongjiang 157011, China
| | - Shan Bao
- Department of Gynaecology and Obstetrics, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China.
| |
Collapse
|
20
|
Barchi A, Dell’Anna G, Massimino L, Mandarino FV, Vespa E, Viale E, Passaretti S, Annese V, Malesci A, Danese S, Ungaro F. Unraveling the pathogenesis of Barrett's esophagus and esophageal adenocarcinoma: the "omics" era. Front Oncol 2025; 14:1458138. [PMID: 39950103 PMCID: PMC11821489 DOI: 10.3389/fonc.2024.1458138] [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: 07/06/2024] [Accepted: 12/10/2024] [Indexed: 02/16/2025] Open
Abstract
Barrett's esophagus (BE) represents a pre-cancerous condition that is characterized by the metaplastic conversion of the squamous esophageal epithelium to a columnar intestinal-like phenotype. BE is the consequence of chronic reflux disease and has a potential progression burden to esophageal adenocarcinoma (EAC). The pathogenesis of BE and EAC has been extensively studied but not completely understood, and it is based on two main hypotheses: "transdifferentiation" and "transcommitment". Omics technologies, thanks to the potentiality of managing huge amounts of genetic and epigenetic data, sequencing the whole genome, have revolutionized the understanding of BE carcinogenesis, paving the way for biomarker development helpful in early diagnosis and risk progression assessment. Genomics and transcriptomics studies, implemented with the most advanced bioinformatics technologies, have brought to light many new risk loci and genomic alterations connected to BE and its progression to EAC, further exploring the complex pathogenesis of the disease. Early mutations of the TP53 gene, together with late aberrations of other oncosuppressor genes (SMAD4 or CKND2A), represent a genetic driving force behind BE. Genomic instability, nonetheless, is the central core of the disease. The implementation of transcriptomic and proteomic analysis, even at the single-cell level, has widened the horizons, complementing the genomic alterations with their transcriptional and translational bond. Increasing interest has been gathered around small circulating genetic traces (circulating-free DNA and micro-RNAs) with a potential role as blood biomarkers. Epigenetic alterations (such as hyper or hypo-methylation) play a meaningful role in esophageal carcinogenesis as well as the study of the tumor micro-environment, which has led to the development of novel immunological therapeutic options. Finally, the esophageal microbiome could be the protagonist to be investigated, deepening our understanding of the subtle association between the host microbiota and tumor development.
Collapse
Affiliation(s)
- Alberto Barchi
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Giuseppe Dell’Anna
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS Policlinico San Donato, Milan, Italy
| | - Luca Massimino
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Faculty of Medicine, Milan, Italy
| | | | - Edoardo Vespa
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Edi Viale
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Sandro Passaretti
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Vito Annese
- Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS Policlinico San Donato, Milan, Italy
- Università Vita-Salute San Raffaele, Faculty of Medicine, Milan, Italy
| | - Alberto Malesci
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS Policlinico San Donato, Milan, Italy
| | - Silvio Danese
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Faculty of Medicine, Milan, Italy
| | - Federica Ungaro
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
21
|
Tang S, Xu J, Wan P, Jin S, Zhang Y, Xun L, Wang J, Luo M, Chen W, Zuo Z, Tang H, Qi J. Recent advances in the role of high-salt diet in anti- and pro-cancer progression. Front Immunol 2025; 16:1542157. [PMID: 39944693 PMCID: PMC11814453 DOI: 10.3389/fimmu.2025.1542157] [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: 12/09/2024] [Accepted: 01/13/2025] [Indexed: 05/09/2025] Open
Abstract
Dietary behaviors significantly influence tumor progression, with increasing focus on high-salt diets (HSD) in recent years. Traditionally, HSD has been regarded as a major risk factor for multiple health issues, including hypertension, cardiovascular disease, kidney disease, cancer, and osteoporosis. However, recent studies have uncovered a novel aspect of HSD, suggesting that HSD may inhibit tumor growth in specific pathological conditions by modulating the activity of immune cells that infiltrate tumors and enhancing the effectiveness of PD-1 immunotherapy. This review focused on the duel molecular mechanisms of HSD in cancer development, which are based on the tumor microenvironment, the gut microbiota, and the involvement of sodium transporter channels. The objective of this review is to explore whether HSD could be a potential future oncological therapeutic strategy under specific situation.
Collapse
Affiliation(s)
- Shiwei Tang
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Juan Xu
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Ping Wan
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Province Clinical Research Center for Senile Diseases, First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Shumen Jin
- Yunnan Institute of Food and Drug Supervision and Control, Medical Products Administration of Yunnan Province, Kuming, Yunnan, China
| | - Ying Zhang
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Linting Xun
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jinli Wang
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Mei Luo
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Wenjie Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
- Guangdong-Hongkong-Macao Joint Laboratory of Respiratory Infectious Disease, Guangzhou Medical University, Guangzhou, China
| | - Zan Zuo
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Clinical Virology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Hui Tang
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jialong Qi
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Province Clinical Research Center for Senile Diseases, First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Clinical Virology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Birth Defects and Genetic Diseases, First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| |
Collapse
|
22
|
Syed Altaf RR, Mohan A, Palani N, Mendonce KC, Monisha P, Rajadesingu S. A review of innovative design strategies: Artificial antigen presenting cells in cancer immunotherapy. Int J Pharm 2025; 669:125053. [PMID: 39667594 DOI: 10.1016/j.ijpharm.2024.125053] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 11/07/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024]
Abstract
Developing nanocarriers that can carry medications directly to tumors is an exciting development in cancer nanomedicine. The efficacy of this intriguing therapeutic approach is, however, compromised by intricate and immunosuppressive circumstances that arise concurrently with the onset of cancer. The artificial antigen presenting cell (aAPC), a micro or nanoparticle based device that mimics an antigen presenting cell by providing crucial signal proteins to T lymphocytes to activate them against cancer, is one cutting-edge method for cancer immunotherapy. This review delves into the critical design considerations for aAPCs, particularly focusing on particle size, shape, and the non-uniform distribution of T cell activating proteins on their surfaces. Adequate surface contact between T cells and aAPCs is essential for activation, prompting engineers to develop nano-aAPCs with microscale contact areas through techniques such as shape modification and nanoparticle clustering. Additionally, we explore recommendations for future advancements in this field.
Collapse
Affiliation(s)
- Rabiya Riffath Syed Altaf
- Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India; Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Agilandeswari Mohan
- Department of BioChemistry, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India; Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Naveen Palani
- Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India; Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Keren Celestina Mendonce
- Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India; Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - P Monisha
- PG & Research Department of Physics, Sri Sarada College for Women, Salem - 636016, Tamil Nadu, India
| | - Suriyaprakash Rajadesingu
- Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India.
| |
Collapse
|
23
|
Fan LL, Wang XW, Zhang XM, Wei ZY, Wu HY, Yang QX, Fu D, de Mello RA, Lin JW, Yu H, Jiang GX. GNGT1 remodels the tumor microenvironment and promotes immune escape through enhancing tumor stemness and modulating the fibrinogen beta chain-neutrophil extracellular trap signaling axis in lung adenocarcinoma. Transl Lung Cancer Res 2025; 14:239-259. [PMID: 39958208 PMCID: PMC11826275 DOI: 10.21037/tlcr-2024-1200] [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: 12/09/2024] [Accepted: 01/17/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND Despite the recent advancements in the treatment of cancer, the 5-year survival of patients with non-small cell lung cancer (NSCLC) remains unsatisfactory. Lung adenocarcinoma (LUAD) is NSCLC's most common subtype, and metastasis is the major cause of death in patients with cancer. Therefore, identifying novel targets associated with metastasis in NSCLC is crucial to improving treatment. This study aimed to characterize the expression of GNGT1 in LUAD and to clarify the mechanism underlying the association between the higher expression level of GNGT1 and worse prognosis in patients. METHODS The transcriptome datasets and clinical information of patients with LUAD were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database. Bioinformatics analyses were performed in 515 patients who were stratified into two groups (high- and low-GNGT1 expression group) according to the GNGT1 level. Overall survival, DNA promotor methylation, immune cell infiltration, gene set enrichment analysis (GSEA), and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed to elucidate the functions of GNGT1 and to identify the related hub genes in LUAD. Their expression and functions in LUAD were verified using tissues from patients and transgenic mice overexpressing GNGT1 under the control of a lung-specific promoter (Scgb1a1-Cre). RESULTS GNGT1 was overexpressed in patients with LUAD and was associated with poor prognosis. GNGT1 expression was significantly correlated with gene alteration and hypomethylated promoter status. High GNGT1 expression in patients with LUAD was associated with advanced lymph node metastasis and the degree of immune cell infiltration. Functional enrichment analyses indicated that differentially expressed genes (DEGs) in the high-GNGT1 group participated in DNA replication, DNA replication preinitiation, and M phase, while cell adhesion molecules, apoptosis, and natural killer cell-mediated cytotoxicity were all downregulated. Messenger RNA and protein levels were correspondingly regulated in human LUAD tissues and the Scgb1a1-Cre; LSL-GNGT1 mouse model (GNGT1fl/+ mice). CONCLUSIONS GNGT1 was associated with tumor cell proliferation via the enhancement of tumor cell stemness and interaction with driver genes. Elevated GNGT1 expression promoted epithelial-mesenchymal transformation, remodeled the tumor microenvironment, and led to tumor metastasis, ultimately worsening the survival-related prognosis of patients with LUAD.
Collapse
Affiliation(s)
- Lin-Lin Fan
- Department of Pathology, The Affiliated Taizhou People’s Hospital of Dalian Medical University, Taizhou, China
- Department of Pathology, Linyi People’s Hospital, Linyi, China
- Department of Thoracic Surgery, Navy Military Medical University Affiliated Changhai Hospital, Shanghai, China
| | - Xiao-Wei Wang
- Department of Thoracic Surgery, Navy Military Medical University Affiliated Changhai Hospital, Shanghai, China
| | - Xiu-Mei Zhang
- Department of Pathology, The People’s Hospital of Xinghua City, Xinghua, China
| | - Zhi-Yong Wei
- Department of Pathology, Linyi People’s Hospital, Linyi, China
| | - Hui-Yi Wu
- Department of Pathology, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, China
- Department of Pathology, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Qin-Xin Yang
- Department of Pathology, The Affiliated Taizhou People’s Hospital of Dalian Medical University, Taizhou, China
- Department of Pathology, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Da Fu
- Department of Pathology, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Ramon Andrade de Mello
- Division of Medical Oncology, Nine of July University (UNINOVE), São Paulo, SP, Brazil
- Medical Oncology MSc Programme, University of Buckingham, Buckingham, UK
| | - Jie-Wei Lin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hong Yu
- Department of Pathology, The Affiliated Taizhou People’s Hospital of Dalian Medical University, Taizhou, China
- Department of Pathology, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, China
- Department of Pathology, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Geng-Xi Jiang
- Department of Thoracic Surgery, Navy Military Medical University Affiliated Changhai Hospital, Shanghai, China
| |
Collapse
|
24
|
Ring BZ, Cronister CT, Ring HZ, Ross DT, Seitz RS. Immune infiltrate populations within distinct tumor immune microenvironments predictive of immune checkpoint treatment outcome. Sci Rep 2025; 15:3126. [PMID: 39856115 PMCID: PMC11760962 DOI: 10.1038/s41598-024-83915-1] [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: 09/14/2024] [Accepted: 12/18/2024] [Indexed: 01/27/2025] Open
Abstract
Understanding the dynamic tumor immune microenvironment (TIME) is important in guiding immunotherapy. We have previously validated signatures predictive of checkpoint inhibitor efficacy which distinguish immunomodulatory, mesenchymal stem-like, and mesenchymal phenotypes. Here we use twenty tumor types (7162 samples) to identify potentially conserved immune biology within these TIME spaces, genes co-expressed across distinct cell types involved these immune processes, and the association of these signatures with ICI response. One signature, which contained multiple B-cell markers, was associated with immunotherapy efficacy in three cohorts, including IMvigor210. This signature of potentially conserved B-cell biology in co-infiltrated immune cell ecosystems had a more consistent association with outcome than comparable single cell type models and likely reflects a complex immunological response involving multilayered relationships between distinct immune effector cell types. These signatures were most highly expressed in tumors with prominent immune cell invasion, however there was consistent identification of infiltrate presence in relatively immune restricted cases. This suggests that these immune population signatures may identify conserved immune cell type co-infiltrate physiology of the TIME that best captures immune physiology with potential clinical utility.
Collapse
Affiliation(s)
| | | | - Huijun Z Ring
- Department of Medicine, Stanford University, Stanford, CA, USA
| | | | | |
Collapse
|
25
|
Zakurdaev EI, Bagateliya ZA, Titov KS, Elkhouli E, Chizhikov NP, Kharina DV. The Role of Tumor-Associated Neutrophils in Early Luminal HER2-Negative Breast Cancer Progression. Asian Pac J Cancer Prev 2025; 26:207-213. [PMID: 39874003 PMCID: PMC12082427 DOI: 10.31557/apjcp.2025.26.1.207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 01/11/2025] [Indexed: 01/30/2025] Open
Abstract
OBJECTIVES To study the predictive role of tumor-associated neutrophils in early luminal HER2-negative breast cancer. MATERIALS AND METHODS This is a retrospective study conducted on 60 women cases aged from 31 to 79 years underwent surgery for luminal HER2-negative ductal breast cancer in tertiary care cancer centre. We first estimated basic morphological signs: tumor size, tumor grade (by Nottingham Histologic Score), tumor infiltrating lymphocytes (TILs), Lymphovascular invasion, hormonal receptors status, proliferative index, and regional lymph nodes metastasis. The expression of intratumoral neutrophils was studied by CD15 immunohistochemistry which was performed using tissue microarrays. The total number of intratumoral neutrophils, were counted in 5 high-power fields. RESULTS According to the Nottingham histologic score system, grade I cases were detected in 10 cases (16%), grade II in 34 cases (57%), and grade III in 16 cases (27%). Lymphovascular invasion was determined in 23 cases (38%), and perineural invasion in 14 cases (23%). Number of TILs varied from 0 to 14 (counted in 5 HPF) and averaged 4.2±0.5. Luminal A tumor phenotype was detected in 35 cases (58%), and luminal B HER2-negative in 25 cases (42%). Nineteen (32%) women had metastases in regional lymph nodes (N+). The number of tumor microenvironment neutrophils in luminal HER2-negative breast carcinomas ranged from 1 to 10 (counted in 5 HPF) with an average value of 2.7±0.4. High tumor-associated neutrophils concentration significantly correlated with tumor size (<5mm and >20mm) with p=0.05, high grade (p=0.01), high proliferative index ((r=0.67; p=0.05), TILs (p=0.05), Lymphovascular space invasion (p=0.01)and positive regional lymph nodes metastasis (p=0.001), but not perineural invasion (p=0.1) and also, did not correlate with the expression of estrogen (r=0.18) and progesterone (r=0.14) receptors. CONCLUSION Tumor-associated neutrophils strongly predict a worse prognosis in early luminal HER2-negative breast cancer.
Collapse
MESH Headings
- Humans
- Female
- Neutrophils/pathology
- Neutrophils/metabolism
- Neutrophils/immunology
- Middle Aged
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Breast Neoplasms/surgery
- Retrospective Studies
- Receptor, ErbB-2/metabolism
- Aged
- Adult
- Lymphatic Metastasis
- Prognosis
- Disease Progression
- Follow-Up Studies
- Lymphocytes, Tumor-Infiltrating/pathology
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/secondary
- Carcinoma, Ductal, Breast/surgery
- Biomarkers, Tumor/metabolism
- Neoplasm Invasiveness
- Receptors, Estrogen/metabolism
Collapse
Affiliation(s)
| | | | | | - Ekbal Elkhouli
- Pathology Department, Mansoura University, Mansoura, Egypt.
| | | | | |
Collapse
|
26
|
Wu Q, Zhao H. Prognostic and clinicopathological role of pretreatment systemic inflammation response index (SIRI) in gastric cancer: a systematic review and meta-analysis. World J Surg Oncol 2024; 22:333. [PMID: 39707359 DOI: 10.1186/s12957-024-03602-3] [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: 03/23/2024] [Accepted: 11/24/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND The systemic inflammatory response index (SIRI) is calculated via the following formula: SIRI = monocyte count × neutrophil count/lymphocyte count. The value of the SIRI in predicting the prognosis of gastric cancer (GC) remains controversial. This study revealed the precise effect of the SIRI in predicting GC prognosis through a meta-analysis. METHODS The ability of the SIRI to predict GC prognosis was evaluated by calculating combined hazard ratios (HRs) and 95% confidence intervals (CIs). Furthermore, the combined odds ratios (ORs) and 95% CIs were determined to analyze the associations between the SIRI and the clinicopathological characteristics of patients with GC. RESULTS Seven publications on a total of 1763 cases were included in this study. The SIRI threshold was between 0.58 and 1.35, and the median value was 0.85. Our pooled findings revealed that a higher SIRI was significantly linked with poor overall survival (OS) (HR = 1.87, 95% CI = 1.59-2.20, p < 0.001) and disease-free survival (DFS; HR = 1.88, 95% CI = 1.50-2.36, p < 0.001) in GC patients. However, the SIRI did not exhibit a significant association with sex (OR = 1.98, 95% CI = 0.82-4.75, p = 0.126), surgery type (OR = 0.96, 95% CI = 0.61-1.51, p = 0.847), tumor differentiation (OR = 0.75, 95% CI = 0.54-1.06, p = 0.099), or TNM stage (OR = 1.25, 95% CI = 0.34-4.62, p = 0.743) in patients with GC. CONCLUSIONS An elevated SIRI was significantly associated with unfavorable OS and DFS in patients with GC. Thus, the SIRI is a reliable biomarker for predicting GC prognosis in clinical practice.
Collapse
Affiliation(s)
- Quan Wu
- Clinical Laboratory, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, 313000, Zhejiang, China
| | - Hui Zhao
- Department of Gastroenterology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, 313000, Zhejiang, China.
| |
Collapse
|
27
|
Wang X, Yin X, Li Y, Zhang S, Hu M, Wei M, Li Z. Novel insight and perspectives of nanoparticle-mediated gene delivery and immune-modulating therapies for pancreatic cancer. J Nanobiotechnology 2024; 22:771. [PMID: 39696302 DOI: 10.1186/s12951-024-02975-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 11/04/2024] [Indexed: 12/20/2024] Open
Abstract
Current standard-of-care therapies have failed to improve the survival of patients with metastatic pancreatic cancer (PCA). Therefore, exploring novel therapeutic approaches for cancer targeting is of utmost need. During the past few years, many efforts have been made to develop conventional treatment strategies to reduce chemotherapy resistance. However, critical challenges have impeded current cancer management outcomes, and limited clinical responses have been achieved due to unfavorable off-target effects. Advances in nanotechnology-based gene and immune-modulator delivery systems have excellent advantages for improving the therapeutic efficacy of PCA and provide promising avenues for overcoming the immunosuppressive tumor microenvironment and enhancing patient treatment outcomes. This review article provides insight into the challenges, opportunities, and future perspectives of these novel emerging nanoparticles based on lipid, polymer, and inorganic metal carriers to modulate genes and immunotherapy paradigms for PCA anticancer activity.
Collapse
Affiliation(s)
- Xinqiao Wang
- School of Pharmacy, China Medical University, Shenyang, Liaoning Province, 110122, P.R. China
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang, Liaoning Province, 110001, P.R. China
| | - Xue Yin
- School of Pharmacy, China Medical University, Shenyang, Liaoning Province, 110122, P.R. China
| | - Yuxin Li
- School of Pharmacy, China Medical University, Shenyang, Liaoning Province, 110122, P.R. China
| | - Shuhui Zhang
- School of Pharmacy, China Medical University, Shenyang, Liaoning Province, 110122, P.R. China
| | - Meie Hu
- School of Pharmacy, China Medical University, Shenyang, Liaoning Province, 110122, P.R. China
| | - Minjie Wei
- School of Pharmacy, China Medical University, Shenyang, Liaoning Province, 110122, P.R. China.
| | - Zhenhua Li
- School of Pharmacy, China Medical University, Shenyang, Liaoning Province, 110122, P.R. China.
| |
Collapse
|
28
|
Weijie S. Annexin A2: the feasibility of being a therapeutic target associated with cancer metastasis and drug resistance in cancer microenvironment. Discov Oncol 2024; 15:783. [PMID: 39692932 DOI: 10.1007/s12672-024-01693-8] [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: 09/26/2024] [Accepted: 12/10/2024] [Indexed: 12/19/2024] Open
Abstract
At present, there is still a lack of effective treatment strategies for cancer metastasis and drug resistance, so finding effective biomarkers is particularly important. AnnexinA2 (ANXA2), a vital membrane protein, critically influences cancer progression, tumor invasion, and tumor microenvironment modulation. To assess the possible application of ANXA2 as a therapeutic target against cancer cell metastasis and drug resistance to chemotherapeutic drugs in the tumor microenvironment, we elucidated the functionality of ANXA2 in stromal cells, angiogenic vascular cells, and infiltrated immune cells that mediate metastasis and drug resistance, as well as its potential as a therapeutic target. ANXA2 shows a high expression level in many tissues, and its expression level is even higher in several tumors and their microenvironments. ANXA2 is a crucial regulator of many factors and may serve as a target against drug-resistant cancers.
Collapse
Affiliation(s)
- Song Weijie
- Laboratory Animal Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin, 300060, China.
| |
Collapse
|
29
|
Fang J, Rao X, Wang C, Wang Y, Wu C, Zhou R. Role of exosomes in modulating non-small cell lung cancer radiosensitivity. Front Pharmacol 2024; 15:1471476. [PMID: 39737074 PMCID: PMC11683128 DOI: 10.3389/fphar.2024.1471476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 12/02/2024] [Indexed: 01/01/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) constitutes a significant proportion of lung cancer cases, and despite advancements in treatment modalities, radiotherapy resistance remains a substantial hurdle in effective cancer management. Exosomes, which are small vesicles secreted by cells, have emerged as pivotal players in intercellular communication and influence various biological processes, including cancer progression and the response to therapy. This review discusses the intricate role of exosomes in the modulation of NSCLC radiosensitivity. The paper focuses on NSCLC and highlights how tumor-derived exosomes contribute to radioresistance by enhancing DNA repair, modulating immune responses, and altering the tumor microenvironment. We further explore the potential of mesenchymal stem cell-derived exosomes to overcome radiotherapy resistance and their potential as biomarkers for predicting therapeutic outcomes. Understanding the mechanisms by which exosomes affect radiotherapy can provide new avenues for enhancing treatment efficacy and improving the survival rates of patients with NSCLC.
Collapse
Affiliation(s)
- Jincheng Fang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinrui Rao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Changjian Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yangchenxi Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Chuangyan Wu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science, Wuhan, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| |
Collapse
|
30
|
Deng X, Zhang Q, Jin F, Lu F, Duan G, Han L, Zhu M, Yang Z, Zhang G. Ailanthone disturbs cross-talk between cancer cells and tumor-associated macrophages via HIF1-α/LINC01956/FUS/β-catenin signaling pathway in glioblastoma. Cancer Cell Int 2024; 24:397. [PMID: 39639311 PMCID: PMC11619249 DOI: 10.1186/s12935-024-03594-w] [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: 07/08/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND An increasing number of studies have focused on ailanthone (aila) due to its antitumor activity. However, the role of ailanthone in glioblastoma(GBM) has not been investigated before. This study aims to explore the biological function and the underlying mechanism of ailanthone in GBM. METHODS The microarray analysis was used to screen out down-stream long non-coding RNAs (lncRNAs) targeted by ailanthone. Real-time PCR(RT-PCR) assay was used to examine LINC01956 expression levels. Colony-formation, Methylthiazolyldiphenyl-tetrazolium bromide(MTT), cell-cycle, organoids culture and in-vivo tumorigenesis assays were used to examine cell growth in vitro and in vivo. Boyden assay was used to examine cell invasion ability in vitro. RNA immunoprecipitation and RNA-protein pull-down assays were used to examine the interaction between LINC01956 and FUS protein. Chromatin Immunoprecipitation(ChIP) assay was used to examine HIF1-α-binding sites in the LINC01956 promoter. RESULTS Ailanthone decreased GBM cell growth in vitro and in vivo via inducing ferroptosis. Ailanthone treatment exhibited blood‒brain barrier(BBB) permeability and specifically targeted the tumor area. LINC01956 was identified as a down-stream target of Ailanthone. LINC01956 exerted as an onco-lncRNA in GBM. M2 polarization of macrophages induced by exosomes derived from glioma cells overexpressing LINC01956 accelerated GBM progression. Mechanistically, we found that LINC01956 bound to FUS and reduced its ubiquitination. LINC01956 evoked nuclear translocation of phosphorylated (p)-β-catenin by recruiting FUS. Furthermore, under hypoxic conditions, LINC01956 was regulated by HIF-1α. Ailanthone decreased the expression of LINC01956 via suppressing HIF-1α. CONCLUSION Taken together, our data revealed for the first time that ailanthone regulated HIF-1α/LINC01956/FUS/β-catenin signaling pathway and thereby inhibited GBM progression.
Collapse
Affiliation(s)
- Xubin Deng
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qianbing Zhang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Fa Jin
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangzhou, China
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Fengfei Lu
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangzhou, China
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Guosheng Duan
- The Radiotherapy Department of Shanxi Province People Hospital, Taiyuan, China
| | - Luwei Han
- The Radiotherapy Department of Shanxi Province People Hospital, Taiyuan, China
| | - Meiling Zhu
- The Radiotherapy Department of Shanxi Province People Hospital, Taiyuan, China
| | - Zhengyan Yang
- The Radiotherapy Department of Shanxi Province People Hospital, Taiyuan, China
| | - Gong Zhang
- The Radiotherapy Department of Shanxi Province People Hospital, Taiyuan, China.
| |
Collapse
|
31
|
Tobe-Nishimoto A, Morita Y, Nishimura J, Kitahira Y, Takayama S, Kishimoto S, Matsumiya-Matsumoto Y, Matsunaga K, Imai T, Uzawa N. Tumor microenvironment dynamics in oral cancer: unveiling the role of inflammatory cytokines in a syngeneic mouse model. Clin Exp Metastasis 2024; 41:891-908. [PMID: 39126553 PMCID: PMC11607012 DOI: 10.1007/s10585-024-10306-1] [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: 02/20/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024]
Abstract
The process of cervical lymph node metastasis is dependent on the phenotype of the tumor cells and their interaction with the host microenvironment and immune system; conventional research methods that focus exclusively on tumor cells are limited in their ability to elucidate the metastatic mechanism. In cancer tissues, a specialized environment called the tumor microenvironment (TME) is established around tumor cells, and inflammation in the TME has been reported to be closely associated with the development and progression of many types of cancer and with the response to anticancer therapy. In this study, to elucidate the mechanism of metastasis establishment, including the TME, in the cervical lymph node metastasis of oral cancer, we established a mouse-derived oral squamous cell carcinoma cervical lymph node highly metastatic cell line and generated a syngeneic orthotopic transplantation mouse model. In the established highly metastatic cells, epithelial-mesenchymal transition (EMT) induction was enhanced compared to that in parental cells. In the syngeneic mouse model, lymph node metastasis was observed more frequently in tumors of highly metastatic cells than in parental cells, and Cyclooxygenase-2 (COX-2) expression and lymphatic vessels in primary tumor tissues were increased, suggesting that this model is highly useful. Moreover, in the established highly metastatic cells, EMT induction was enhanced compared to that in the parent cell line, and CCL5 and IL-6 secreted during inflammation further enhanced EMT induction in cancer cells. This suggests the possibility of a synergistic effect between EMT induction and inflammation. This model, which allows for the use of two types of cells with different metastatic and tumor growth potentials, is very useful for oral cancer research involving the interaction between cancer cells and the TME in tumor tissues and for further searching for new therapeutic agents.
Collapse
Affiliation(s)
- Ayano Tobe-Nishimoto
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan
| | - Yoshihiro Morita
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan.
| | - Junya Nishimura
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan
| | - Yukiko Kitahira
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan
| | - Shun Takayama
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan
| | - Satoko Kishimoto
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan
| | - Yuka Matsumiya-Matsumoto
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan
| | - Kazuhide Matsunaga
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan
| | - Tomoaki Imai
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan
| | - Narikazu Uzawa
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan
| |
Collapse
|
32
|
Dalpati N, Rai SK, Dash SP, Kumar P, Singh D, Sarangi PP. Integrins α5β1 and αvβ3 Differentially Participate in the Recruitment and Reprogramming of Tumor-associated Macrophages in the In Vitro and In Vivo Models of Breast Tumor. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1553-1568. [PMID: 39330703 DOI: 10.4049/jimmunol.2400180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024]
Abstract
Tumor-associated macrophages (TAMs) drive the protumorigenic responses and facilitate tumor progression via matrix remodeling, angiogenesis, and immunosuppression by interacting with extracellular matrix proteins via integrins. However, the expression dynamics of integrin and its correlation with TAM functional programming in the tumors remain unexplored. In this study, we examined surface integrins' role in TAM recruitment and phenotypic programming in a 4T1-induced murine breast tumor model. Our findings show that integrin α5β1 is upregulated in CD11b+Ly6Chi monocytes in the bone marrow and blood by day 10 after tumor induction. Subsequent analysis revealed elevated integrin α5β1 expression on tumor-infiltrating monocytes (Ly6ChiMHC class II [MHCII]low) and M1 TAMs (F4/80+Ly6ClowMHCIIhi), whereas integrin αvβ3 was predominantly expressed on M2 TAMs (F4/80+Ly6ClowMHCIIlow), correlating with higher CD206 and MERTK expression. Gene profiling of cells sorted from murine tumors showed that CD11b+Ly6G-F4/80+α5+ TAMs had elevated inflammatory genes (IL-6, TNF-α, and STAT1/2), whereas CD11b+Ly6G-F4/80+αv+ TAMs exhibited a protumorigenic phenotype (IL-10, Arg1, TGF-β, and STAT3/6). In vitro studies demonstrated that blocking integrin α5 and αv during macrophage differentiation from human peripheral blood monocytes reduced cell spreading and expression of CD206 and CD163 in the presence of specific matrix proteins, fibronectin, and vitronectin. Furthermore, RNA sequencing data analysis (GEO dataset: GSE195857) from bone marrow-derived monocytes and TAMs in 4T1 mammary tumors revealed differential integrin α5 and αv expression and their association with FAK and SRC kinase. In line with this, FAK inhibition during TAM polarization reduced SRC, STAT1, and STAT6 phosphorylation. In conclusion, these findings underscore the crucial role of integrins in TAM recruitment, polarization, and reprogramming in tumors.
Collapse
Affiliation(s)
- Nibedita Dalpati
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Shubham Kumar Rai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Shiba Prasad Dash
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Puneet Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Divya Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Pranita P Sarangi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| |
Collapse
|
33
|
Pan Y, Fei L, Wang S, Chen H, Jiang C, Li H, Wang C, Yang Y, Zhang Q, Chen Y. Integrated analysis of single-cell, spatial and bulk RNA-sequencing identifies a cell-death signature for predicting the outcomes of head and neck cancer. Front Immunol 2024; 15:1487966. [PMID: 39575251 PMCID: PMC11578999 DOI: 10.3389/fimmu.2024.1487966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/16/2024] [Indexed: 11/24/2024] Open
Abstract
Background Cell death plays an essential role in carcinogenesis, but its function in the recurrence and postoperative prognosis of head and neck cancer (HNC), which ranks as the 7th most common malignancy globally, remains unclear. Methods Data from five main subtypes of HNC related single-cell RNA sequencing (scRNA-seq) were recruited to establish a single-cell atlas, and the distribution of cell death models (CDMs) across different tissues as well as cell subtypes were analyzed. Bulk RNA-seq from the Cancer Genome Atlas Program (TCGA) dataset was subjected to a machine learning-based integrative procedure for constructing a consensus cell death-related signature risk score (CDRscore) model and validated by external data. The biofunctions including different expression analysis, immune cell infiltration, genomic mutations, enrichment analysis as well as cellchat analysis were compared between the high- and low- risk score groups categorized by this CDRscore model. Finally, samples from laryngeal squamous cell cancer (LSCC) were conducted by spatial transcriptomics (ST) to further validate the results of CDRscore model. Results T cells from HNC patients manifested the highest levels of cell death while HPV infection attenuates malignant cell death based on single-cell atlas. CDMs are positively correlated with the tumor-cell stemness, immune-related score and T cells are infiltrated. A CDRscore model was established based on the transcription of ten cell death prognostic genes (MRPL10, DDX19A, NDFIP1, PCMT1, HPRT1, SLC2A3, EFNB2, HK1, BTG3 and MAP2K7). It functions as an independent prognostic factor for overall survival in HNC and displays stable and powerful performance validated by GSE41613 and GSE65858 datasets. Patients in high CDRscore manifested worse overall survival, more active of epithelial mesenchymal transition, TGF-β-related pathways and hypoxia, higher transcription of T cell exhausted markers, and stronger TP53 mutation. ST from LSCC showed that spots with high-risk scores were colocalized with TGF-β and the proliferating malignant cells, additionally, the risk scores have a negative correlation with TCR signaling but positive association with LAG3 transcription. Conclusion The CDRscore model could be utilized as a powerful prognostic indicator for HNC.
Collapse
Affiliation(s)
- Yue Pan
- Institute of Immunology, People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
| | - Lei Fei
- Institute of Immunology, People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
| | - Shihua Wang
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Hua Chen
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Changqing Jiang
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Hong Li
- Chongqing Renpin Otolaryngology Head and Neck Surgery Hospital, Chongqing, China
| | - Changsong Wang
- Department of Pathology, People’s Liberation Army Joint Logistic Support Force 989 Hospital, Luoyang, Henan, China
| | - Yao Yang
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Qinggao Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China
| | - Yongwen Chen
- Institute of Immunology, People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
| |
Collapse
|
34
|
Zhen X, Kim J, Kang JS, Choi BJ, Park KH, Lee DS, Hong SH, Lee JH. Homology-independent targeted insertion-mediated derivation of M1-biased macrophages harbouring Megf10 and CD3ζ from human pluripotent stem cells. EBioMedicine 2024; 109:105390. [PMID: 39383607 PMCID: PMC11497429 DOI: 10.1016/j.ebiom.2024.105390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND Macrophages engineered with chimeric antigen receptors (CAR) are suitable for immunotherapy based on their immunomodulatory activity and ability to infiltrate solid tumours. However, the production and application of genetically edited, highly effective, and mass-produced CAR-modified macrophages (CAR-Ms) are challenging. METHODS Here, we used homology-independent targeted insertion (HITI) for site-directed CAR integration into the safe-harbour region of human pluripotent stem cells (hPSCs). This approach, together with a simple differentiation protocol, produced stable and highly effective CAR-Ms without heterogeneity. FINDINGS These engineered cells phagocytosed cancer cells, leading to significant inhibition of cancer-cell proliferation in vitro and in vivo. Furthermore, the engineered CARs, which incorporated a combination of CD3ζ and Megf10 (referred to as FRP5Mζ), markedly enhanced the antitumour effect of CAR-Ms by promoting M1, but not M2, polarisation. FRP5Mζ promoted M1 polarisation via nuclear factor kappa B (NF-κB), ERK, and STAT1 signalling, and concurrently inhibited STAT3 signalling even under M2 conditions. These features of CAR-Ms modulated the tumour microenvironment by activating inflammatory signalling, inducing M1 polarisation of bystander non-CAR macrophages, and enhancing the infiltration of T cells in cancer spheroids. INTERPRETATION Our findings suggest that CAR-Ms have promise as immunotherapeutics. In conclusion, the guided insertion of CAR containing CD3ζ and Megf10 domains is an effective strategy for the immunotherapy of solid tumours. FUNDING This work was supported by KRIBB Research Initiative Program Grant (KGM4562431, KGM5282423) and a Korean Fund for Regenerative Medicine (KFRM) grant funded by the Korean government (Ministry of Science and ICT,Ministry of Health and Welfare) (22A0304L1-01).
Collapse
Affiliation(s)
- Xing Zhen
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea; Department of Nanoscience and Nanotechnology, Graduate School, Kyungpook National University, Daegu, 41566, Republic of Korea.
| | - Jieun Kim
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea; Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| | - Jong Soon Kang
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea.
| | - Byeong Jo Choi
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea.
| | - Ki Hwan Park
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea.
| | - Dong-Seok Lee
- Department of Nanoscience and Nanotechnology, Graduate School, Kyungpook National University, Daegu, 41566, Republic of Korea; School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea.
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea; KW-Bio Co., Ltd, Chuncheon, 24252, South Korea.
| | - Jong-Hee Lee
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
| |
Collapse
|
35
|
Liu Z, Li S, Xiao Y, Liu X, Zhang B, Zeng Q, Ao Q, Zhang X. A Multi-Functional Nanoadjuvant Coupling Manganese with Toll-Like 9 Agonist Stimulates Potent Innate and Adaptive Anti-Tumor Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402678. [PMID: 39258810 PMCID: PMC11538688 DOI: 10.1002/advs.202402678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/20/2024] [Indexed: 09/12/2024]
Abstract
The effectiveness of Toll-like 9 agonists (CpG) as an adjuvant for tumor immunotherapy is restricted due to their insufficient ability to activate anti-tumor immunity. To address that, the common nutrient metal ions are explored (Mn2+, Cu2+, Ca2+, Mg2+, Zn2+, Fe3+, and Al3+), identifying Mn2+ as a key enhancer of CpG to mediate immune activation by augmenting the STING-NF-κB pathway. Mn2+ and CpG are then self-assembled with epigallocatechin gallate (EGCG) into a nanoadjuvant MPN/CpG. Local delivery of MPN/CpG effectively inhibits tumor growth in a B16 melanoma-bearing mouse model, reshaping the tumor microenvironment (TME) by repolarizing M2-type tumor-associated macrophages (TAMs) to an M1-type and boosting intra-tumoral infiltration of CD8+/CD4+ T lymphocytes and DCs. Furthermore, compared to free CpG, MPN/CpG exhibits heightened accumulation in lymph nodes, enhancing CpG uptake and DC activation, consequently inducing significant antigen-specific cytotoxic CD8+ T cell immune response and humoral immunity. In a prophylactic tumor-bearing mouse model, MPN/CpG vaccination with OVA antigen significantly delays B16-OVA melanoma growth and extends mouse survival. These findings underscore the potential of MPN/CpG as a multifunctional adjuvant platform to drive powerful innate and adaptive immunity and regulate TME against tumors.
Collapse
MESH Headings
- Animals
- Female
- Mice
- Catechin/analogs & derivatives
- Catechin/chemistry
- CpG Islands
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Immunity, Humoral/drug effects
- Immunity, Innate/drug effects
- Injections, Intralesional
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Manganese/administration & dosage
- Manganese/chemistry
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice, Inbred C57BL
- Nanoparticle Drug Delivery System/chemistry
- Oligodeoxyribonucleotides/administration & dosage
- Oligodeoxyribonucleotides/chemistry
- Oligodeoxyribonucleotides/genetics
- RAW 264.7 Cells
- Skin Neoplasms/immunology
- Skin Neoplasms/pathology
- Skin Neoplasms/therapy
- Toll-Like Receptor 9/agonists
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
- Tumor-Associated Macrophages/drug effects
- Tumor-Associated Macrophages/immunology
- Immunotherapy, Active/methods
- Adjuvants, Vaccine/administration & dosage
- Adjuvants, Vaccine/chemistry
Collapse
Affiliation(s)
- Zhongjie Liu
- College of Biomedical EngineeringSichuan UniversityChengdu610064China
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial & Institute of Regulatory Science for Medical Device & National Engineering Research Center for BiomaterialsSichuan UniversityChengduSichuan610064China
| | - Shu Li
- College of Biomedical EngineeringSichuan UniversityChengdu610064China
| | - Yang Xiao
- College of Biomedical EngineeringSichuan UniversityChengdu610064China
| | - Xiaoyang Liu
- Orthopedic Research Institution, Department of OrthopedicsWest China Hospital, Sichuan UniversityChengdu610041China
| | - Bin Zhang
- College of Biomedical EngineeringSichuan UniversityChengdu610064China
| | - Qin Zeng
- College of Biomedical EngineeringSichuan UniversityChengdu610064China
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial & Institute of Regulatory Science for Medical Device & National Engineering Research Center for BiomaterialsSichuan UniversityChengduSichuan610064China
| | - Qiang Ao
- College of Biomedical EngineeringSichuan UniversityChengdu610064China
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial & Institute of Regulatory Science for Medical Device & National Engineering Research Center for BiomaterialsSichuan UniversityChengduSichuan610064China
| | - Xingdong Zhang
- College of Biomedical EngineeringSichuan UniversityChengdu610064China
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial & Institute of Regulatory Science for Medical Device & National Engineering Research Center for BiomaterialsSichuan UniversityChengduSichuan610064China
| |
Collapse
|
36
|
Mastrogiovanni M, Donnadieu E, Pathak R, Di Bartolo V. Subverting Attachment to Prevent Attacking: Alteration of Effector Immune Cell Migration and Adhesion as a Key Mechanism of Tumor Immune Evasion. BIOLOGY 2024; 13:860. [PMID: 39596815 PMCID: PMC11591779 DOI: 10.3390/biology13110860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024]
Abstract
Cell adhesion regulates specific migratory patterns, location, communication with other cells, physical interactions with the extracellular matrix, and the establishment of effector programs. Proper immune control of cancer strongly depends on all these events occurring in a highly accurate spatiotemporal sequence. In response to cancer-associated inflammatory signals, effector immune cells navigating the bloodstream shift from their patrolling exploratory migration mode to establish adhesive interactions with vascular endothelial cells. This interaction enables them to extravasate through the blood vessel walls and access the cancer site. Further adhesive interactions within the tumor microenvironment (TME) are crucial for coordinating their distribution in situ and for mounting an effective anti-tumor immune response. In this review, we examine how alterations of adhesion cues in the tumor context favor tumor escape by affecting effector immune cell infiltration and trafficking within the TME. We discuss the mechanisms by which tumors directly modulate immune cell adhesion and migration patterns to affect anti-tumor immunity and favor tumor evasion. We also explore indirect immune escape mechanisms that involve modifications of TME characteristics, such as vascularization, immunogenicity, and structural topography. Finally, we highlight the significance of these aspects in designing more effective drug treatments and cellular immunotherapies.
Collapse
Affiliation(s)
- Marta Mastrogiovanni
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Emmanuel Donnadieu
- Equipe Labellisée Ligue Contre le Cancer, CNRS, INSERM, Institut Cochin, Université Paris Cité, F-75014 Paris, France;
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Vincenzo Di Bartolo
- Immunoregulation Unit, Institut Pasteur, Université Paris Cité, F-75015 Paris, France;
| |
Collapse
|
37
|
Enemark MH, Jensen ML, Andersen MD, Plesner TL, Hamilton-Dutoit S, Ludvigsen M. Impact of the Immune Landscape in Follicular Lymphoma: Insights into Histological Transformation in the Rituximab Era. Cancers (Basel) 2024; 16:3553. [PMID: 39456647 PMCID: PMC11506075 DOI: 10.3390/cancers16203553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Follicular lymphoma (FL) presents significant clinical heterogeneity, with some patients experiencing transformation into an aggressive disease, a key contributor to FL-related mortality. Based on gene expression profiles, this study aimed to provide insights into immunological differences associated with transformation. Methods: Gene expression analysis using the NanoString nCounter Tumor Signaling 360 Panel was performed on diagnostic lymphoma samples from 70 FL patients diagnosed in the rituximab era, either non-transforming FL (nt-FL, n = 34) or subsequently transforming FL (st-FL, n = 36), with paired high-grade transformed FL (tFL, n = 36) samples available. In silico immunophenotyping was performed to infer immune cell infiltration using the CIBERSORTx algorithm. Results: The gene expression analysis revealed 164 significantly differentially expressed genes, distinguishing st-FL from nt-FL and generally presenting an upregulation of B cell-related genes (CD40, IRF4, RELB), immunosuppressive molecules (IL10, SOCS3), and immune checkpoint molecules (CD276, TIM3). Analysis of immune cell proportions indicated significant differences in infiltrates of M1-like macrophages (p = 0.007) and neutrophils (p = 0.012) in nt-FL versus st-FL samples. Transformation-free survival (TFS) was associated with high numbers of both these cellular subsets (p = 0.006 and 0 = 0.002, respectively). This was even more evident when combined with inferior TFS in lymphomas with high infiltrates of both cell types (p < 0.001). After transformation, tFL samples showed a reduction in T follicular helper cells (p = 0.008) and an increase in immunosuppressive M2-like macrophages and neutrophils (p < 0.001 and p = 0.028, respectively). Conclusion: By elucidating the distinct molecular and immune landscapes of FL at the time of diagnosis and transformation, this study underscores the importance of immune microenvironment in FL transformation and patient outcome.
Collapse
Affiliation(s)
- Marie Hairing Enemark
- Department of Hematology, Aarhus University Hospital, 8200 Aarhus, Denmark; (M.H.E.); (M.L.J.); (M.D.A.)
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Maja Lund Jensen
- Department of Hematology, Aarhus University Hospital, 8200 Aarhus, Denmark; (M.H.E.); (M.L.J.); (M.D.A.)
| | - Maja Dam Andersen
- Department of Hematology, Aarhus University Hospital, 8200 Aarhus, Denmark; (M.H.E.); (M.L.J.); (M.D.A.)
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | | | | | - Maja Ludvigsen
- Department of Hematology, Aarhus University Hospital, 8200 Aarhus, Denmark; (M.H.E.); (M.L.J.); (M.D.A.)
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
38
|
Wang Q, Yin X, Huang X, Zhang L, Lu H. Impact of mitochondrial dysfunction on the antitumor effects of immune cells. Front Immunol 2024; 15:1428596. [PMID: 39464876 PMCID: PMC11502362 DOI: 10.3389/fimmu.2024.1428596] [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: 05/08/2024] [Accepted: 09/24/2024] [Indexed: 10/29/2024] Open
Abstract
Mitochondrial dysfunction, a hallmark of immune cell failure, affects the antitumor effects of immune cells through metabolic reprogramming, fission, fusion, biogenesis, and immune checkpoint signal transduction of mitochondria. According to researchers, restoring damaged mitochondrial function can enhance the efficacy of immune cells. Nevertheless, the mechanism of mitochondrial dysfunction in immune cells in patients with cancer is unclear. In this review, we recapitulate the impact of mitochondrial dysfunction on the antitumor effects of T cells, natural killer cells, dendritic cells, and tumor-associated macrophage and propose that targeting mitochondria can provide new strategies for antitumor therapy.
Collapse
Affiliation(s)
- Quan Wang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiangzhi Yin
- Department of Orthopaedics, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaotong Huang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lu Zhang
- Department of Radiation Oncology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haijun Lu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
39
|
Huang T, Bei C, Hu Z, Li Y. CAR-macrophage: Breaking new ground in cellular immunotherapy. Front Cell Dev Biol 2024; 12:1464218. [PMID: 39421021 PMCID: PMC11484238 DOI: 10.3389/fcell.2024.1464218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Chimeric Antigen Receptor (CAR) technology has revolutionized cellular immunotherapy, particularly with the success of CAR-T cells in treating hematologic malignancies. However, CAR-T cells have the limited efficacy of against solid tumors. To address these limitations, CAR-macrophages (CAR-Ms) leverage the innate properties of macrophages with the specificity and potency of CAR technology, offering a novel and promising approach to cancer immunotherapy. Preclinical studies have shown that CAR-Ms can effectively target and destroy tumor cells, even within challenging microenvironments, by exhibiting direct cytotoxicity and enhancing the recruitment and activation of other immune cells. Additionally, the favorable safety profile of macrophages and their persistence within solid tumors position CAR-Ms as potentially safer and more durable therapeutic options compared to CAR-T cells. This review explores recent advancements in CAR-Ms technology, including engineering strategies to optimize their anti-tumor efficacy and preclinical evidence supporting their use. We also discuss the challenges and future directions in developing CAR-Ms therapies, emphasizing their potential to revolutionize cellular immunotherapy. By harnessing the unique properties of macrophages, CAR-Ms offer a groundbreaking approach to overcoming the current limitations of CAR-T cell therapies, paving the way for more effective and sustainable cancer treatments.
Collapse
Affiliation(s)
- Ting Huang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chenqi Bei
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhenhua Hu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
| | - Yuanyuan Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
40
|
Tian CF, Jing HY, Sinicrope FA, Wang JS, Gao BB, Sun XG, Yao ZG, Li LP, Saberzadeh-Ardestani B, Song W, Sha D. Tumor microenvironment characteristics association with clinical outcome in patients with resected intestinal-type gastric cancer. Oncologist 2024; 29:e1280-e1290. [PMID: 38907674 PMCID: PMC11448893 DOI: 10.1093/oncolo/oyae124] [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: 01/22/2024] [Accepted: 05/04/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND Tumor microenvironment (TME) characteristics including tumor stroma ratio (TSR), tumor budding (TB), and tumor-infiltrating lymphocytes (TILs) were examined in resected gastric cancer. These TME features have been shown to indicate metastatic potential in colon cancer, and intestinal-type gastric cancer (IGC) has pathological similarities with that malignancy. METHODS TSR, TB, and TILs were quantified in routine histological sections from 493 patients with IGC who underwent radical resection at 2 university hospitals in China from 2010 to 2016. TME variables were dichotomized as follows: TSR (50%), TILs (median), TB per international guidelines (4 buds/0.785mm2), and platelet-lymphocyte ratio (PLR) per survival ROC. Association of TME features with patient clinicopathological characteristics, time-to-recurrence (TTR), and cancer-specific-survival (CSS) were examined using univariate and multivariate analysis, including a relative contribution analysis by Cox regression. RESULTS Patients whose tumors showed high TSR or high TB or low TILs were each significantly associated with increased T and N stage, higher histological grade, and poorer TTR and CSS at 5 years. Only TSR and N stage were independently associated with TTR and CSS after adjustment for covariates. PLR was only independently associated with TTR after adjustment for covariates. Among the variables examined, only TSR was significantly associated with both TTR (HR 1.72, 95% CI, 1.14-2.60, P = .01) and CSS (HR 1.62, 95% CI, 1.05-2.51, P = .03) multivariately. Relative contribution to TTR revealed that the top 3 contributors were N stage (45.1%), TSR (22.5%), and PLR (12.9%), while the top 3 contributors to CSS were N stage (59.9%), TSR (14.7%), and PLR (10.9%). CONCLUSIONS Among the examined TME features, TSR was the most robust for prognostication and was significantly associated with both TTR and CSS. Furthermore, the relative contribution of TSR to patient TTR and CSS was second only to nodal status.
Collapse
Affiliation(s)
- Chun-Fang Tian
- Department of Minimally Invasive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Hai-Yan Jing
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Frank A Sinicrope
- Department of Oncology, Mayo Clinic, Rochester, 55905, United States
| | - Jin-Shen Wang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Bin-Bin Gao
- Department of Minimally Invasive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Xiao-Gang Sun
- Department of Minimally Invasive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Zhi-Gang Yao
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Le-Ping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | | | - Wei Song
- Department of Minimally Invasive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Dan Sha
- Department of Minimally Invasive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| |
Collapse
|
41
|
Um‐e‐Kalsoom, Wang S, Qu J, Liu L. Innovative optical imaging strategies for monitoring immunotherapy in the tumor microenvironments. Cancer Med 2024; 13:e70155. [PMID: 39387259 PMCID: PMC11465031 DOI: 10.1002/cam4.70155] [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: 01/13/2024] [Revised: 08/01/2024] [Accepted: 08/16/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND The tumor microenvironment (TME) plays a critical role in cancer progression and response to immunotherapy. Immunotherapy targeting the immune system has emerged as a promising treatment modality, but challenges in understanding the TME limit its efficacy. Optical imaging strategies offer noninvasive, real-time insights into the interactions between immune cells and the TME. OBJECTIVE This review assesses the progress of optical imaging technologies in monitoring immunotherapy within the TME and explores their potential applications in clinical trials and personalized cancer treatment. METHODS This is a comprehensive literature review based on the advances in optical imaging modalities including fluorescence imaging (FLI), bioluminescence imaging (BLI), and photoacoustic imaging (PAI). These modalities were analyzed for their capacity to provide high-resolution, real-time imaging of immune cell dynamics, tumor vasculature, and other critical components of the TME. RESULTS Optical imaging techniques have shown significant potential in tracking immune cell infiltration, assessing immune checkpoint inhibitors, and visualizing drug delivery within the TME. Technologies like FLI and BLI are pivotal in tracking immune responses in preclinical models, while PAI provides functional imaging with deeper tissue penetration. The integration of these modalities with immunotherapy holds promise for improving treatment monitoring and outcomes. CONCLUSION Optical imaging is a powerful tool for understanding the complexities of the TME and optimizing immunotherapy. Further advancements in imaging technologies, combined with nanomaterial-based approaches, could pave the way for enhanced diagnostic accuracy and therapeutic efficacy in cancer treatment.
Collapse
Affiliation(s)
- Um‐e‐Kalsoom
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Shiqi Wang
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Liwei Liu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| |
Collapse
|
42
|
He Y, Li G, Wu Y, Cai N, Chen Z, Mei B, Chen X, Zhang B, Jin G, Ding Z. Actin like 6A is a prognostic biomarker and associated with immune cell infiltration in cancers. Discov Oncol 2024; 15:503. [PMID: 39333441 PMCID: PMC11436596 DOI: 10.1007/s12672-024-01388-0] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024] Open
Abstract
PURPOSE To investigate the role of Actin like 6 A (ACTL6A) in cancer and explore the potential mechanism of its function. METHODS Differential expression of ACTL6A was analyzed using Oncomine and TIMER database. Then, we downloaded data sets from TCGA database. The correlation between ACTL6A expression and survival in pan-cancer were analyzed by "survival", "survminer" R package and PrognoScan database. STRING (v 11.0) and stringAPP for Cytoscape v3.7.2 were used to predict ACTL6A associated genes. Copy number and methylation alterations of ACTL6A were analyzed using cBioPortal and GSCALite. Transcription factors were downloaded from The Human Transcription Factors Database and analyzed using "limma" R package, JASPAR and PROMO database. Correlations analysis between ACTL6A and immune cells were performed using TIMER and GEPIA database. RESULTS In our studies, we found that ACTL6A was widely upregulated in cancers, which might be attributed to its gene amplifications. Moreover, ACTL6A might regulated by transcription factors (TFs), including E2F1, YY1, CDX2 and HOXD10. In addition, high ACTL6A expression was associated with poor prognosis in most cancers. Meanwhile, ACTL6A was associated with the infiltration of immune cells, especially in liver hepatocellular carcinoma and brain lower grade glioma. CONCLUSION Amplification of ACTL6A is correlated with poor prognosis and contribute to immune cells infiltration in LIHC and LGG, which may provide immune-related therapeutic targets to guide clinical strategies.
Collapse
Affiliation(s)
- Yi He
- Hepatic Surgery Center, Clinical Medicine Research Centre for Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jie Fang Avenue, Wuhan, 430030, China
| | - Ganxun Li
- Hepatic Surgery Center, Clinical Medicine Research Centre for Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jie Fang Avenue, Wuhan, 430030, China
| | - Yu Wu
- Hepatic Surgery Center, Clinical Medicine Research Centre for Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jie Fang Avenue, Wuhan, 430030, China
| | - Ning Cai
- Hepatic Surgery Center, Clinical Medicine Research Centre for Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jie Fang Avenue, Wuhan, 430030, China
| | - Zeyu Chen
- Hepatic Surgery Center, Clinical Medicine Research Centre for Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jie Fang Avenue, Wuhan, 430030, China
| | - Bin Mei
- Hepatic Surgery Center, Clinical Medicine Research Centre for Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jie Fang Avenue, Wuhan, 430030, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Clinical Medicine Research Centre for Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jie Fang Avenue, Wuhan, 430030, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Clinical Medicine Research Centre for Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jie Fang Avenue, Wuhan, 430030, China
| | - Guannan Jin
- Department of Internal Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jie Fang Avenue, Wuhan, 430000, China.
| | - Zeyang Ding
- Hepatic Surgery Center, Clinical Medicine Research Centre for Hepatic Surgery of Hubei Province, and Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jie Fang Avenue, Wuhan, 430030, China.
| |
Collapse
|
43
|
Wen J, Liu D, Zhu H, Shu K. Microenvironmental regulation of tumor-associated neutrophils in malignant glioma: from mechanism to therapy. J Neuroinflammation 2024; 21:226. [PMID: 39285276 PMCID: PMC11406851 DOI: 10.1186/s12974-024-03222-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024] Open
Abstract
Glioma is the most common primary intracranial tumor in adults, with high incidence, recurrence, and mortality rates. Tumor-associated neutrophils (TANs) are essential components of the tumor microenvironment (TME) in glioma and play a crucial role in glioma cell proliferation, invasion and proneural-mesenchymal transition. Besides the interactions between TANs and tumor cells, the multi-dimensional crosstalk between TANs and other components within TME have been reported to participate in glioma progression. More importantly, several therapies targeting TANs have been developed and relevant preclinical and clinical studies have been conducted in cancer therapy. In this review, we introduce the origin of TANs and the functions of TANs in malignant behaviors of glioma, highlighting the microenvironmental regulation of TANs. Moreover, we focus on summarizing the TANs-targeted methods in cancer therapy, aiming to provide insights into the mechanisms and therapeutic opportunities of TANs in the malignant glioma microenvironment.
Collapse
Affiliation(s)
- Jiayi Wen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Dan Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongtao Zhu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China.
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China.
| |
Collapse
|
44
|
Schmidt E, Distel L, Erber R, Büttner-Herold M, Rosahl MC, Ott OJ, Strnad V, Hack CC, Hartmann A, Hecht M, Fietkau R, Schnellhardt S. Tumor-Associated Neutrophils Are a Negative Prognostic Factor in Early Luminal Breast Cancers Lacking Immunosuppressive Macrophage Recruitment. Cancers (Basel) 2024; 16:3160. [PMID: 39335132 PMCID: PMC11430230 DOI: 10.3390/cancers16183160] [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: 08/19/2024] [Revised: 09/08/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Tumor-associated neutrophils (TANs) are important modulators of the tumor microenvironment with opposing functions that can promote and inhibit tumor progression. The prognostic role of TANs in early luminal breast cancer is unclear. METHODS A total of 144 patients were treated for early-stage hormone-receptor-positive breast cancer as part of an Accelerated Partial Breast Irradiation (APBI) phase II trial. Resection samples from multiple locations were processed into tissue microarrays and sections thereof immunohistochemically stained for CD66b+ neutrophils. CD66b+ neutrophil density was measured separately in the stromal and intraepithelial compartment. RESULTS High stromal and intraepithelial CD66b+ TAN density was a negative prognostic factor in central tumor samples. In addition, neutrophil density in adjacent normal breast tissue and lymph node samples also correlated with reduced disease-free survival. TAN density correlated with CD163+ M2-like tumor-associated macrophage (TAM) density, which we analyzed in a previous study. TANs were a negative prognostic factor in tumors with an elevated M1/M2 TAM ratio, while this impact on patient outcome was lost in tumors with a low M1/M2 ratio. A combined multivariate analysis of TAM and TAN density revealed that only TAM polarization status was an independent prognostic factor. CONCLUSIONS CD66b+ neutrophils were a negative prognostic factor in early-stage luminal breast cancer in single-marker analysis. Combined analysis with TAMs could be necessary to correctly evaluate their prognostic impact in future studies. TAN recruitment might act as a compensatory mechanism of immunoevasion and disease progression in tumors that are unable to sufficiently attract and polarize TAMs.
Collapse
Affiliation(s)
- Eva Schmidt
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (E.S.)
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
| | - Luitpold Distel
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (E.S.)
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
| | - Ramona Erber
- Institute of Pathology, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Maike Büttner-Herold
- Department of Nephropathology, Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Marie-Charlotte Rosahl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (E.S.)
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
| | - Oliver J. Ott
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (E.S.)
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
| | - Vratislav Strnad
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (E.S.)
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
| | - Carolin C. Hack
- Department of Gynecology and Obstetrics, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Markus Hecht
- Department of Radiotherapy and Radiation Oncology, Saarland University Medical Center, 66421 Homburg, Germany (S.S.)
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (E.S.)
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
| | - Sören Schnellhardt
- Department of Radiotherapy and Radiation Oncology, Saarland University Medical Center, 66421 Homburg, Germany (S.S.)
| |
Collapse
|
45
|
Quoniou R, Moreau E, Cachin F, Blavignac C, Bortoli E, Chautard E, Peyrode C. Chondrosarcoma Co-Culture 3D Model─An Insight to Evaluate Drugs Acting on TAMs. ACS Biomater Sci Eng 2024; 10:5832-5843. [PMID: 39121344 DOI: 10.1021/acsbiomaterials.4c00625] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2024]
Abstract
Chondrosarcoma (CHS), also known as malignant cartilage tumors, is the second most common bone cancer after osteosarcoma. This tumor is particularly chemo- and radioresistant, and the only therapeutic alternative is surgery with wide margins. The tumor immune microenvironment reveals an infiltration of tumor-associated macrophages (TAMs) sometimes approaching 50% of the tumor mass, mainly differentiated into M2-like phenotype and correlated with poor prognosis and metastasis. Thus, macrophage-targeting therapies could have an interest in the management of CHS. To evaluate these strategies, we propose here the development of a three-dimensional (3D) tumoroid co-culture model between two human CHS cell lines (JJ012 and CH2879) and a human leukemia monocytic cell line (THP-1) in a methylcellulose matrix. These two models were compared to the in vivo xenograft models in terms of macrophage phenotypes, proteoglycans, MMP-9, and COX-2 expression. Finally, mifamurtide, an immunomodulator acting on TAMs, was evaluated on the most in vitro relevant model: 3D co-culture CH2879 model. Our results showed that it is now possible to develop 3D models that very accurately mimic what is found in vivo with the possibility of evaluating treatments specific to a tumor cell component.
Collapse
Affiliation(s)
- Rohan Quoniou
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, 63000 Clermont-Ferrand, France
| | - Emmanuel Moreau
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, 63000 Clermont-Ferrand, France
| | - Florent Cachin
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, 63000 Clermont-Ferrand, France
- Service de Médecine Nucléaire, Centre Jean PERRIN, 63000 Clermont-Ferrand, France
| | | | - Elisa Bortoli
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, 63000 Clermont-Ferrand, France
| | - Emmanuel Chautard
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, 63000 Clermont-Ferrand, France
- Service de Pathologie, Centre Jean PERRIN, 63000 Clermont-Ferrand, France
| | - Caroline Peyrode
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, 63000 Clermont-Ferrand, France
| |
Collapse
|
46
|
Feng F, Shen J, Qi Q, Zhang Y, Ni S. Empowering brain tumor management: chimeric antigen receptor macrophage therapy. Theranostics 2024; 14:5725-5742. [PMID: 39310093 PMCID: PMC11413779 DOI: 10.7150/thno.98290] [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: 05/10/2024] [Accepted: 08/17/2024] [Indexed: 09/25/2024] Open
Abstract
Brain tumors pose formidable challenges in oncology due to the intricate biology and the scarcity of effective treatment modalities. The emergence of immunotherapy has opened new avenues for innovative therapeutic strategies. Chimeric antigen receptor, originally investigated in T cell-based therapy, has now expanded to encompass macrophages, presenting a compelling avenue for augmenting anti-tumor immune surveillance. This emerging frontier holds promise for advancing the repertoire of therapeutic options against brain tumors, offering potential breakthroughs in combating the formidable malignancies of the central nervous system. Tumor-associated macrophages constitute a substantial portion, ranging from 30% to 50%, of the tumor tissue and exhibit tumor-promoting phenotypes within the immune-compromised microenvironment. Constructing CAR-macrophages can effectively repolarize M2-type macrophages towards an M1-type phenotype, thereby eliciting potent anti-tumor effects. CAR-macrophages can recruit T cells to the brain tumor site, thereby orchestrating a remodeling of the immune niche to effectively inhibit tumor growth. In this review, we explore the potential limitations as well as strategies for optimizing CAR-M therapy, offering insights into the future direction of this innovative therapeutic approach.
Collapse
Affiliation(s)
| | | | - Qichao Qi
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Yulin Zhang
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Shilei Ni
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| |
Collapse
|
47
|
Cao W, Xie Y, Cai L, Wang M, Chen Z, Wang Z, Xv J, Wang Y, Li R, Liu X, Wang W. Pan‑cancer analysis on the role of KMT2C expression in tumor progression and immunotherapy. Oncol Lett 2024; 28:444. [PMID: 39091583 PMCID: PMC11292467 DOI: 10.3892/ol.2024.14577] [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: 12/04/2023] [Accepted: 05/10/2024] [Indexed: 08/04/2024] Open
Abstract
Histone lysine N-methyltransferase 2C (KMT2C) is involved in transcriptional regulation and DNA damage repair. Mutations in KMT2C have been implicated in the progression, metastasis, and drug resistance of multiple cancer types. However, the roles of KMT2C in the regulation of tumor prognosis, immune cell infiltration and the immune microenvironment in these multiple cancer types remain unclear. Therefore, in the present study, data from The Cancer Genome Atlas and Genotype-Tissue Expression databases were used for KMT2C expression analyses. Kaplan-Meier and univariate Cox regression analyses were also performed to investigate the prognostic role of KMT2C. In addition, Gene Set Enrichment Analysis (GSEA) was conducted to study the KMT2C-related signaling pathways. Tumor immune estimation resource 2 and single-sample GSEA were conducted to investigate the correlation between KMT2C expression and immune cell infiltrations, and Spearman's analysis was conducted to study the correlations among KMT2C, tumor mutational burden, microsatellite instability, immune regulators, chemokines and immune receptors. Immunohistochemistry of patient kidney tumor samples was performed to verify the correlation between KMT2C and programmed death-ligand 1 (PD-L1) expression. Finally, RNA interference, wound healing and colony formation assays were conducted to evaluate the effects of KMT2C expression on cell proliferation and metastasis. The results of the present study demonstrated that KMT2C was highly expressed in multiple cancer types, was a protective factor in kidney renal clear cell carcinoma and ovarian serous cystadenocarcinoma, and a risk factor for lung squamous cell carcinoma and uveal melanoma. In addition, KMT2C levels were negatively correlated with immune-activated pathways and the infiltration of immune cells, and positively correlated with inhibitory immune factors and tumor angiogenesis. Patients with low KMT2C expression had higher objective response rates to immunotherapy, and drug sensitivity analysis indicated that topoisomerase, histone deacetylase, DOT1-like histone H3K79 methyltransferase and G9A nuclear histone lysine methyltransferase inhibitors could potentially be used to treat tumors with high KMT2C expression levels. Finally, the KMT2C and PD-L1 expression levels were shown to be positively correlated, and KMT2C knockdown markedly promoted the proliferation and invasion capacities of A549 cells. In conclusion, the present study revealed that low KMT2C expression may be a promising biomarker for predicting the response of patients with cancer to immunotherapy. Conversely, high KMT2C expression was shown to promote tumor angiogenesis, which may contribute to the formation of the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Wei Cao
- Department of Thoracic Surgery, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Yawen Xie
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Li Cai
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Mengqing Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Zhuoying Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Ziteng Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Jiajia Xv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yuqing Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Rong Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Xuesong Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Wenliang Wang
- Institute of Clinical Immunology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
48
|
Kikuta S, Abe Y, Hino K, Imai S, Matsuo K, Shinozaki K, Nakamura M, Seki N, Kusukawa J. What prognostic factors have impacted the efficacy of immune checkpoint inhibitors in patients with recurrent or metastatic oral cancer? JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2024; 125:101961. [PMID: 38960025 DOI: 10.1016/j.jormas.2024.101961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are widely adapted for recurrent or metastatic head and neck cancer (RM-HNC), and various studies on its prognostic factors have been reported. We aimed to elucidate the prognostic factors of ICI treatment for RM oral cancer (RM-OC) in a retrospective study. METHODS We retrospectively reviewed patients with RM-OC treated with ICIs (nivolumab and pembrolizumab) at our department from May 2017 to February 2023. The objective response rate (ORR) for ICI treatment and the relationship between several potential prognostic factors, progression-free survival (PFS), and overall survival (OS) were analyzed statistically. RESULTS The investigation enrolled 31 patients, 16 with nivolumab and 15 with pembrolizumab. There were no significant differences in the ORR or disease control rate between the nivolumab and pembrolizumab groups (p = 0.4578 and 0.2524). In multivariate analysis, the prognostic nutritional index (PNI) and C-reactive protein to albumin ratio (CAR) exhibited statistical correlations with PFS, whereas the use of antibiotics and proton pump inhibitors (PPIs), neutrophil to lymphocyte ratio (NLR), and PNI demonstrated statistical associations with OS. CONCLUSION Our findings imply that the use of antibiotics and PPIs, which can modify the gut microbiota, may also serve as a prognostic determinant for ICI treatment in RM-OC, consistent with previous studies. Additionally, PNI may be essential in affecting the survival rates of both PFS and OS and could be an exceedingly valuable inflammatory biomarker for RM-OC.
Collapse
Affiliation(s)
- Shogo Kikuta
- Dental and Oral Medical Center, Kurume University School of Medicine, Fukuoka, Japan.
| | - Yushi Abe
- Dental and Oral Medical Center, Kurume University School of Medicine, Fukuoka, Japan
| | - Kiyosato Hino
- Dental and Oral Medical Center, Kurume University School of Medicine, Fukuoka, Japan
| | - Sho Imai
- Dental and Oral Medical Center, Kurume University School of Medicine, Fukuoka, Japan
| | - Katsuhisa Matsuo
- Dental and Oral Medical Center, Kurume University School of Medicine, Fukuoka, Japan; Department of Dentistry and Oral Surgery, Takagi Hospital, Fukuoka, Japan
| | - Katsumi Shinozaki
- Dental and Oral Medical Center, Kurume University School of Medicine, Fukuoka, Japan
| | - Moriyoshi Nakamura
- Dental and Oral Medical Center, Kurume University School of Medicine, Fukuoka, Japan
| | - Naoko Seki
- Dental and Oral Medical Center, Kurume University School of Medicine, Fukuoka, Japan
| | - Jingo Kusukawa
- Dental and Oral Medical Center, Kurume University School of Medicine, Fukuoka, Japan
| |
Collapse
|
49
|
Yu LC, Wang CA, Hu CY, Lin KC, Ou CH, Jan HC. Preoperative systemic inflammation response index enhances the prognostic value of tumor multifocalityin upper tract urothelial carcinoma. Oncol Lett 2024; 28:436. [PMID: 39081967 PMCID: PMC11287106 DOI: 10.3892/ol.2024.14569] [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: 12/14/2023] [Accepted: 05/20/2024] [Indexed: 08/02/2024] Open
Abstract
In cancer, tumor-related inflammation affects disease progression and survival outcomes. However, the role of systemic inflammation in tumor multifocality in upper tract urothelial carcinoma (UTUC) is not well understood. The aim of the present study was to evaluate the impact of the systemic inflammation response index (SIRI) on tumor multifocality for predicting oncological outcomes in patients with UTUC after radical nephroureterectomy (RNU). For this purpose, data from 645 patients with non-metastatic UTUC who underwent RNU between 2008 and 2020 were retrospectively analyzed. Survival outcomes such as overall survival (OS), cancer-specific survival (CSS) and recurrence-free survival (RFS) RATES were assessed using the Kaplan-Meier method, and independent prognostic factors were identified through a multivariable Cox proportional hazards regression model. Of the 645 patients with UTUC included in the present study, 163 (25%) had multifocal UTUC. Kaplan-Meier analysis indicated that multifocal UTUC synchronous with a high-level SIRI was significantly associated with poorer outcomes after RNU. Furthermore, the results of the multivariate Cox proportional hazards model analysis demonstrated that multifocal tumor coupled with a high-level SIRI was an independent factor for predicting a shorter survival and disease progression. In conclusion, the results of the present study indicated that an elevated SIRI significantly influenced the survival rate of patients with multifocal UTUC. Specifically, integrating multifocal UTUC with a high-level SIRI emerged as an independent risk factor for poorer OS, CSS and RFS. These findings highlighted the potential role of SIRI in the risk stratification and management of patients with multifocal UTUC.
Collapse
Affiliation(s)
- Lian-Ching Yu
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan, R.O.C
| | - Chu-An Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, R.O.C
| | - Che-Yuan Hu
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan, R.O.C
- Department of Urology, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan, R.O.C
| | - Kun-Che Lin
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan, R.O.C
| | - Chien-Hui Ou
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan, R.O.C
- Department of Urology, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan, R.O.C
| | - Hau-Chern Jan
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan, R.O.C
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan, R.O.C
- Division of Urology, Department of Surgery, National Cheng Kung University Hospital Dou-Liou Branch, Yunlin 64043, Taiwan, R.O.C
| |
Collapse
|
50
|
Zhang XC, Zhou YW, Wei GX, Luo YQ, Qiu M. Locoregional therapies combined with immune checkpoint inhibitors for liver metastases. Cancer Cell Int 2024; 24:302. [PMID: 39217341 PMCID: PMC11365172 DOI: 10.1186/s12935-024-03484-1] [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: 06/11/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have achieved remarkable success in clinical research and practice. Notably, liver metastasis is not sensitive to ICIs. Liver locoregional therapies can cause irreversible damage to tumor cells and release tumor antigens, thereby providing a rationale for immunotherapy treatments in liver metastasis. The combination therapy of ICIs with locoregional therapies is a promising option for patients with liver metastasis. Preclinical studies have demonstrated that combining ICIs with locoregional therapies produces a significantly synergistic anti-tumor effect. However, the current evidence for the efficacy of ICIs combined with locoregional therapies remains insufficient. Therefore, we review the literature on the mechanisms of locoregional therapies in treating liver metastasis and the clinical research progress of their combination with ICIs.
Collapse
Affiliation(s)
- Xing-Chen Zhang
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, 610041, Sichuan Province, China
| | - Yu-Wen Zhou
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, 610041, Sichuan Province, China
| | - Gui-Xia Wei
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yi-Qiao Luo
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Meng Qiu
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, 610041, Sichuan Province, China.
| |
Collapse
|