1
|
Liu ZH, Cai ZL, Tong XJ, Sun YY, Zhuang XY, Yang XF, Fan JKM. Modified scoring model incorporating waist-hip ratio for predicting advanced colorectal neoplasia. World J Clin Oncol 2025; 16:106409. [DOI: 10.5306/wjco.v16.i5.106409] [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: 02/25/2025] [Revised: 03/29/2025] [Accepted: 04/25/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Designing a feasible risk prediction model for advanced colorectal neoplasia (ACN) can enhance colonoscopy screening efficiency. Abdominal obesity is associated with colorectal cancer development.
AIM To propose and evaluate a modified scoring model incorporating waist-hip ratio for the prediction of ACN.
METHODS A total of 6483 patients who underwent their first screening or diagnostic colonoscopy in our center between 2020 and 2023 were recruited, in which 4592 were in the derivation cohort and 1891 formed a validation cohort. Multivariate logistic regression was used to investigate the risk factors of ACN in the derivation cohort based on endoscopic findings, and a new scoring model for ACN prediction was developed. The discriminatory capability of the scoring model was validated by the validation cohort.
RESULTS Age, male gender, smoking, and wait-to-hip ratio were identified as independent risk factors for ACN, and a 7-point scoring model was developed. The prevalence of ACN was 3.3%, 9.3% and 18.5% in participants with scores of 0-2 [low risk (LR)], 3–4 [moderate risk (MR)], and 5–7 [high risk (HR)], respectively, in the derivation cohort. With the scoring model, 49.9%, 38.4%, and 11.7% of patients in the validation cohort were categorized as LR, MR, and HR, respectively. The corresponding prevalence rates of ACN were 5.0%, 10.3%, and 17.6%, respectively. The C-statistic of the new scoring model was 0.66, which was higher than that of the Asia-Pacific Colorectal Screening model (0.63).
CONCLUSION A modified scoring model incorporating waist-hip ratio has an improved predictive performance in the prediction of ACN.
Collapse
Affiliation(s)
- Zhong-Hui Liu
- Department of Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518000, Guangdong Province, China
| | - Zong-Lin Cai
- Department of Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518000, Guangdong Province, China
| | - Xiao-Jun Tong
- Department of Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518000, Guangdong Province, China
| | - Yang-Yang Sun
- Endoscopy Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518000, Guangdong Province, China
| | - Xin-Yu Zhuang
- Department of Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518000, Guangdong Province, China
| | - Xue-Fei Yang
- Department of Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518000, Guangdong Province, China
| | - Joe KM Fan
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
2
|
Zhu X, Zhang L, Yu X, Yan P, Zhang X, Zhao Y, Wang D, Yang XA. Elucidating the tumor microenvironment interactions in breast, cervical, and ovarian cancer through single-cell RNA sequencing. Sci Rep 2025; 15:17846. [PMID: 40404741 PMCID: PMC12098903 DOI: 10.1038/s41598-025-03017-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 05/19/2025] [Indexed: 05/24/2025] Open
Abstract
This study aimed to identify the key cell types and their interactions in gynecological oncology of breast cancer, cervical cancer, and ovarian cancer. Single-cell RNA sequencing was performed on tumor samples of gynecological oncology from the GEO database. Cell types were identified using SingleR and cell composition was analyzed to understand the tumor microenvironment (TME). CellChat was used to analyze cell interactions, and pseudotemporal analysis was conducted on cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs) to understand their differentiation status. Four CAF subtypes were identified: iCAF, myCAF, proCAF, and matCAF. The iCAF subpopulation secreted COL1A1 and promoted tumor cell migration, while myCAF was involved in angiogenesis. The matCAF subpopulation was present throughout tumor development. TAMs were found to promote angiogenesis through the VEGFA_VEGFR2 signaling pathway. CAFs and TAMs play pivotal roles in tumor progression through their interactions and signaling pathways.
Collapse
Affiliation(s)
- Xiaoyue Zhu
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
- Graduate School of Chengde Medical University, Chengde, 067000, China
| | - Liang Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Xiaomin Yu
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
| | - Pengxian Yan
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
| | - Xiaoyu Zhang
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
- Graduate School of Chengde Medical University, Chengde, 067000, China
| | - Yunlong Zhao
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
- Graduate School of Chengde Medical University, Chengde, 067000, China
| | - Dongze Wang
- Clinical and Basic Medical College, Shandong First Medical University, Jinan, 250000, China
| | - Xiu-An Yang
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China.
- Hebei Key Laboratory of Nerve Injury and Repair, Chengde Medical University, Chengde, 067000, China.
| |
Collapse
|
3
|
Wei Y, Jia H, Guo X, Zhang H, Yang X, Can C, He N, Wu H, Liu W, Ma D. A novel LncRNA risk model for disulfidptosis-related prognosis prediction and response to chemotherapy in acute myeloid leukemia. Sci Rep 2025; 15:16995. [PMID: 40379722 PMCID: PMC12084325 DOI: 10.1038/s41598-025-01730-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/07/2025] [Indexed: 05/19/2025] Open
Abstract
Acute myeloid leukemia (AML), the most prevalent acute leukemia in adults, is characterized by its heterogeneity, which contributes to a poor prognosis and high recurrence rate. Recently, a unique form of cell death, called disulfidptosis, has been identified, which could transforming our understanding of and strategy for cancer treatment. Consequently, further inquiry is necessary to explore the possible link between disulfidptosis and AML. To facilitate this analysis, the researchers obtained single-cell RNA sequencing (scRNA-seq) data from AML patients using the Gene Expression Omnibus (GEO) database. By applying the Cox proportional hazards model and least absolute shrinkage and selection operator (LASSO) regression analysis, we created a signature of disulfidptosis-related long non-coding RNAs (DRLs). This predictive model was established based on six specific DRLs (AC005076.1, AP002807.1, HDAC4-AS1, L3MBTL4-AS1, LINC01694, and THAP9-AS1). The utility of this model in forecasting the prognosis of AML patients was corroborated by the receiver operating characteristic (ROC) curve. Moreover, significant variations in the biological functions and signaling pathways were discovered by gene ontology (GO) and Gene Set Enrichment Analysis (GSEA). To further investigate the relationship between immune infiltration, the study assessed variations in immune checkpoint expression and immune cell subset infiltration. Additionally, we used real-time quantitative PCR (RT-qPCR) to detect lncRNA expression in AML and healthy control to substantiate our analysis results. In conclusion, the results of this study may help discover novel therapeutic targets and prognostic biomarkers for AML, paving the way for customized precision chemotherapy.
Collapse
MESH Headings
- Humans
- RNA, Long Noncoding/genetics
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/mortality
- Prognosis
- Female
- Male
- ROC Curve
- Middle Aged
- Gene Expression Regulation, Leukemic
- Biomarkers, Tumor/genetics
- Proportional Hazards Models
- Disulfidptosis
Collapse
Affiliation(s)
- Yihong Wei
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Hexiao Jia
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Xiaodong Guo
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Hailei Zhang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Xinyu Yang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Can Can
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Na He
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Hanyang Wu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Wancheng Liu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Daoxin Ma
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
4
|
Malactou C, Hayes M, Zhang T, Ward S, Harwood C, Dalli J, Strid J. Eosinophils and Bioactive Lipid Mediators Regulate Skin Inflammation and Cancer Growth. J Invest Dermatol 2025:S0022-202X(25)00461-0. [PMID: 40320162 DOI: 10.1016/j.jid.2025.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/24/2025] [Accepted: 04/08/2025] [Indexed: 05/25/2025]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the second most common form of skin cancer, with local immunity playing a key role in regulating outcome. Nonresolving inflammation has been suggested to drive cancer growth; however, a fundamental understanding of the tumor immune microenvironment in cSCC is still lacking. In this study, we demonstrate tissue-associated tumor eosinophilia in both human and murine cSCC and show that eosinophils promote epithelial hyperplasia and tumor outgrowth. The skin eosinophils display tissue adaptation and dynamically alter their transcriptome according to the microenvironment. Furthermore, we show that the skin bioactive lipidome is significantly altered during inflammation and in cSCC and that this is regulated by eosinophils. Inflamed skin and cSCC tissue are dominated by lipid mediators synthesized by lipoxygenase enzymes, and our results suggest that eosinophils influence the balance between proinflammatory and proresolving lipid mediators. We demonstrate that lipid mediators can regulate epithelial cell expansion, with PCTR1 reducing inflammation-driven growth of tumor cells and eosinophil-driven eoxin C4 blocking this effect. Together, these findings highlight that eosinophils and specific bioactive lipid mediators play important roles in skin biology and carcinogenesis and point to potential previously unreported strategies for treatment of inflammatory disorders and epithelial malignancies.
Collapse
Affiliation(s)
- Christina Malactou
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Mark Hayes
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Tingfeng Zhang
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Sophie Ward
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Catherine Harwood
- Centre for Cell Biology and Cutaneous Research, Queen Mary University of London, London, United Kingdom
| | - Jesmond Dalli
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom; Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| | - Jessica Strid
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom.
| |
Collapse
|
5
|
Dora D, Revisnyei P, Pasic A, Galffy G, Dulka E, Mihucz A, Roskó B, Szincsak S, Iliuk A, Weiss GJ, Lohinai Z. Host and bacterial urine proteomics might predict treatment outcomes for immunotherapy in advanced non-small cell lung cancer patients. Front Immunol 2025; 16:1543817. [PMID: 40297587 PMCID: PMC12035445 DOI: 10.3389/fimmu.2025.1543817] [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: 12/11/2024] [Accepted: 03/05/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Urine samples are non-invasive approaches to study potential circulating biomarkers from the host organism. Specific proteins cross the bloodstream through the intestinal barrier and may also derive from gut microbiota. In this study, we aimed to evaluate the predictive role of the host and bacterial urine extracellular vesicle (EV) proteomes in patients with non-small cell lung cancer (NSCLC) treated with anti-PD1 immunotherapy. Methods We analyzed the urine EV proteome of 33 advanced-stage NSCLC patients treated with anti-PD1 immunotherapy with LC-MS/MS, stratifying patients according to long (>6 months) and short (≤6 months) progression-free survival (PFS). Gut microbial communities on a subcohort of 23 patients were also analyzed with shotgun metagenomics. Internal validation was performed using the Random Forest (RF) machine learning (ML) algorithm. RF was validated with a non-linear Bayesian ML model. Gene enrichment, and pathway analysis of host urine proteins were analyzed using the Reactome and Gene Ontology databases. Results We identified human (n=3513), bacterial (n=2647), fungal (n=19), and viral (n=4) proteins. 186 human proteins showed differential abundance (p<0.05) according to PFS groups, 101 being significantly more abundant in patients with short PFS and n=85 in patients with long PFS. We found several pathways that were significantly enriched in patients with short PFS (vs long PFS). Multivariate Cox regression showed that human urine proteins MPP5, IGKV6-21, NT5E, and KRT27 were strongly associated with long PFS, and LMAN2, NUTF2, NID1, TNC, IGF1, BCR, GPHN, and PPBP showed the strongest association with short PFS. We revealed that an increased bacterial/host protein ratio in the urine is more frequent in patients with long PFS. Increased abundance of E. coli and E. faecalis proteins in the urine positively correlates with their gut metagenomic abundance. RF ML model supported the reliability in predicting PFS for critical human urine proteins (AUC=0.89), accuracy (95%) and Bacterial proteins (AUC=0.74). Conclusion To our knowledge, this is the first study to depict the predictive role of the host and bacterial urine proteome in anti-PD1-treated advanced NSCLC.
Collapse
Affiliation(s)
- David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Peter Revisnyei
- Department of Telecommunications and Media Informatics, Budapest University of Technology and Economics, Budapest, Hungary
- HUN-REN-BME Information Systems Research Group, Budapest, Hungary
| | - Alija Pasic
- Department of Telecommunications and Media Informatics, Budapest University of Technology and Economics, Budapest, Hungary
| | | | - Edit Dulka
- County Hospital of Torokbalint, Torokbalint, Hungary
| | - Anna Mihucz
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Brigitta Roskó
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Sara Szincsak
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| | - Anton Iliuk
- Tymora Analytical Operations, West Lafayette, IN, United States
| | - Glen J. Weiss
- Department of Medicine, UMass Chan Medical School, Worcester, MA, United States
| | - Zoltan Lohinai
- County Hospital of Torokbalint, Torokbalint, Hungary
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| |
Collapse
|
6
|
Garlanda C, Dambra M, Magrini E. Interplay between the complement system and other immune pathways in the tumor microenvironment. Semin Immunol 2025; 78:101951. [PMID: 40209638 DOI: 10.1016/j.smim.2025.101951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025]
Abstract
Tumor growth and spread are sustained by the tumor microenvironment. Inflammatory cells and pathways have a fundamental role in the tumor microenvironment, driving or conditioning the functional activation of other leukocyte subsets and favoring evasion of anti-tumor immunity. One of the inflammatory pathways contributing to cancer-related inflammation is the complement system. Complement has long been considered an immune mechanism associated with immunosurveillance. More recently it emerged as a tumor promoting pathway, due to direct effects on cancer cells or indirect effects via immunosuppression driven by myeloid cells. The role of complement in cancer is complex and ambiguous, and depends on the tumor type and stage, as well as other factors including oncogenic drivers, leukocyte infiltration, interactions with other tumor microenvironment components or tumor cells. Other factors of complexity include the source of complement molecules, its canonical or non-canonical extracellular functions, its potential intracellular activation, and the interaction with other systems, such as the coagulation or the microbiome. Preclinical studies generally demonstrate the involvement of complement activation in smouldering inflammation in cancer and promotion of an immunosuppressive environment. These studies paved the way for clinical trials aimed at enhancing the potential of immunotherapy, in particular by targeting complement-dependent myeloid-sustained immunosuppression. However, the complex role of complement in cancer and the multiplicity of complement players may represent stumbling blocks and account for failures of clinical trials, and suggest that further studies are required to identify patient subsets who may benefit from specific complement molecule targeting in combination with conventional therapies or immunotherapy. Here, we will discuss the anti- or pro-tumor role of complement activation in cancer, focusing on the interactions of complement with immune cells within the tumor microenvironment, in particular the myeloid compartment. Furthermore, we will examine the potential of complement targeting in cancer treatment, particularly in the context of macrophage reprogramming.
Collapse
Affiliation(s)
- Cecilia Garlanda
- Department of Biomedical Sciences, Humanitas University, Milan, Pieve Emanuele 20072, Italy; IRCCS, Humanitas Research Hospital, Milan, Rozzano 20089, Italy.
| | - Monica Dambra
- IRCCS, Humanitas Research Hospital, Milan, Rozzano 20089, Italy
| | - Elena Magrini
- IRCCS, Humanitas Research Hospital, Milan, Rozzano 20089, Italy
| |
Collapse
|
7
|
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
|
8
|
Gwak HS, Choi BK, Lee YJ, Han NY, Yang KH. Long-Term Remission of Recurrent Anaplastic Oligodendroglioma With WT-1-Specific CD8+ T-Cell Therapy: A Case Report. Brain Tumor Res Treat 2025; 13:65-72. [PMID: 40347129 PMCID: PMC12070078 DOI: 10.14791/btrt.2025.0010] [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: 04/01/2025] [Revised: 04/06/2025] [Accepted: 04/08/2025] [Indexed: 05/12/2025] Open
Abstract
We report a case of complete remission in anaplastic oligodendroglioma following adoptive cell therapy (ACT) with autologous Wilms tumor 1 (WT-1)-specific CD8+ T cells. A 40-year-old woman referred to our hospital for adjuvant chemotherapy after recurrent anaplastic oligodendroglioma initially presented with a left frontal tumor, diagnosed through seizure onset, and subtotal resection confirmed oligodendroglioma (WHO grade 2). Radiation therapy treated the residual tumor, achieving partial remission until recurrence 2.5 years later when malignant transformation to anaplastic oligodendroglioma (WHO grade 3) occurred following a second craniotomy. After three cycles of procarbazine, lomustine, and vincristine chemotherapy, the residual tumor stabilized for 3 years. However, follow-up MRI identified a new enhancing lesion, prompting a third craniotomy. Recurrent anaplastic oligodendroglioma was confirmed, and adjuvant proton beam therapy and temozolomide chemotherapy were initiated. Two years later, another enhancing lesion appeared on the adjacent medial frontal lobe. Following multidisciplinary review, we introduced WT-1-specific ACT. Although transient swelling was observed 1 month post-therapy, the tumor demonstrated a response within 3-9 months. Continued regression led to complete remission-confirmed via MRI at the 15-month follow-up and sustained for 4.7 years. The patient's peripheral blood monocyte profiles and immune-associated cytokine analysis indicated T-cell activation following WT-1 sensitization.
Collapse
Affiliation(s)
- Ho-Shin Gwak
- Department of Cancer Biomedical Science, National Cancer Center, Graduate School of Cancer Science and Policy, Goyang, Korea.
| | - Beom Kyu Choi
- Biomedicine Production Branch, National Cancer Center, Goyang, Korea
| | - Young Joo Lee
- Center for Lung Cancer, National Cancer Center, Goyang, Korea
| | - Na Young Han
- Department of Pathology, National Cancer Center, Goyang, Korea
| | - Kook Hee Yang
- Department of Neurosurgery, National Health Insurance Service Ilsan Hospital, Goyang, Korea
| |
Collapse
|
9
|
Xiao Z, Puré E. The fibroinflammatory response in cancer. Nat Rev Cancer 2025:10.1038/s41568-025-00798-8. [PMID: 40097577 DOI: 10.1038/s41568-025-00798-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/06/2025] [Indexed: 03/19/2025]
Abstract
Fibroinflammation refers to the highly integrated fibrogenic and inflammatory responses mediated by the concerted function of fibroblasts and innate immune cells in response to tissue perturbation. This process underlies the desmoplastic remodelling of the tumour microenvironment and thus plays an important role in tumour initiation, growth and metastasis. More specifically, fibroinflammation alters the biochemical and biomechanical signalling in malignant cells to promote their proliferation and survival and further supports an immunosuppressive microenvironment by polarizing the immune status of tumours. Additionally, the presence of fibroinflammation is often associated with therapeutic resistance. As such, there is increasing interest in targeting this process to normalize the tumour microenvironment and thus enhance the treatment of solid tumours. Herein, we review advances made in unravelling the complexity of cancer-associated fibroinflammation that can inform the rational design of therapies targeting this.
Collapse
Affiliation(s)
- Zebin Xiao
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Ellen Puré
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
10
|
Liu X, Shao Y, Li Y, Chen Z, Shi T, Tong Q, Zou X, Ju L, Pan J, Zhuang R, Pan X. Extensive Review of Nanomedicine Strategies Targeting the Tumor Microenvironment in PDAC. Int J Nanomedicine 2025; 20:3379-3406. [PMID: 40125427 PMCID: PMC11927507 DOI: 10.2147/ijn.s504503] [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: 11/18/2024] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers in the world, mainly because of its powerful pro-connective tissue proliferation matrix and immunosuppressive tumor microenvironment (TME), which promote tumor progression and metastasis. In addition, the extracellular matrix leads to vascular collapse, increased interstitial fluid pressure, and obstruction of lymphatic return, thereby hindering effective drug delivery, deep penetration, and immune cell infiltration. Therefore, reshaping the TME to enhance tumor perfusion, increase deep drug penetration, and reverse immune suppression has become a key therapeutic strategy. Traditional therapies for PDAC, including surgery, radiation, and chemotherapy, face significant limitations. Surgery is challenging due to tumor location and growth, while chemotherapy and radiation are hindered by the dense extracellular matrix and immunosuppressive TME. In recent years, the advancement of nanotechnology has provided new opportunities to improve drug efficacy. Nanoscale drug delivery systems (NDDSs) provide several advantages, including improved drug stability in vivo, enhanced tumor penetration, and reduced systemic toxicity. However, the clinical translation of nanotechnology in PDAC therapy faces several challenges. These include the need for precise targeting and control over drug release, potential immune responses to the nanocarriers, and the scalability and cost-effectiveness of production. This article provides an overview of the latest nanobased methods for achieving better therapeutic outcomes and overcoming drug resistance. We pay special attention to TME-targeted therapy in the context of PDAC, discuss the advantages and limitations of current strategies, and emphasize promising new developments. By emphasizing the enormous potential of NDDSs in improving the treatment outcomes of patients with PDAC, while critically discussing the limitations of traditional therapies and the challenges faced by nanotechnology in achieving clinical breakthroughs, our review paves the way for future research in this rapidly developing field.
Collapse
Affiliation(s)
- Xing Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311400, People’s Republic of China
| | - Yidan Shao
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Yunjiang Li
- Radiology Department, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Zuhua Chen
- Radiology Department, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Tingting Shi
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Qiao Tong
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Xi Zou
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Liping Ju
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Jinming Pan
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Rangxiao Zhuang
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Xuwang Pan
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| |
Collapse
|
11
|
Mao J, Li L, Sun H, Han J, Li J, Dong CS, Zhao H. Investigation of sphingolipid-related genes in lung adenocarcinoma. Front Mol Biosci 2025; 12:1548655. [PMID: 40182622 PMCID: PMC11966433 DOI: 10.3389/fmolb.2025.1548655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Background Lung adenocarcinoma (LUAD) is responsible for majority cases of lung cancer and considered to be the primary cause of cancer-related mortality. The imbalance of cellular proliferation and apoptosis is critically implicated in the pathogenesis and progression of LUAD. Sphingomyelin, a vital lipid component, is integral to the regulation of tumor cell growth and apoptosis, and has garnered significant attention as a target in novel anticancer therapies. The pivotal molecules involved in sphingomyelin metabolism are crucial in modulating tumor cell behavior, thereby influencing clinical outcomes. Methods A comprehensive consensus clustering analysis was conducted by collecting clinical LUAD figures from the TCGA and GEO databases. By employing Cox regression and Lasso regression analysis, a prognostic model for LUAD patients was established by identifying seven sphingolipid-related genes (SRGs), and validated in the GEO database. The study also delved into the clinical relevance, functional capabilities, and immune implications of prognostic signals associated with sphingolipid metabolism. Finally, experiments conducted in vitro confirmed the imbalance of sphingolipid-associated genes in LUAD. Results Using the prognostic model, lung adenocarcinoma (LUAD) patients can be divided into high-risk and low-risk groups. Meanwhile, we can observe marked disparities in survival times among these groups. Additionally, the model demonstrates high predictive accuracy in external validation cohorts. Research on the immune microenvironment and immunotherapy points to this risk stratification as a useful reference for immunotherapeutic strategies in LUAD. Finally, our hypothesis was corroborated through in vitro experiments. Conclusion This study demonstrates that sphingolipid-related gene prognostic characteristics correlate with tumor progression and recurrence, long-term prognosis, and immune infiltration in LUAD patients. The outcomes of our study could help shape innovative strategies for early intervention and prognosis prediction in lung adenocarcinoma.
Collapse
Affiliation(s)
- Jibin Mao
- Department of Radiation Oncology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Li Li
- Department of Radiation Oncology, The Affiliated Hospital of Nantong University, Nantong, China
| | - Hui Sun
- Department of Pathology, The Affiliated Hospital of Nantong University, Nantong, China
| | - Jie Han
- Department of Radiation Oncology, The Affiliated Hospital of Nantong University, Nantong, China
| | - Jinqiao Li
- Department of Radiation Oncology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Chang-Sheng Dong
- Cancer Institute of Traditional Chinese Medicine/Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongyu Zhao
- Department of Radiation Oncology, The Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
12
|
Juthi RT, Sazed SA, Mareboina M, Zaravinos A, Georgakopoulos-Soares I. Characterization of Exhausted T Cell Signatures in Pan-Cancer Settings. Int J Mol Sci 2025; 26:2311. [PMID: 40076932 PMCID: PMC11899893 DOI: 10.3390/ijms26052311] [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: 01/07/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
T cells play diverse roles in cancer immunology, acting as tumor suppressors, cytotoxic effectors, enhancers of cytotoxic T lymphocyte responses and immune suppressors; providing memory and surveillance; modulating the tumor microenvironment (TME); or activating innate immune cells. However, cancer cells can disrupt T cell function, leading to T cell exhaustion and a weakened immune response against the tumor. The expression of exhausted T cell (Tex) markers plays a pivotal role in shaping the immune landscape of multiple cancers. Our aim was to systematically investigate the role of known T cell exhaustion (Tex) markers across multiple cancers while exploring their molecular interactions, mutation profiles, and potential implications for immunotherapy. The mRNA expression profile of six Tex markers, LAG-3, PDCD1, TIGIT, HAVCR2, CXCL13, and LAYN was investigated in pan-cancer. Utilizing data from The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), The Cancer Proteome Atlas (TCPA), and other repositories, we characterized the differential expression of the Tex markers, their association with the patients' survival outcome, and their mutation profile in multiple cancers. Additionally, we analyzed the effects on cancer-related pathways and immune infiltration within the TME, offering valuable insights into mechanisms of cancer immune evasion and progression. Finally, the correlation between their expression and sensitivity to multiple anti-cancer drugs was investigated extensively. Differential expression of all six markers was significantly associated with KIRC and poor prognosis in several cancers. They also played a potential activating role in apoptosis, EMT, and hormone ER pathways, as well as a potential inhibitory role in the DNA damage response and RTK oncogenic pathways. Infiltration of different immune cells was also found to be associated with the expression of the Tex-related genes in most cancer types. These findings underline that the reviving of exhausted T cells can be used to enhance the efficacy of immunotherapy in cancer patients.
Collapse
Affiliation(s)
- Rifat Tasnim Juthi
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh;
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (S.A.S.); (M.M.)
| | - Saiful Arefeen Sazed
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (S.A.S.); (M.M.)
| | - Manvita Mareboina
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (S.A.S.); (M.M.)
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, 22006, 1516 Nicosia, Cyprus
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), 22006, 1516 Nicosia, Cyprus
| | - Ilias Georgakopoulos-Soares
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (S.A.S.); (M.M.)
| |
Collapse
|
13
|
Xu Y, Kang K, Coakley BA, Eisenstein S, Parveen A, Mai S, Wang YS, Zheng J, Boral D, Mai J, Pan W, Zhang L, Aaronson SA, Fang B, Divino C, Zhang B, Song WM, Hung MC, Pan PY, Chen SH. Modulation of tumor inflammatory signaling and drug sensitivity by CMTM4. EMBO J 2025; 44:1866-1883. [PMID: 39948411 PMCID: PMC11914105 DOI: 10.1038/s44318-024-00330-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 03/19/2025] Open
Abstract
Although inflammation has been widely associated with cancer development, how it affects the outcomes of immunotherapy and chemotherapy remains incompletely understood. Here, we show that CKLF-like MARVEL transmembrane domain-containing member 4 (CMTM4) is highly expressed in multiple human and murine cancer types including Lewis lung carcinoma, triple-negative mammary cancer and melanoma. In lung carcinoma, loss of CMTM4 significantly reduces tumor growth and impairs NF-κB, mTOR, and PI3K/Akt pathway activation. Furthermore, we demonstrate that CMTM4 can regulate epidermal growth factor (EGF) signaling post-translationally by promoting EGFR recycling and preventing its Rab-dependent degradation. Consequently, CMTM4 knockout sensitizes human lung tumor cells to EGFR inhibitors. In addition, CMTM4 knockout tumors stimulated with EGF show a decreased ability to produce inflammatory cytokines including granulocyte colony-stimulating factor (G-CSF), leading to decreased recruitment of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) and therefore establishing a less suppressive tumor immune environment in both lung and mammary cancers. We also present evidence indicating that CMTM4-targeting siRNA-loaded liposomes reduce lung tumor growth in vivo and prolong animal survival. Knockout of CMTM4 enhances immune checkpoint blockade or chemotherapy to further reduce lung tumor growth. These data suggest that CMTM4 represents a novel target for the inhibition of tumor inflammation, and improvement of the immune response and tumor drug sensitivity.
Collapse
Affiliation(s)
- Yitian Xu
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Kyeongah Kang
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Brian A Coakley
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Samuel Eisenstein
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Arshiya Parveen
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Sunny Mai
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Yuan Shuo Wang
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Junjun Zheng
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Debasish Boral
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Junhua Mai
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - William Pan
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Licheng Zhang
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Stuart A Aaronson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bingliang Fang
- Department of Thoracic and Cardiovascular Surgery, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Celia Divino
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Won-Min Song
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Ping-Ying Pan
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Shu-Hsia Chen
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medical Science and Graduate School of Medical Sciences, New York, NY, 10065, USA.
- Graduate and professional school at Texas A&M University, 400 Bizzell St., College Station, TX, 77840, USA.
| |
Collapse
|
14
|
Yang Y, Liu T, Mi S, Liu X, Jabbour SK, Liang N, Deng G, Hu P, Zhang J. Radiotherapy as salvage therapy and an adjunct to immunotherapy: exploring local and abscopal mechanisms to overcome immunotherapy resistance: a narrative review. Transl Lung Cancer Res 2025; 14:591-606. [PMID: 40114936 PMCID: PMC11921301 DOI: 10.21037/tlcr-2025-57] [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: 02/17/2025] [Indexed: 03/22/2025]
Abstract
Background and Objective Immune checkpoint inhibitors (ICIs) have ushered in a new era of therapies and play a significant role in the clinical treatment of a variety of tumors. However, immune resistance has increasingly created a bottleneck in treatment, making the question of how to overcome drug resistance an urgent issue to address. In this article, the mechanism of drug resistance is briefly described with a focus on how radiotherapy (RT) acts on the immune system to reverse immunotherapy failure. Combinations of existing treatment modalities need to be optimized to overcome resistance problems. Research has shown that some RT modalities reverse immune resistance or enhance efficacy when used in combination, which shows some value for immune resistance and is worthy of in-depth research. Methods In this review, we searched the literature published from 2000 to 2023 surrounding immunotherapy, RT and cancer. Key Content and Findings Based on the immune effects and immunosuppressive effects induced by RT, this review examined the preclinical rationales of RT and its clinical results. The findings indicate that RT might provide a novel regimen for patients with locally advanced tumors, especially oligometastatic tumors. Conclusions Salvage therapy with RT after immunotherapy resistance is the focus of current research. Other strategies, such as multidrug combination therapies, have made preliminary progress in preclinical experiments. Further research on the roles of different RT doses, fractionation regimens, and other treatment sequences in salvage therapy need to be conducted in the future. The optimal site and timing of low-dose radiotherapy are also undetermined, and prospective studies are need to determine the best regimen for optimizing patient treatment.
Collapse
Affiliation(s)
- Yunxin Yang
- Department of Oncology, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| | - Tong Liu
- Department of Oncology, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| | - Song Mi
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
- Department of Oncology, Shandong University of Traditional Chinese Medicine, Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xin Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Ning Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| | - Guodong Deng
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| | - Pingping Hu
- Department of Oncology, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| | - Jiandong Zhang
- Department of Oncology, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| |
Collapse
|
15
|
Tan H, Cai M, Wang J, Yu T, Xia H, Zhao H, Zhang X. Harnessing Macrophages in Cancer Therapy: from Immune Modulators to Therapeutic Targets. Int J Biol Sci 2025; 21:2235-2257. [PMID: 40083710 PMCID: PMC11900799 DOI: 10.7150/ijbs.106275] [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: 11/03/2024] [Accepted: 02/14/2025] [Indexed: 03/16/2025] Open
Abstract
Macrophages, as the predominant phagocytes, play an essential role in pathogens defense and tissue homeostasis maintenance. In the context of cancer, tumor-associated macrophages (TAMs) have evolved into cunning actors involved in angiogenesis, cancer cell proliferation and metastasis, as well as the construction of immunosuppressive microenvironment. Once properly activated, macrophages can kill tumor cells directly through phagocytosis or attack tumor cells indirectly by stimulating innate and adaptive immunity. Thus, the prospect of targeting TAMs has sparked significant interest and emerged as a promising strategy in immunotherapy. In this review, we summarize the diverse roles and underlying mechanisms of TAMs in cancer development and immunity and highlight the TAM-based therapeutic strategies such as inhibiting macrophage recruitment, inhibiting the differentiation reprogramming of TAMs, blocking phagocytotic checkpoints, inducing trained macrophages, as well as the potential of engineered CAR-armed macrophages in cancer therapy.
Collapse
Affiliation(s)
- Huabing Tan
- Department of Infectious Diseases, Hepatology Institute, Renmin Hospital, Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, Hubei Province, China
- General internal medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Meihe Cai
- Department of Traditional Chinese Medicine, Zhushan Renmin Hospital, Zhushan, 442200, China
| | | | - Tao Yu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Houjun Xia
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Huanbin Zhao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Present: Division of Pharmaceutical Sciences, Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiaoyu Zhang
- Department of Gastrointestinal Surgery, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| |
Collapse
|
16
|
Zhou H, Zheng Z, Fan C, Zhou Z. Mechanisms and strategies of immunosenescence effects on non-small cell lung cancer (NSCLC) treatment: A comprehensive analysis and future directions. Semin Cancer Biol 2025; 109:44-66. [PMID: 39793777 DOI: 10.1016/j.semcancer.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/29/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
Non-small cell lung cancer (NSCLC), the most prevalent form of lung cancer, remains a leading cause of cancer-related mortality worldwide, particularly among elderly individuals. The phenomenon of immunosenescence, characterized by the progressive decline in immune cell functionality with aging, plays a pivotal role in NSCLC progression and contributes to the diminished efficacy of therapeutic interventions in older patients. Immunosenescence manifests through impaired immune surveillance, reduced cytotoxic responses, and increased chronic inflammation, collectively fostering a pro-tumorigenic microenvironment. This review provides a comprehensive analysis of the molecular, cellular, and genetic mechanisms of immunosenescence and its impact on immune surveillance and the tumor microenvironment (TME) in NSCLC. We explore how aging affects various immune cells, including T cells, B cells, NK cells, and macrophages, and how these changes compromise the immune system's ability to detect and eliminate tumor cells. Furthermore, we address the challenges posed by immunosenescence to current therapeutic strategies, particularly immunotherapy, which faces significant hurdles in elderly patients due to immune dysfunction. The review highlights emerging technologies, such as single-cell sequencing and CRISPR-Cas9, which offer new insights into immunosenescence and its potential as a therapeutic target. Finally, we outline future research directions, including strategies for rejuvenating the aging immune system and optimizing immunotherapy for older NSCLC patients, with the goal of improving treatment efficacy and survival outcomes. These efforts hold promise for the development of more effective, personalized therapies for elderly patients with NSCLC.
Collapse
Affiliation(s)
- Huatao Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China
| | - Zilong Zheng
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China.
| | - Zijing Zhou
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China.
| |
Collapse
|
17
|
Zhang R, Shi W, Wu X, Yu Q, Xiao Y. Application of hydrogen sulfide donor conjugates in different diseases. Nitric Oxide 2025; 154:128-139. [PMID: 39662602 DOI: 10.1016/j.niox.2024.11.008] [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: 05/07/2024] [Revised: 07/05/2024] [Accepted: 11/19/2024] [Indexed: 12/13/2024]
Abstract
As an endogenous gas signaling molecule, hydrogen sulfide (H2S) has been proved to have a variety of biological activities. Studies have shown that in some disease state H2S concentration in the body is lower than normal state. Based on these findings, exogenous H2S supplementation is expected to be an effective treatment for many diseases. In recent years, a lot of H2S-releasing substances, namely H2S donors, have emerged as H2S sources. Specifically, various H2S donors also could be connected to drugs or compounds to form H2S donor conjugates. Many studies have found that H2S donor conjugates can not only retain the activity of the parent drug, but also reduce the adverse effects of the parent drug, this makes H2S donor conjugates to be a new kind of drug candidates. In this article, H2S donor conjugates will be reviewed and classified according to different diseases, such as inflammation, cardiovascular and cerebrovascular diseases, diseases of central nervous system and cancer. This review aims to provide an idea for researchers for further study of H2S and H2S donor conjugates.
Collapse
Affiliation(s)
- Rui Zhang
- College of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Wumei Shi
- College of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaoyan Wu
- College of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Qingfeng Yu
- College of Science, China Pharmaceutical University, Nanjing, 211198, China.
| | - Ying Xiao
- College of Science, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
18
|
Yang XY, Chen N, Wen Q, Zhou Y, Zhang T, Zhou J, Liang CH, Han LP, Wang XY, Kang QM, Zheng XX, Zhai XJ, Jiang HY, Shen TH, Xiao JW, Zou YX, Deng Y, Lin S, Duan JJ, Wang J, Yu SC. The microenvironment cell index is a novel indicator for the prognosis and therapeutic regimen selection of cancers. J Transl Med 2025; 23:61. [PMID: 39806464 PMCID: PMC11727790 DOI: 10.1186/s12967-024-05950-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND It is worthwhile to establish a prognostic prediction model based on microenvironment cells (MCs) infiltration and explore new treatment strategies for triple-negative breast cancer (TNBC). METHODS The xCell algorithm was used to quantify the cellular components of the TNBC microenvironment based on bulk RNA sequencing (bulk RNA-seq) data. The MCs index (MCI) was constructed using the least absolute shrinkage and selection operator Cox (LASSO-Cox) regression analysis. Single-cell RNA sequencing (scRNA-seq), spatially resolved transcriptomics (SRT), and multiplex immunofluorescence (mIF) staining analyses verified MCI. The mechanism of action of the MCI was investigated in tumor-bearing mice. RESULTS MCI consists of the six types of MCs, which can precisely predict the prognosis of the TNBC patients. scRNA-seq, SRT, and mIF analyses verified the existence and proportions of these cells. Furthermore, combined with the spatial distribution characteristics of the six types of MCs, an MCI-enhanced (MCI-e) model was constructed, which could predict the prognosis of the TNBC patients more accurately. More importantly, inhibition of the insulin signaling pathway activated in the cancer cells of the MCIhigh the TNBC patients significantly prolonged the survival time of tumor-bearing mice. CONCLUSIONS Overall, our results demonstrate that MCs infiltration can be exploited as a novel indicator for the prognosis and therapeutic regimen selection of the TNBC patients.
Collapse
Affiliation(s)
- Xian-Yan Yang
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-Feng Laboratory, Chongqing, 401329, China
| | - Nian Chen
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Qian Wen
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Yu Zhou
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Tao Zhang
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Ji Zhou
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Cheng-Hui Liang
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Li-Ping Han
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Xiao-Ya Wang
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Qing-Mei Kang
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Xiao-Xia Zheng
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Xue-Jia Zhai
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Hong-Ying Jiang
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Tian-Hua Shen
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Jin-Wei Xiao
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Yu-Xin Zou
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Yun Deng
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Shuang Lin
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jiang-Jie Duan
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-Feng Laboratory, Chongqing, 401329, China
| | - Jun Wang
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China.
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China.
- Jin-Feng Laboratory, Chongqing, 401329, China.
| | - Shi-Cang Yu
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China.
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China.
- Jin-Feng Laboratory, Chongqing, 401329, China.
| |
Collapse
|
19
|
Lichtenstein AV. Rethinking the Evolutionary Origin, Function, and Treatment of Cancer. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:19-31. [PMID: 40058971 DOI: 10.1134/s0006297924603575] [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: 09/29/2024] [Revised: 09/29/2024] [Accepted: 12/08/2024] [Indexed: 05/13/2025]
Abstract
Despite remarkable progress in basic oncology, practical results remain unsatisfactory. This discrepancy is partly due to the exclusive focus on processes within the cancer cell, which results in a lack of recognition of cancer as a systemic disease. It is evident that a wise balance is needed between two alternative methodological approaches: reductionism, which would break down complex phenomena into smaller units to be studied separately, and holism, which emphasizes the study of complex systems as integrated wholes. A consistent holistic approach has so far led to the notion of cancer as a special organ, stimulating debate about its function and evolutionary significance. This article discusses the role of cancer as a mechanism of purifying selection of the gene pool, the correlation between hereditary and sporadic cancer, the cancer interactome, and the role of metastasis in a lethal outcome. It is also proposed that neutralizing the cancer interactome may be a novel treatment strategy.
Collapse
Affiliation(s)
- Anatoly V Lichtenstein
- N. N. Blokhin National Medical Research Centre of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia.
| |
Collapse
|
20
|
Huang Y, Chen Z, Shen G, Fang S, Zheng J, Chi Z, Zhang Y, Zou Y, Gan Q, Liao C, Yao Y, Kong J, Fan X. Immune regulation and the tumor microenvironment in anti-PD-1/PDL-1 and anti-CTLA-4 therapies for cancer immune evasion: A bibliometric analysis. Hum Vaccin Immunother 2024; 20:2318815. [PMID: 38419524 PMCID: PMC11789735 DOI: 10.1080/21645515.2024.2318815] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/26/2024] [Accepted: 02/11/2024] [Indexed: 03/02/2024] Open
Abstract
This study aims to conduct a bibliometric analysis, employing visualization tools to examine literature pertaining to tumor immune evasion related to anti-CTLA-4 and anti-PD-1/PD-L1 therapy from 1999 to 2022. A special emphasis is placed on the interplay between tumor microenvironment, signaling pathways, immune cells and immune evasion, with data sourced from the Web of Science core collection (WoSCC). Advanced tools, including VOSviewer, Citespace, and Scimago Graphica, were utilized to analyze various parameters, such as co-authorship/co-citation patterns, regional contributions, journal preferences, keyword co-occurrences, and significant citation bursts. Out of 4778 publications reviewed, there was a marked increase in research focusing on immune evasion, with bladder cancer being notably prominent. Geographically, China, the USA, and Japan were the leading contributors. Prestigious institutions like MD Anderson Cancer Center, Harvard Medical School, Fudan University, and Sun Yat Sen University emerged as major players. Renowned journals in this domain included Frontiers in Immunology, Cancers, and Frontiers in Oncology. Ehen LP and Wang W were identified as prolific authors on this topic, while Topalian SL stood out as one of the most cited. Research current situation is notably pivoting toward challenges like immunotherapy resistance and the intricate signaling pathways driving drug resistance. This bibliometric study seeks to provide a comprehensive overview of past and current research trends, emphasizing the potential role of tumor microenvironment, signaling pathways and immune cells in the context of immune checkpoint inhibitors (ICIs) and tumor immune evasion.
Collapse
Affiliation(s)
- Yi Huang
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Zhijian Chen
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Gang Shen
- Department of Urology, DUSHU Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Shuogui Fang
- Department of Radiotherapy, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
| | - Junjiong Zheng
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Zepai Chi
- Department of urology, Shantou Central Hospital, Shantou, China
| | - Yuanfeng Zhang
- Department of urology, Shantou Central Hospital, Shantou, China
| | - Yitong Zou
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Qinghua Gan
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Chengxiao Liao
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Yuhui Yao
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Jianqiu Kong
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Xinxiang Fan
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen University, Guangzhou, P. R. China
| |
Collapse
|
21
|
Quinlivan KM, Howard IV, Southan F, Bayer RL, Torres KL, Serhan CN, Panigrahy D. Exploring the unique role of specialized pro-resolving mediators in cancer therapeutics. Prostaglandins Other Lipid Mediat 2024; 178:106944. [PMID: 39722403 DOI: 10.1016/j.prostaglandins.2024.106944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/26/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Unresolved chronic inflammation, a hallmark of cancer, promotes tumor growth and metastasis in various cancer types. In contrast to blocking inflammation, stimulation of resolution of inflammation is an entirely novel approach to "resolve" inflammation. Resolution of inflammation mechanisms in cancer includes clearance of tumor debris, counter-regulation of pro-inflammatory eicosanoids and cytokines, and suppression of leukocyte infiltration. Conventional cytotoxic chemotherapy, radiation, anti-angiogenic therapy, and immune checkpoint inhibitors directly or indirectly can lead to the generation of pro-tumorigenic cellular debris. Over the past two decades, a potential paradigm shift has emerged in the inflammation field with the discovery of specialized pro-resolving mediators (SPMs), including resolvins, lipoxins, maresins, and protectins. SPMs are structurally distinct families of mediators grouped together by their pro-resolving "debris-clearing" functions. "Pro-resolving" therapies are in clinical development for various inflammation-driven diseases, including cancer. SPMs, as novel cancer therapeutics, have tremendous potential to enhance current cancer therapy. The mechanisms of SPMs as anti-cancer therapeutics are under active investigation by various laboratories worldwide. Here, we explore the current appreciation of the SPMs as innovative potential treatments designed to harness the unique anti-cancer activity of SPMs.
Collapse
Affiliation(s)
- Katherine M Quinlivan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States.
| | - Isabella V Howard
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Franciska Southan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Rachel L Bayer
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Kimberly L Torres
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Dipak Panigrahy
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| |
Collapse
|
22
|
Song Y, Zhang J, Li Y, Cheng L, Song H, Zhang Y, Du G, Yu S, Zou Y, Xu Q. Exploring Bioinformatics Tools to Analyze the Role of CDC6 in the Progression of Polycystic Ovary Syndrome to Endometrial Cancer by Promoting Immune Infiltration. Int J Mol Sci 2024; 25:12974. [PMID: 39684684 DOI: 10.3390/ijms252312974] [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: 11/01/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Cell division cycle 6 (CDC6) is essential for the initiation of DNA replication in eukaryotic cells and contributes to the development of various human tumors. Polycystic ovarian syndrome (PCOS) is a reproductive endocrine disease in women of childbearing age, with a significant risk of endometrial cancer (EC). However, the role of CDC6 in the progression of PCOS to EC is unclear. Therefore, we examined CDC6 expression in patients with PCOS and EC. We evaluated the relationship between CDC6 expression and its prognostic value, potential biological functions, and immune infiltrates in patients with EC. In vitro analyses were performed to investigate the effects of CDC6 knockdown on EC proliferation, migration, invasion, and apoptosis. CDC6 expression was significantly upregulated in patients with PCOS and EC. Moreover, this protein caused EC by promoting the aberrant infiltration of macrophages into the immune microenvironment in patients with PCOS. A functional enrichment analysis revealed that CDC6 exerted its pro-cancer and pro-immune cell infiltration functions via the PI3K-AKT pathway. Moreover, it promoted EC proliferation, migration, and invasion but inhibited apoptosis. This protein significantly reduced EC survival when mutated. These findings demonstrate that CDC6 regulates the progression of PCOS to EC and promotes immune infiltration.
Collapse
Affiliation(s)
- Yuhang Song
- School of Basic Medicine, Xinjiang Medical University, Urumqi 830054, China
- School of Clinical Medicine, Xinjiang Medical University, Urumqi 830054, China
| | - Jing Zhang
- Department of Immunology, School of Basic Medicine, Central South University, Changsha 410017, China
| | - Yao Li
- School of Basic Medicine, Xinjiang Medical University, Urumqi 830054, China
| | - Lufeng Cheng
- Basic Medical College, Xinjiang Medical University, Urumqi 830054, China
| | - Hua Song
- School of Clinical Medicine, Xinjiang Medical University, Urumqi 830054, China
| | - Yuhang Zhang
- School of Clinical Medicine, Xinjiang Medical University, Urumqi 830054, China
| | - Guoqing Du
- School of Basic Medicine, Xinjiang Medical University, Urumqi 830054, China
| | - Sunyue Yu
- School of Clinical Medicine, Xinjiang Medical University, Urumqi 830054, China
| | - Yizhou Zou
- Department of Immunology, School of Basic Medicine, Central South University, Changsha 410017, China
| | - Qi Xu
- School of Basic Medicine, Xinjiang Medical University, Urumqi 830054, China
| |
Collapse
|
23
|
Guo J, Lei T, Yu X, Wang P, Xie H, Jian G, Zhang Q, Qing Y. Analysis of the Potential Link Between Dermatomyositis and Cancer. J Inflamm Res 2024; 17:10163-10182. [PMID: 39649426 PMCID: PMC11624688 DOI: 10.2147/jir.s480744] [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: 07/26/2024] [Accepted: 11/27/2024] [Indexed: 12/10/2024] Open
Abstract
Background Dermatomyositis (DM) is an inflammatory muscle disease that increases the risk of cancer, although the precise connection is not fully understood. The aim of this study was to investigate the mechanisms linking DM to cancer and identify potential therapeutic targets. Methods We conducted differential gene expression analysis on the GSE128470 dataset and employed WGCNA to pinpoint key genes related to DM. Central genes were identified with the LASSO and SVM-RFE methods. The expression levels and diagnostic relevance of these genes were confirmed via the GSE1551 dataset. Immune cell infiltration was analyzed in relation to central genes, and RT‒qPCR was utilized to evaluate the expression of key genes across various cancers. Results In total, differentially expressed genes (DEGs), involved mainly in innate immunity, cytokine responses, and autoimmune diseases, were identified. In the WGCNA, 399 significant genes related to DM were identified, with central genes including MIF, C1QA, and CDKN1A. Immune infiltration analysis revealed diverse immune cell populations in DM patients, with significant correlations between central genes and these immune cells. MIF levels were notably elevated in various tumors and correlated with the prognosis of specific cancers. Furthermore, MIF was negatively associated with most immune cells but positively correlated with CD4+ Th1 cells, NKT cells, and MDSCs. Factors such as immune regulatory elements, TMB, and MSI indicated that MIF may affect immunotherapy outcomes. The increased expression of MIF mRNA was confirmed via RT‒qPCR. Conclusion The findings demonstrate that MIF, C1QA, and CDKN1A are differentially expressed in DM patients, with MIF showing significant alterations in DM patients with cancer. MIF may serve as a crucial prognostic biomarker and therapeutic target for various cancers, playing a pivotal role in linking DM to cancer through the modulation of CD4+ Th1 cells, NKT cells, and MDSCs.
Collapse
Affiliation(s)
- Jianwei Guo
- Research Center of Hyperuricemia and Gout, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
- Department of Geriatrics, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Tianyi Lei
- Research Center of Hyperuricemia and Gout, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Xiang Yu
- Research Center of Hyperuricemia and Gout, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Peng Wang
- Research Center of Hyperuricemia and Gout, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
- Department of Geriatrics, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Hongyuan Xie
- Research Center of Hyperuricemia and Gout, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Guilin Jian
- Research Center of Hyperuricemia and Gout, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
- Emergency Department,Suining Third People, S Hospital, Suining, Sichuan, 629000, People’s Republic of China
| | - Quanbo Zhang
- Research Center of Hyperuricemia and Gout, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
- Department of Geriatrics, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Yufeng Qing
- Research Center of Hyperuricemia and Gout, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| |
Collapse
|
24
|
Singh SS, Calvo R, Kumari A, Sable RV, Fang Y, Tao D, Hu X, Castle SG, Nahar S, Li D, Major E, Sanchez TW, Kato R, Xu X, Zhou J, Liu L, LeClair CA, Simeonov A, Baljinnyam B, Henderson MJ, Marugan J, Rudloff U. Fatty Acid Derivatization and Cyclization of the Immunomodulatory Peptide RP-182 Targeting CD206high Macrophages Improve Antitumor Activity. Mol Cancer Ther 2024; 23:1827-1841. [PMID: 39212669 PMCID: PMC11612619 DOI: 10.1158/1535-7163.mct-23-0790] [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: 11/22/2023] [Revised: 06/04/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
As tumor-associated macrophages (TAM) exercise a plethora of protumor and immune evasive functions, novel strategies targeting TAMs to inhibit tumor progression have emerged within the current arena of cancer immunotherapy. Activation of the mannose receptor 1 (CD206) is a recent approach that recognizes immunosuppressive CD206high M2-like TAMs as a drug target. Ligation of CD206 both induces reprogramming of CD206high TAMs toward a proinflammatory phenotype and selectively triggers apoptosis in these cells. CD206-activating therapeutics are currently limited to the linear, 10mer peptide RP-182, 1, which is not a drug candidate. In this study, we sought to identify a better suitable candidate for future clinical development by synthesizing and evaluating a series of RP-182 analogs. Surprisingly, fatty acid derivative 1a [RP-182-PEG3-K(palmitic acid)] not only showed improved stability but also increased affinity to the CD206 receptor through enhanced interaction with a hydrophobic binding motif of CD206. Peptide 1a showed superior in vitro activity in cell-based assays of macrophage activation which was restricted to CD206high M2-polarized macrophages. Improvement in responses was disproportionally skewed toward improved induction of phagocytosis including cancer cell phagocytosis. Peptide 1a reprogrammed the immune landscape in genetically engineered murine KPC pancreatic tumors toward increased innate immune surveillance and improved tumor control and effectively suppressed tumor growth of murine B16 melanoma allografts.
Collapse
Affiliation(s)
- Sitanshu S. Singh
- Rare Tumor Initiative, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Raul Calvo
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Anju Kumari
- Rare Tumor Initiative, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Rushikesh V. Sable
- Rare Tumor Initiative, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Yuhong Fang
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Dingyin Tao
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Xin Hu
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Sarah Gray Castle
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Saifun Nahar
- Rare Tumor Initiative, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Dandan Li
- Rare Tumor Initiative, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Emily Major
- Rare Tumor Initiative, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Tino W. Sanchez
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Rintaro Kato
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Xin Xu
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | | | - Liang Liu
- CPC Scientific Inc., San Jose, California
| | - Christopher A. LeClair
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Anton Simeonov
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Bolormaa Baljinnyam
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Mark J. Henderson
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Juan Marugan
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Udo Rudloff
- Rare Tumor Initiative, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
25
|
Garcia-Vicién G, Ruiz N, Micke P, Ruffinelli JC, Mils K, Bañuls M, Molina N, Pardo MA, Lladó L, Mezheyeuski A, Molleví DG. The histological growth patterns in liver metastases from colorectal cancer display differences in lymphoid, myeloid, and mesenchymal cells. MedComm (Beijing) 2024; 5:e70000. [PMID: 39563958 PMCID: PMC11574879 DOI: 10.1002/mco2.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 07/17/2024] [Accepted: 08/15/2024] [Indexed: 11/21/2024] Open
Abstract
Colorectal liver metastases grow following different histologic growth patterns (HGPs), classified as desmoplastic and nondesmoplastic (dHGP, non-dHGP), being the latter associated with worst prognosis. This study aimed to investigate the tumor microenvironment (TME) between HGPs supporting different survival. Multiplexed immunohistochemical staining was performed with the Opal7 system in a 100-patients cohort to evaluate the tumor-liver interface with three different cell panels: lymphoid, myeloid, and carcinoma-associated fibroblasts. Differences between HGPs were assessed by Mann-Whitney U test with Pratt correction and Holm-Bonferroni multitest adjustment. Cytotoxic T-cells were more abundant in tumoral areas of dHGP, while non-dHGP had higher macrophages infiltration, Th2, CD163+, and Calprotectin+ cells as well as higher pSMAD2 expression. Regarding carcinoma-associated fibroblasts, several subsets expressing COL1A1 were enriched in dHGP, while αSMAlow_single cells were present at higher densities in non-dHGP. Interestingly, Calprotectin+ cells confer better prognoses in non-dHGP, identifying a subgroup of good outcome patients that unexpectedly also show an enrichment in other myeloid cells. In summary, our results illustrate different TME landscapes with respect to HGPs. dHGP presents a higher degree of immunocompetence, higher amounts of Collagen 1 as well as lesser presence of myeloid cell populations, features that might be influencing on the better prognosis of encapsulated metastases.
Collapse
Affiliation(s)
- Gemma Garcia-Vicién
- Tumoural and Stromal Chemoresistance Group, Oncobell Program, IDIBELL L'Hospitalet de Llobregat Barcelona Catalonia Spain
| | - Núria Ruiz
- Tumoural and Stromal Chemoresistance Group, Oncobell Program, IDIBELL L'Hospitalet de Llobregat Barcelona Catalonia Spain
- Department of Pathology Hospital Universitari de Bellvitge L'Hospitalet de Llobregat Barcelona Catalonia Spain
| | - Patrick Micke
- Department of Immunology, Genetics and Pathology Uppsala University Uppsala Sweden
| | - José Carlos Ruffinelli
- Tumoural and Stromal Chemoresistance Group, Oncobell Program, IDIBELL L'Hospitalet de Llobregat Barcelona Catalonia Spain
- Department of Medical Oncology Institut Català d'Oncologia, L'Hospitalet de Llobregat Barcelona Catalonia Spain
| | - Kristel Mils
- Tumoural and Stromal Chemoresistance Group, Oncobell Program, IDIBELL L'Hospitalet de Llobregat Barcelona Catalonia Spain
- Department of Surgery Hospital Universitari de Bellvitge L'Hospitalet de Llobregat Barcelona Catalonia Spain
| | - María Bañuls
- Program Against Cancer Therapeutic Resistance (ProCURE) Institut Català d'Oncologia, L'Hospitalet de Llobregat Barcelona Catalonia Spain
| | - Natalia Molina
- Tumoural and Stromal Chemoresistance Group, Oncobell Program, IDIBELL L'Hospitalet de Llobregat Barcelona Catalonia Spain
| | - Miguel A Pardo
- Program Against Cancer Therapeutic Resistance (ProCURE) Institut Català d'Oncologia, L'Hospitalet de Llobregat Barcelona Catalonia Spain
| | - Laura Lladó
- Tumoural and Stromal Chemoresistance Group, Oncobell Program, IDIBELL L'Hospitalet de Llobregat Barcelona Catalonia Spain
- Department of Surgery Hospital Universitari de Bellvitge L'Hospitalet de Llobregat Barcelona Catalonia Spain
| | - Artur Mezheyeuski
- Department of Immunology, Genetics and Pathology Uppsala University Uppsala Sweden
- Molecular Oncology Group, Vall d'Hebron Institute of Oncology Barcelona Catalonia Spain
| | - David G Molleví
- Tumoural and Stromal Chemoresistance Group, Oncobell Program, IDIBELL L'Hospitalet de Llobregat Barcelona Catalonia Spain
- Program Against Cancer Therapeutic Resistance (ProCURE) Institut Català d'Oncologia, L'Hospitalet de Llobregat Barcelona Catalonia Spain
| |
Collapse
|
26
|
Yang J, Zhang K, Shi J, Li Z, Dai H, Yang W. Perfluoroalkyl and polyfluoroalkyl substances and Cancer risk: results from a dose-response Meta-analysis. JOURNAL OF ENVIRONMENTAL HEALTH SCIENCE & ENGINEERING 2024; 22:455-469. [PMID: 39464822 PMCID: PMC11499464 DOI: 10.1007/s40201-024-00899-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 03/06/2024] [Indexed: 10/29/2024]
Abstract
Background Perfluoroalkyl and polyfluoroalkyl substances (PFASs) are persistent organic pollutants in the environment. While some studies suggest that PFASs may contribute to cancer development, the link between PFAS exposure and cancer risk remains debated. Methods This dose-response meta-analysis explores the relationship between PFASs and cancer. It employs odds ratio (OR) and standardized mean difference (SMD), along with their 95% confidence interval (CI), to assess the effects of PFASs on cancer risk. Relevant studies were sourced from Web of Science, PubMed, Embase, Medline, and CNKI databases. The dose-response relationship was assessed by the fixed-effects model and least-squares regression. Results Forty studies, involving a total of 748,188 participants, were included in this meta-analysis. Out of these, 13 studies were specifically analyzed for the dose-response relationship. Findings revealed that exposure to PFASs, especially PFDA, significantly raises the risk of genitourinary cancers, and PFDA exposure shows a dose-dependent increase in overall and breast cancer risk. Additionally, PFOS exposure is associated with an increased cancer risk, and elevated PFOA levels were significantly observed in breast cancer patients. Conclusions The findings suggest that PFAS exposure is a potential cancer risk factor, with the carcinogenic potential of PFDA being dose-dependent. Supplementary Information The online version contains supplementary material available at 10.1007/s40201-024-00899-w.
Collapse
Affiliation(s)
- Jingxuan Yang
- Department of Physiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Kui Zhang
- Department of Forensic Pathology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Jingyi Shi
- Department of Physiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Zhuo Li
- Department of Forensic Pathology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Hao Dai
- Department of Forensic Pathology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Wenxing Yang
- Department of Physiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| |
Collapse
|
27
|
Kur IM, Weigert A. Phosphatidylserine externalization as immune checkpoint in cancer. Pflugers Arch 2024; 476:1789-1802. [PMID: 38573347 PMCID: PMC11582130 DOI: 10.1007/s00424-024-02948-7] [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: 01/18/2024] [Revised: 03/13/2024] [Accepted: 03/16/2024] [Indexed: 04/05/2024]
Abstract
Cancer is the second leading cause of mortality worldwide. Despite recent advances in cancer treatment including immunotherapy with immune checkpoint inhibitors, new unconventional biomarkers and targets for the detection, prognosis, and treatment of cancer are still in high demand. Tumor cells are characterized by mutations that allow their unlimited growth, program their local microenvironment to support tumor growth, and spread towards distant sites. While a major focus has been on altered tumor genomes and proteomes, crucial signaling molecules such as lipids have been underappreciated. One of these molecules is the membrane phospholipid phosphatidylserine (PS) that is usually found at cytosolic surfaces of cellular membranes but can be rapidly and massively shuttled to the extracellular leaflet of the plasma membrane during apoptosis to serve as a limiting factor for immune responses. These immunosuppressive interactions are exploited by tumor cells to evade the immune system. In this review, we describe mechanisms of immune regulation in tumors, discuss if PS may constitute an inhibitory immune checkpoint, and describe current and future strategies for targeting PS to reactivate the tumor-associated immune system.
Collapse
Affiliation(s)
- Ivan-Maximiliano Kur
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Andreas Weigert
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.
- Frankfurt Cancer Institute, Goethe-University Frankfurt, 60596, Frankfurt, Germany.
- German Cancer Consortium (DKTK), Partner Site, Frankfurt, Germany.
- Cardiopulmonary Institute (CPI), 60590, Frankfurt, Germany.
| |
Collapse
|
28
|
Torres MJ, Ríos JC, Valle A, Indo S, GV KB, López-Moncada F, Faúndez M, Castellón EA, Contreras HR. Alpha-Lipoic Acid-Mediated Inhibition of LTB 4 Synthesis Suppresses Epithelial-Mesenchymal Transition, Modulating Functional and Tumorigenic Capacities in Non-Small Cell Lung Cancer A549 Cells. CURRENT THERAPEUTIC RESEARCH 2024; 102:100765. [PMID: 39816494 PMCID: PMC11731977 DOI: 10.1016/j.curtheres.2024.100765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/22/2024] [Indexed: 01/18/2025]
Abstract
Background Leukotriene B4 (LTB4) plays a crucial role in carcinogenesis by inducing epithelial-mesenchymal transition (EMT), a process associated with tumor progression. The synthesis of LTB4 is mediated by leukotriene A4 hydrolase (LTA4H), and it binds to the receptors BLT1 and BLT2. Dysregulation in LTB4 production is linked to the development of various pathologies. Therefore, the identification or design of inhibitors of LTB4 synthesis or receptor antagonists represents an ongoing challenge. In this context, our laboratory previously demonstrated that alpha-lipoic acid (ALA) inhibits LTA4H. The objective of this study was to evaluate the effect of ALA on the expression of canonical EMT markers and the functional and tumorigenic capacities induced by LTB4 in A549 cells. Methods The expression of cPLA2, 5LOX, FLAP, LTA4H, BLT1, and LTB4 production in human adenocarcinomic alveolar basal epithelial A549 cells was assessed using Western blot, RT-qPCR, and ELISA, respectively. Subsequently, the expression of canonical EMT markers was evaluated by Western blot. Functional assays were performed to assess cell viability, proliferation, invasion, migration, and clonogenicity using MTT, Western blot, Transwell assays, and colony formation assays, respectively. Results were expressed as median with interquartile range (n≥3) and analyzed using the Kruskal-Wallis or Tukey multiple comparisons tests. Results A549 cells express key proteins involved in LTB4 synthesis and receptor binding, including LTA4H and BLT1, and ALA inhibits the production of LTB4. Additionally, LTA4H and BLT1 were detected in lung adenocarcinoma tissue samples. LTB4 was found to induce EMT, whereas ALA treatment enhanced the expression of epithelial markers and reduced the expression of mesenchymal markers. Furthermore, ALA treatment resulted in a decrease in LTB4 levels and attenuated the functional and tumorigenic capacities of A549 cells, including their viability, migration, invasion, and clonogenic potential. Conclusions These findings suggest that ALA may offer therapeutic potential in the context of lung cancer, as it could be integrated into conventional pharmacological therapies to enhance treatment efficacy and mitigate the adverse effects associated with chemotherapy. Further studies are warranted to confirm the clinical applicability of ALA as an adjunctive treatment in lung cancer.
Collapse
Affiliation(s)
- María José Torres
- Laboratorio de Farmacología y Toxicología Molecular. Escuela de Química y Farmacia. Facultad de Química y de Farmacia. Pontificia Universidad Católica de Chile, Santiago, Chile
- Programa de Farmacología y Toxicología. Facultad de Medicina. Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Carlos Ríos
- Programa de Farmacología y Toxicología. Facultad de Medicina. Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Laboratorios Clínicos. Facultad de Medicina. Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexandra Valle
- Laboratorio de Farmacología y Toxicología Molecular. Escuela de Química y Farmacia. Facultad de Química y de Farmacia. Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sebastián Indo
- Laboratorio de Oncología Celular y Molecular. Departamento de Oncología Básico-Clínica. Facultad de Medicina. Universidad de Chile, Santiago, Chile
- Departamento de Tecnología Médica. Facultad de Medicina. Universidad de Chile, Santiago, Chile
- Centro para la Prevención y el Control del Cáncer (CECAN), Santiago, Chile
| | - Kevin Brockway GV
- Laboratorio de Farmacología y Toxicología Molecular. Escuela de Química y Farmacia. Facultad de Química y de Farmacia. Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratorio de Oncología Celular y Molecular. Departamento de Oncología Básico-Clínica. Facultad de Medicina. Universidad de Chile, Santiago, Chile
| | | | - Mario Faúndez
- Laboratorio de Farmacología y Toxicología Molecular. Escuela de Química y Farmacia. Facultad de Química y de Farmacia. Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Enrique A. Castellón
- Laboratorio de Oncología Celular y Molecular. Departamento de Oncología Básico-Clínica. Facultad de Medicina. Universidad de Chile, Santiago, Chile
- Centro para la Prevención y el Control del Cáncer (CECAN), Santiago, Chile
| | - Héctor R. Contreras
- Laboratorio de Oncología Celular y Molecular. Departamento de Oncología Básico-Clínica. Facultad de Medicina. Universidad de Chile, Santiago, Chile
- Centro para la Prevención y el Control del Cáncer (CECAN), Santiago, Chile
| |
Collapse
|
29
|
Murrey MW, Ng IT, Pixley FJ. The role of macrophage migratory behavior in development, homeostasis and tumor invasion. Front Immunol 2024; 15:1480084. [PMID: 39588367 PMCID: PMC11586339 DOI: 10.3389/fimmu.2024.1480084] [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: 08/13/2024] [Accepted: 10/23/2024] [Indexed: 11/27/2024] Open
Abstract
Tumor-associated macrophages (TAMs) recapitulate the developmental and homeostatic behaviors of tissue resident macrophages (TRMs) to promote tumor growth, invasion and metastasis. TRMs arise in the embryo and colonize developing tissues, initially to guide tissue morphogenesis and then to form complex networks in adult tissues to constantly search for threats to homeostasis. The macrophage growth factor, colony-stimulating factor-1 (CSF-1), which is essential for TRM survival and differentiation, is also responsible for the development of the unique motility machinery of mature macrophages that underpins their ramified morphologies, migratory capacity and ability to degrade matrix. Two CSF-1-activated kinases, hematopoietic cell kinase and the p110δ catalytic isoform of phosphatidylinositol 3-kinase, regulate this machinery and selective inhibitors of these proteins completely block macrophage invasion. Considering tumors co-opt the invasive capacity of TAMs to promote their own invasion, these proteins are attractive targets for drug development to inhibit tumor progression to invasion and metastasis.
Collapse
Affiliation(s)
| | | | - Fiona J. Pixley
- Macrophage Biology and Cancer Laboratory, School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
30
|
Mantovani A, Marchesi F, Di Mitri D, Garlanda C. Macrophage diversity in cancer dissemination and metastasis. Cell Mol Immunol 2024; 21:1201-1214. [PMID: 39402303 PMCID: PMC11528009 DOI: 10.1038/s41423-024-01216-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/06/2024] [Indexed: 11/02/2024] Open
Abstract
Invasion and metastasis are hallmarks of cancer. In addition to the well-recognized hematogenous and lymphatic pathways of metastasis, cancer cell dissemination can occur via the transcoelomic and perineural routes, which are typical of ovarian and pancreatic cancer, respectively. Macrophages are a universal major component of the tumor microenvironment and, in established tumors, promote growth and dissemination to secondary sites. Here, we review the role of tumor-associated macrophages (TAMs) in cancer cell dissemination and metastasis, emphasizing the diversity of myeloid cells in different tissue contexts (lungs, liver, brain, bone, peritoneal cavity, nerves). The generally used models of lung metastasis fail to capture the diversity of pathways and tissue microenvironments. A better understanding of TAM diversity in different tissue contexts may pave the way for tailored diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Alberto Mantovani
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy.
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy.
- William Harvey Research Institute, Queen Mary University, London, UK.
| | - Federica Marchesi
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
- Department Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Diletta Di Mitri
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy
| | - Cecilia Garlanda
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy
| |
Collapse
|
31
|
Shen Q, Murakami K, Sotov V, Butler M, Ohashi PS, Reedijk M. Inhibition of Notch enhances efficacy of immune checkpoint blockade in triple-negative breast cancer. SCIENCE ADVANCES 2024; 10:eado8275. [PMID: 39475614 PMCID: PMC11524187 DOI: 10.1126/sciadv.ado8275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024]
Abstract
Aberrant Notch, which is a defining feature of triple-negative breast cancer (TNBC) cells, regulates intercellular communication in the tumor immune microenvironment (TIME). This includes tumor-associated macrophage (TAM) recruitment through Notch-dependent cytokine secretion, contributing to an immunosuppressive TIME. Despite the low response rate of TNBC to immune checkpoint blockade (ICB), here, we report that inhibition of Notch-driven cytokine-mediated programs reduces TAMs and induces responsiveness to sequentially delivered ICB. This is characterized by the emergence of GrB+ cytotoxic T lymphocytes (CTLs) in the primary tumor. A more impressive effect of sequential treatment is observed in the lung where TAM depletion and increased CTLs are accompanied by near-complete abolition of metastases. This is due to (i) therapeutic reduction in Notch-dependent, prometastatic circulating factors released by the primary tumor, and (ii) elevated PD ligand 1 (PD-L1) in lung metastases, rendering them profoundly sensitive to ICB. These findings highlight the potential of combination cytokine inhibition and ICB as an immunotherapeutic strategy in TNBC.
Collapse
Affiliation(s)
- Qiang Shen
- Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| | - Kiichi Murakami
- Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| | - Valentin Sotov
- Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| | - Marcus Butler
- Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, Division of Medical Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Pamela S. Ohashi
- Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
- Department of Immunology, University of Toronto, Medical Sciences Building, 1 King’s College Circle, Room 7205, Toronto, Ontario M5S 1A8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto Medical Discovery Tower, MaRS Centre, 101 College Street, Room 15-701, Toronto, Ontario M5G 2M9, Canada
| | - Michael Reedijk
- Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
- Department of Medical Biophysics, University of Toronto, Toronto Medical Discovery Tower, MaRS Centre, 101 College Street, Room 15-701, Toronto, Ontario M5G 2M9, Canada
- Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Suite 8-411, Toronto, Ontario M5G 2M9, Canada
| |
Collapse
|
32
|
Ji H, Lan Y, Xing P, Wang Z, Zhong X, Tang W, Wei Q, Chen H, Liu B, Guo H. IL-18, a therapeutic target for immunotherapy boosting, promotes temozolomide chemoresistance via the PI3K/AKT pathway in glioma. J Transl Med 2024; 22:951. [PMID: 39434175 PMCID: PMC11492732 DOI: 10.1186/s12967-024-05755-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024] Open
Abstract
Interleukin-18, a member of the interleukin - 1 family of cytokines, is upregulated in glioma. However, its effects on glioma remain unclear. This study aimed to explore the role and underlying mechanisms of interleukin-18 expression in glioma. Here, we demonstrated that interleukin-18 enhanced resistance to temozolomide by increasing proliferation and inhibiting apoptosis in cultured glioma cells. Further in vivo studies revealed that interleukin-18 promoted temozolomide resistance in BALB/c nude mice bearing tumor. Mechanical exploration indicated that interleukin-18 stimulation could activate the PI3K/AKT signaling pathway in glioma cells, and PI3K inhibition could reduce the temozolomide resistance promoted by interleukin-18. We found that interleukin-18 upregulated CD274 expression in glioma, revealing its potential effects on the microenvironment. Furthermore, we established a tumor xenograft model and explored the therapeutic efficacy of anti-interleukin-18 monoclonal antibody. Targeting interleukin-18 prolonged survival and attenuated CD274 expression in the mice bearing tumor. Combined treatment with anti-interleukin-18 and anti-PD-1 monoclonal antibody showed better efficacy in suppressing tumor growth than either treatment alone in mice bearing tumor. Collectively, these data present that interleukin-18 promotes temozolomide chemoresistance in glioma cells via PI3K/Akt activation and establishes an immunosuppressive milieu by modulating CD274. This study highlights the therapeutic value of interleukin-18 in glioma.
Collapse
Affiliation(s)
- Huangyi Ji
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Yufei Lan
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Pengpeng Xing
- ZhiXin High School, No. 152, ZhiXin South Road, Yuexiu District, Guangzhou, 510080, China
| | - Zhao Wang
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Xiangyang Zhong
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Wenhui Tang
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Quantang Wei
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Hongbin Chen
- The Second Clinical School, Southern Medical University, Guangzhou, 510515, China
| | - Boyang Liu
- Department of Neurosurgery, Department of Neuro-Oncological Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| | - Hongbo Guo
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
33
|
Wu B, Zhang B, Li B, Wu H, Jiang M. Cold and hot tumors: from molecular mechanisms to targeted therapy. Signal Transduct Target Ther 2024; 9:274. [PMID: 39420203 PMCID: PMC11491057 DOI: 10.1038/s41392-024-01979-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/20/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Immunotherapy has made significant strides in cancer treatment, particularly through immune checkpoint blockade (ICB), which has shown notable clinical benefits across various tumor types. Despite the transformative impact of ICB treatment in cancer therapy, only a minority of patients exhibit a positive response to it. In patients with solid tumors, those who respond well to ICB treatment typically demonstrate an active immune profile referred to as the "hot" (immune-inflamed) phenotype. On the other hand, non-responsive patients may exhibit a distinct "cold" (immune-desert) phenotype, differing from the features of "hot" tumors. Additionally, there is a more nuanced "excluded" immune phenotype, positioned between the "cold" and "hot" categories, known as the immune "excluded" type. Effective differentiation between "cold" and "hot" tumors, and understanding tumor intrinsic factors, immune characteristics, TME, and external factors are critical for predicting tumor response and treatment results. It is widely accepted that ICB therapy exerts a more profound effect on "hot" tumors, with limited efficacy against "cold" or "altered" tumors, necessitating combinations with other therapeutic modalities to enhance immune cell infiltration into tumor tissue and convert "cold" or "altered" tumors into "hot" ones. Therefore, aligning with the traits of "cold" and "hot" tumors, this review systematically delineates the respective immune characteristics, influencing factors, and extensively discusses varied treatment approaches and drug targets based on "cold" and "hot" tumors to assess clinical efficacy.
Collapse
Affiliation(s)
- Bo Wu
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bo Zhang
- Department of Youth League Committee, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bowen Li
- Department of Pancreatic and Gastrointestinal Surgery, Ningbo No. 2 Hospital, Ningbo, China
| | - Haoqi Wu
- Department of Gynaecology and Obstetrics, The Second Hospital of Dalian Medical University, Dalian, China
| | - Meixi Jiang
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
34
|
Zhao C, Ma M, Yang J, Ye Z, Ma P, Song D. "Hedgehog Ball"-Shaped Nanoprobes for Multimodal Detection and Imaging of Inflammatory Markers in Osteosarcoma Using Fluorescence and Electrochemiluminescence. Anal Chem 2024; 96:16053-16062. [PMID: 39316735 DOI: 10.1021/acs.analchem.4c03739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Inflammation can affect the progression of cancer at tumor sites, such as in osteosarcoma, by intensifying metastasis and complicating outcomes. The current diagnostic methods lack the specificity and sensitivity required for early and accurate detection, particularly in differentiating between inflammation-induced changes and tumor activities. To address this, a novel "hedgehog ball"-shaped nanoprobe, Fe3O4@Au-pep-CQDs, was developed and designed to enhance the detection of caspase-1, a key marker of inflammation. This magnetic nanoprobe facilitates simultaneous fluorescence (FL) and electrochemiluminescence (ECL) detection. Magnetic separation minimizes the quenching of nanoparticles in solution and eliminates the need for frequent electrode replacement in ECL tests, thereby simplifying diagnostic procedures. The experimental results showed that in the detection of caspase-1, the nanoprobe had a detection limit of 0.029 U/mL (FL) and 0.033 U/mL (ECL) and had a dynamic range of 0.05 to 1.0 U/mL. Additionally, the nanoprobe achieved high recovery rates of 94.36 to 102.44% (FL) and 94.36-100.12% (ECL) in spiked biological samples. Furthermore, the nanoprobe's capabilities were extended to in vivo bioimaging to provide direct, intuitive visualization of biological processes. These novel nanoprobes were able to significantly enhance the accurate detection of inflammation at tumor sites, thereby optimizing both diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Chen Zhao
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
| | - Mo Ma
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
- School of Pharmacy, Jilin University, Qianjin Street 2699, Changchun 130012, China
| | - Jukun Yang
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
| | - Zhuoxin Ye
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
| | - Pinyi Ma
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
| | - Daqian Song
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
| |
Collapse
|
35
|
Chun KS, Kim EH, Kim DH, Song NY, Kim W, Na HK, Surh YJ. Targeting cyclooxygenase-2 for chemoprevention of inflammation-associated intestinal carcinogenesis: An update. Biochem Pharmacol 2024; 228:116259. [PMID: 38705538 DOI: 10.1016/j.bcp.2024.116259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/18/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Mounting evidence from preclinical and clinical studies suggests that persistent inflammation functions as a driving force in the journey to cancer. Cyclooxygenase-2 (COX-2) is a key enzyme involved in inflammatory signaling. While being transiently upregulated upon inflammatory stimuli, COX-2 has been found to be consistently overexpressed in human colorectal cancer and several other malignancies. The association between chronic inflammation and cancer has been revisited: cancer can arise when inflammation fails to resolve. Besides its proinflammatory functions, COX-2 also catalyzes the production of pro-resolving as well as anti-inflammatory metabolites from polyunsaturated fatty acids. This may account for the side effects caused by long term use of some COX-2 inhibitory drugs during the cancer chemopreventive trials. This review summarizes the latest findings highlighting the dual functions of COX-2 in the context of its implications in the development, maintenance, and progression of cancer.
Collapse
Affiliation(s)
- Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 42601, Korea
| | - Eun-Hee Kim
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam 13488, South Korea
| | - Do-Hee Kim
- Department of Chemistry, College of Convergence and Integrated Science, Kyonggi University, Suwon, Gyeonggi-do 16227, South Korea
| | - Na-Young Song
- Department of Oral Biology, BK21 Four Project, Yonsei University College of Dentistry, Seoul 03722, South Korea
| | - Wonki Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Hye-Kyung Na
- Department of Food Science and Biotechnology, College of Knowledge-Based Services Engineering, Sungshin Women's University, Seoul 01133, South Korea
| | - Young-Joon Surh
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
36
|
Wang M, Min M, Mai J, Liu X. Systematic analysis of the expression profiles and prognostic significance of the MED gene family in renal clear cell carcinoma. Oncol Lett 2024; 28:398. [PMID: 38979551 PMCID: PMC11228927 DOI: 10.3892/ol.2024.14531] [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: 01/11/2024] [Accepted: 04/23/2024] [Indexed: 07/10/2024] Open
Abstract
The mediator complex (MED) family is a contributing factor in the regulation of transcription and proliferation of cells, and is closely associated with the development of various types of cancer. However, the significance of the expression levels and prognostic value of MED genes in kidney renal clear cell carcinoma (KIRC) have rarely been reported. The present study analyzed the expression and prognostic potential of MED genes in KIRC. The Search Tool for the Retrieval of Interacting Genes/Proteins was used to construct the protein-protein interaction network (PPI), the Assistant for Clinical Bioinformatics database was used to perform correlation analysis, GEPIA 2 was utilized to draw the Kaplan-Meier plot and analyze prognostic significance and the Tumor Immune Estimation Resource was used to assess the association of MED genes with the infiltration of immune cells in patients with KIRC. A total of 30 MED genes were identified, and among these genes, 11 were selected for the creation of a prognostic gene signature based on the results of a LASSO Cox regression analysis. Furthermore, according to univariate and multivariate analyses, MED7, MED16, MED21, MED25 and MED29 may be valuable independent predictive biomarkers for the prognosis of individuals with KIRC. Furthermore, there were significant differences in the expression levels of MED7, MED21 and MED25 in KIRC among different tumor grades. Additionally, patients with KIRC with high transcription levels of MED7, MED21 and MED29 had considerably longer overall survival times. The expression levels of MED genes were also linked to the infiltration of several immune cells. Overall, MED genes may have potential significance in predicting the prognosis of patients with KIRC.
Collapse
Affiliation(s)
- Min Wang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Min Min
- Outpatient Department, The Air Force Hospital of Western Theater, People's Liberation Army, Chengdu, Sichuan 500643, P.R. China
| | - Jia Mai
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Xiaojuan Liu
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
37
|
Xia Y, Wang X, Lin J, Li Y, Dong L, Liang X, Wang HY, Ding X, Wang Q. Gastric cancer fibroblasts affect the effect of immunotherapy and patient prognosis by inducing micro-vascular production. Front Immunol 2024; 15:1375013. [PMID: 39040110 PMCID: PMC11260615 DOI: 10.3389/fimmu.2024.1375013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/28/2024] [Indexed: 07/24/2024] Open
Abstract
Introduction Immunotherapy is critical for treating many cancers, and its therapeutic success is linked to the tumor microenvironment. Although anti-angiogenic drugs are used to treat gastric cancer (GC), their efficacy remains limited. Cancer-associated fibroblast (CAF)-targeted therapies complement immunotherapy; however, the lack of CAF-specific markers poses a challenge. Therefore, we developed a CAF angiogenesis prognostic score (CAPS) system to evaluate prognosis and immunotherapy response in patients with GC, aiming to improve patient stratification and treatment efficacy. Methods We assessed patient-derived GC CAFs for promoting angiogenesis using EdU, cell cycle, apoptosis, wound healing, and angiogenesis analysis. Results We then identified CAF-angiogenesis-associated differentially-expressed genes, leading to the development of CAPS, which included THBS1, SPARC, EDNRA, and VCAN. We used RT-qPCR to conduct gene-level validation, and eight GEO datasets and the HPA database to validate the CAPS system at the gene and protein levels. Six independent GEO datasets were utilized for validation. Overall survival time was shorter in the high- than the low-CAPS group. Immune microenvironment and immunotherapy response analysis showed that the high-CAPS group had a greater tendency toward immune escape and reduced immunotherapy efficacy than the low-CAPS group. Discussion CAPS is closely associated with GC prognosis and immunotherapy outcomes. It is therefore an independent predictor of GC prognosis and immunotherapy efficacy.
Collapse
Affiliation(s)
- Yan Xia
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaolu Wang
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Lin
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuan Li
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Lidan Dong
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xue Liang
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Huai-Yu Wang
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xia Ding
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qi Wang
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
38
|
Tsutsumi E, Macy AM, LoBello J, Hastings KT, Kim S. Tumor immune microenvironment permissive to metastatic progression of ING4-deficient breast cancer. PLoS One 2024; 19:e0304194. [PMID: 38968186 PMCID: PMC11226078 DOI: 10.1371/journal.pone.0304194] [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: 05/06/2024] [Accepted: 06/20/2024] [Indexed: 07/07/2024] Open
Abstract
Deficiencies in the ING4 tumor suppressor are associated with advanced stage tumors and poor patient survival in cancer. ING4 was shown to inhibit NF-kB in several cancers. As NF-kB is a key mediator of immune response, the ING4/NF-kB axis is likely to manifest in tumor-immune modulation but has not been investigated. To characterize the tumor immune microenvironment associated with ING4-deficient tumors, three approaches were employed in this study: First, tissue microarrays composed of 246 primary breast tumors including 97 ING4-deficient tumors were evaluated for the presence of selective immune markers, CD68, CD4, CD8, and PD-1, using immunohistochemical staining. Second, an immune-competent mouse model of ING4-deficient breast cancer was devised utilizing CRISPR-mediated deletion of Ing4 in a Tp53 deletion-derived mammary tumor cell line; mammary tumors were evaluated for immune markers using flow cytometry. Lastly, the METABRIC gene expression dataset was evaluated for patient survival related to the immune markers associated with Ing4-deleted tumors. The results showed that CD68, CD4, CD8, or PD-1, was not significantly associated with ING4-deficient breast tumors, indicating no enrichment of macrophages, T cells, or exhausted T cell types. In mice, Ing4-deleted mammary tumors had a growth rate comparable to Ing4-intact tumors but showed increased tumor penetrance and metastasis. Immune marker analyses of Ing4-deleted tumors revealed a significant increase in tumor-associated macrophages (Gr-1loCD11b+F4/80+) and a decrease in granzyme B-positive (GzmB+) CD4+ T cells, indicating a suppressive and/or less tumoricidal immune microenvironment. The METABRIC data analyses showed that low expression of GZMB was significantly associated with poor patient survival, as was ING4-low expression, in the basal subtype of breast cancer. Patients with GZMB-low/ING4-low tumors had the worst survival outcomes (HR = 2.80, 95% CI 1.36-5.75, p = 0.0004), supportive of the idea that the GZMB-low immune environment contributes to ING4-deficient tumor progression. Collectively, the study results demonstrate that ING4-deficient tumors harbor a microenvironment that contributes to immune evasion and metastasis.
Collapse
Affiliation(s)
- Emily Tsutsumi
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, United States of America
- Cancer and Cell Biology Division, Translational Genomic Research Institute, Phoenix, Arizona, United States of America
| | - Anne M. Macy
- Department of Dermatology, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, United States of America
- Phoenix Veterans Affairs Health Care System, Phoenix, Arizona, United States of America
| | - Janine LoBello
- Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Karen T. Hastings
- Department of Dermatology, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, United States of America
- Phoenix Veterans Affairs Health Care System, Phoenix, Arizona, United States of America
| | - Suwon Kim
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, United States of America
- Cancer and Cell Biology Division, Translational Genomic Research Institute, Phoenix, Arizona, United States of America
| |
Collapse
|
39
|
Dang Q, Zuo L, Hu X, Zhou Z, Chen S, Liu S, Ba Y, Zuo A, Xu H, Weng S, Zhang Y, Luo P, Cheng Q, Liu Z, Han X. Molecular subtypes of colorectal cancer in the era of precision oncotherapy: Current inspirations and future challenges. Cancer Med 2024; 13:e70041. [PMID: 39054866 PMCID: PMC11272957 DOI: 10.1002/cam4.70041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/07/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is among the most hackneyed malignancies. Even patients with identical clinical symptoms and the same TNM stage still exhibit radically different clinical outcomes after receiving equivalent treatment regimens, indicating extensive heterogeneity of CRC. Myriad molecular subtypes of CRC have been exploited for decades, including the most compelling consensus molecular subtype (CMS) classification that has been broadly applied for patient stratification and biomarker-drug combination formulation. Encountering barriers to clinical translation, however, CMS classification fails to fully reflect inter- or intra-tumor heterogeneity of CRC. As a consequence, addressing heterogeneity and precisely managing CRC patients with unique characteristics remain arduous tasks for clinicians. REVIEW In this review, we systematically summarize molecular subtypes of CRC and further elaborate on their clinical applications, limitations, and future orientations. CONCLUSION In recent years, exploration of subtypes through cell lines, animal models, patient-derived xenografts (PDXs), organoids, and clinical trials contributes to refining biological insights and unraveling subtype-specific therapies in CRC. Therapeutic interventions including nanotechnology, clustered regulatory interspaced short palindromic repeat/CRISPR-associated nuclease 9 (CRISPR/Cas9), gut microbiome, and liquid biopsy are powerful tools with the possibility to shift the immunologic landscape and outlook for CRC precise medicine.
Collapse
Affiliation(s)
- Qin Dang
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Lulu Zuo
- Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xinru Hu
- Department of Cardiology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Zhaokai Zhou
- Department of UrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Shuang Chen
- Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Shutong Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yuhao Ba
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Anning Zuo
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Hui Xu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Siyuan Weng
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yuyuan Zhang
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Peng Luo
- Department of Oncology, Zhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Quan Cheng
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Zaoqu Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanChina
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanChina
- Institute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xinwei Han
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanChina
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
40
|
Mahmood K, Wang H, Ji Z, Giovacchini CX, Wahidi MM, Dorry M, Shofer SL, Clarke JM, Antonia SJ, Shaz BH, Steadman K, Weinhold KJ, Yi J. Differences in microenvironment of lung cancer and pleural effusions by single-cell RNA sequencing. Lung Cancer 2024; 193:107847. [PMID: 38889499 DOI: 10.1016/j.lungcan.2024.107847] [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: 03/31/2024] [Revised: 05/20/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Direct comparison of tumor microenvironment of matched lung cancer biopsies and pleural effusions (PE) from the same patients is critical in understanding tumor biology but has not been performed. This is the first study to compare the lung cancer and PE microenvironment by single-cell RNA sequencing (scRNA-seq). METHODS Matched lung cancer biopsies and PE were obtained prospectively from ten patients. We isolated CD45+ cells and performed scRNA-seq to compare the biopsies and PE. RESULTS PE had a higher proportion of CD4+ T cells but lower proportion of CD8+ T cells (False detection rate, FDR = 0.0003) compared to biopsies. There was a higher proportion of naïve CD4+ T cells (FDR = 0.04) and naïve CD8+ T cells (FDR = 0.0008) in PE vs. biopsies. On the other hand, there was a higher proportion of Tregs (FDR = 0.04), effector CD8+ (FDR = 0.006), and exhausted CD8+ T cells (FDR = 0.01) in biopsies. The expression of inflammatory genes in T cells was increased in biopsies vs. PE, including TNF, IFN-ɣ, IL-1R1, IL-1R2, IL-2, IL-12RB2, IL-18R1, and IL-18RAP (FDR = 0.009, 0.013, 0.029, 0.043, 0.009, 0.013, 0.004, and 0.003, respectively). The gene expression of exhaustion markers in T cells was also increased in tumor biopsies including PDCD1, CTLA4, LAG 3, HAVCR2, TIGIT, and CD160 (FDR = 0.008, 0.003, 0.002, 0.011, 0.006, and 0.049, respectively). CONCLUSIONS There is a higher proportion of naïve T cells and lower proportion of exhausted T cells and Tregs in PE compared to lung cancer biopsies, which can be leveraged for prognostic and therapeutic applications.
Collapse
Affiliation(s)
- Kamran Mahmood
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, Duke University, Durham, NC, USA.
| | - Huimin Wang
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Zhicheng Ji
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Coral X Giovacchini
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, Duke University, Durham, NC, USA
| | - Momen M Wahidi
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, Duke University, Durham, NC, USA
| | - Michael Dorry
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, Duke University, Durham, NC, USA
| | - Scott L Shofer
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, Duke University, Durham, NC, USA
| | - Jeffrey M Clarke
- Department of Medicine, Division of Medical Oncology, Duke University, Durham, NC, USA
| | - Scott J Antonia
- Department of Medicine, Division of Medical Oncology, Duke University, Durham, NC, USA
| | - Beth H Shaz
- Department of Pathology, Duke University, Durham, NC, USA
| | - Katelyn Steadman
- Duke Immune Profiling Core, Department of Surgery, Duke University, Durham, NC, USA
| | - Kent J Weinhold
- Duke Immune Profiling Core, Department of Surgery, Duke University, Durham, NC, USA
| | - John Yi
- Duke Immune Profiling Core, Department of Surgery, Duke University, Durham, NC, USA
| |
Collapse
|
41
|
Li Y, Li H, Sun G, Xu S, Tang X, Zhang L, Wan L, Zhang L, Tang M. Integrative analyses of multi-omics data constructing tumor microenvironment and immune-related molecular prognosis model in human colorectal cancer. Heliyon 2024; 10:e32744. [PMID: 38975206 PMCID: PMC11226854 DOI: 10.1016/j.heliyon.2024.e32744] [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: 09/18/2023] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 07/09/2024] Open
Abstract
The increasing prevalence and incidence of colorectal cancer (CRC), particularly in young adults, underscore the imperative to comprehend its fundamental mechanisms, discover novel diagnostic and prognostic markers, and enhance therapeutic strategies. Here, we integrated multi-omics data, including gene expression, somatic mutation data and DNA methylation data, to unravel the intricacies of tumor microenvironment (TME) in CRC and search for novel prognostic markers. By calculating the immune score for each patient from the expression profile, we delineated the differential immune cell fraction, constructed an immune-related multi-omics atlas, and identified molecular characteristics. The entire colorectal dataset (n = 343) was randomly divided into training (n = 249) and testing datasets (n = 94). We screened 144 immune-related genes, 6 mutant genes, and 38 methylation probes associated with overall survival (OS). These makers were then incorporated into a 10-gene prognostic model using Lasso and Cox regression in the training dataset, and the model's performance was evaluated in an independent validation dataset. The model exhibited satisfactory results (average concordance index [C-index] = 0.77), with the average 1-year, 3-year, and 5-year AUCs being 0.79, 0.76, and 0.76 in the training dataset and 0.74, 0.80, and 0.90 in the testing dataset. Furthermore, the prognostic model demonstrated applicability in guiding chemotherapy for CRC patients and exhibited a degree of pan-cancer utility in risk stratification. In conclusion, our integrated analysis of multi-omics data revealed immune-related genetic and epigenetic characteristics of the TME. We propose an integrative prognostic model that can stratify risk and guide chemotherapy for CRC patients. The generalizability of the model in risk stratification across different cancer types was validated in Pan-Cancer cohort.
Collapse
Affiliation(s)
- Yifei Li
- Clinical Biobank, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Hexin Li
- Clinical Biobank, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Gaoyuan Sun
- Clinical Biobank, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Siyuan Xu
- Clinical Biobank, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiaokun Tang
- Clinical Biobank, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Lanxin Zhang
- Clinical Biobank, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Li Wan
- Clinical Biobank, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Lili Zhang
- Clinical Biobank, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Min Tang
- Department of Medical Oncology, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
42
|
Quandt Z, Jacob S, Fadlullah MZH, Wu C, Wu C, Huppert L, Levine LS, Sison P, Tsai KK, Chow M, Kang JH, Hwang J, Lee JC, Oglesby A, Venegas J, Brintz BJ, Tan AC, Anderson MS, Rosenblum MD, Young A, Daud AI. Phase II trial of pembrolizumab, ipilimumab, and aspirin in melanoma: clinical outcomes and translational predictors of response. BJC REPORTS 2024; 2:46. [PMID: 39516257 PMCID: PMC11524064 DOI: 10.1038/s44276-024-00057-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/26/2024] [Accepted: 03/08/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE Many patients with melanoma treated with immune checkpoint inhibitors (ICIs) do not derive response. Preclinical and retrospective studies identified that inhibition of the cyclooxygenase (COX) pathway may improve response to ICI treatment. METHODS This prospective single site phase II trial accrued patients with advanced/metastatic melanoma. Participants underwent high-dose aspirin daily combined with pembrolizumab and ipilimumab every 3 weeks for 4 cycles followed by high-dose aspirin and pembrolizumab monotherapy. The primary endpoint was objective response rate (ORR). Longitudinal sampling of blood was performed to assess peripheral immune correlates. RESULTS Twenty-seven subjects were enrolled with median follow-up of 32 months. An ORR of 62.9% was reached prior to discontinuation due to low likelihood of achieving the pre-specified ORR of 80%. 17 patients (63%) experienced a treatment-related adverse event (TRAEs) grade 3 or higher. A per-protocol analysis showed that patients able to continue aspirin alongside ICI through the induction period experienced significant survival benefit. Ten cytokines and increased regulatory T cells in the periphery correlated with beneficial response. CONCLUSIONS The addition of high-dose aspirin to combination ICI within this study results in response comparable to ICI alone. Future clinical studies of COX inhibition will need to focus on mitigation of AEs to establish the clinical utility of this combination.
Collapse
Affiliation(s)
- Zoe Quandt
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Diabetes Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Saya Jacob
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | | | - Chaorong Wu
- Division of Epidemiology, University of Utah, Salt Lake City, UT, 84112, USA
| | - Clinton Wu
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Laura Huppert
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Lauren S Levine
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Paula Sison
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Katy K Tsai
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Melissa Chow
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Jee Hye Kang
- Diabetes Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Jimmy Hwang
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - James C Lee
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ariel Oglesby
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Jessica Venegas
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, UT, 84112, USA
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Ben J Brintz
- Division of Epidemiology, University of Utah, Salt Lake City, UT, 84112, USA
| | - Aik Choon Tan
- Departments of Oncological Sciences and Biomedical Informatics, University of Utah, Salt Lake City, UT, 84112, USA
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, UT, 84112, USA
| | - Mark S Anderson
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Diabetes Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Michael D Rosenblum
- Dermatology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Arabella Young
- Diabetes Center, University of California San Francisco, San Francisco, CA, 94143, USA.
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, UT, 84112, USA.
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA.
| | - Adil I Daud
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
43
|
Farhat-Younis L, Na M, Zarfin A, Khateeb A, Santana-Magal N, Richter A, Gutwillig A, Rasoulouniriana D, Gleiberman A, Beck L, Giger T, Ashkenazi A, Barzel A, Rider P, Carmi Y. Expression of modified FcγRI enables myeloid cells to elicit robust tumor-specific cytotoxicity. eLife 2024; 12:RP91999. [PMID: 38885133 PMCID: PMC11182644 DOI: 10.7554/elife.91999] [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] [Indexed: 06/20/2024] Open
Abstract
Despite the central role of T cells in tumor immunity, attempts to harness their cytotoxic capacity as a therapy have met limited efficacy, partially as a result of the suppressive microenvironment which limits their migration and activation. In contrast, myeloid cells massively infiltrate tumors and are well adapted to survive these harsh conditions. While they are equipped with cell-killing abilities, they often adopt an immunosuppressive phenotype upon migration to tumors. Therefore, the questions of how to modify their activation programming against cancer, and what signaling cascades should be activated in myeloid cells to elicit their cytotoxicity have remained unclear. Here, we found that activation of IgM-induced signaling in murine myeloid cells results in secretion of lytic granules and massive tumor cell death. These findings open venues for designing novel immunotherapy by equipping monocytes with chimeric receptors that target tumor antigens and consequently, signal through IgM receptor. Nonetheless, we found that myeloid cells do not express the antibody-derived portion used to recognize the tumor antigen due to the induction of an ER stress response. To overcome this limitation, we designed chimeric receptors that are based on the high-affinity FcγRI for IgG. Incubation of macrophages expressing these receptors along with tumor-binding IgG induced massive tumor cell killing and secretion of reactive oxygen species and Granzyme B. Overall, this work highlights the challenges involved in genetically reprogramming the signaling in myeloid cells and provides a framework for endowing myeloid cells with antigen-specific cytotoxicity.
Collapse
Affiliation(s)
- Leen Farhat-Younis
- Department of Pathology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Manho Na
- Department of Pathology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Amichai Zarfin
- Department of Pathology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Aseel Khateeb
- Department of Pathology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | | | - Alon Richter
- Department of Pathology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Amit Gutwillig
- Department of Pathology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | | | - Annette Gleiberman
- Department of Pathology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Lir Beck
- Department of Human Molecular Genetics and Biochemistry, Tel Aviv UniversityTel AvivIsrael
| | - Tamar Giger
- Department of Molecular Cell Biology, Weizmann InstituteRehovotIsrael
| | - Avraham Ashkenazi
- Department of Cell and Developmental Biology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Adi Barzel
- Department of Biochemistry Molecular Biology, George S. Wise Faculty of Life Sciences, Tel Aviv UniversityTel AvivIsrael
| | - Peleg Rider
- Department of Pathology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Yaron Carmi
- Department of Pathology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| |
Collapse
|
44
|
Chen Q, Gao F, Wu J, Zhang K, Du T, Chen Y, Cai R, Zhao D, Deng R, Tang J. Comprehensive pan-cancer analysis of mitochondrial outer membrane permeabilisation activity reveals positive immunomodulation and assists in identifying potential therapeutic targets for immunotherapy resistance. Clin Transl Med 2024; 14:e1735. [PMID: 38899748 PMCID: PMC11187817 DOI: 10.1002/ctm2.1735] [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: 01/25/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Mitochondrial outer membrane permeabilisation (MOMP) plays a pivotal role in cellular death and immune activation. A deeper understanding of the impact of tumour MOMP on immunity will aid in guiding more effective immunotherapeutic strategies. METHODS A comprehensive pan-cancer dataset comprising 30 cancer-type transcriptomic cohorts, 20 immunotherapy transcriptomic cohorts and three immunotherapy scRNA-seq datasets was collected and analysed to determine the influence of tumour MOMP activity on clinical prognosis, immune infiltration and immunotherapy effectiveness. Leveraging 65 scRNA-Seq datasets, the MOMP signature (MOMP.Sig) was developed to accurately reflect tumour MOMP activity. The clinical predictive value of MOMP.Sig was explored through machine learning models. Integration of the MOMP.Sig model and a pan-cancer immunotherapy CRISPR screen further investigated potential targets to overcome immunotherapy resistance, which subsequently underwent clinical validation. RESULTS Our research revealed that elevated MOMP activity reduces mortality risk in cancer patients, drives the formation of an anti-tumour immune environment and enhances the response to immunotherapy. This finding emphasises the potential clinical application value of MOMP activity in immunotherapy. MOMP.Sig, offering a more precise indicator of tumour cell MOMP activity, demonstrated outstanding predictive efficacy in machine-learning models. Moreover, with the assistance of the MOMP.Sig model, FOXO1 was identified as a core modulator that promotes immune resistance. Finally, these findings were successfully validated in clinical immunotherapy cohorts of skin cutaneous melanoma and triple-negative breast cancer patients. CONCLUSIONS This study enhances our understanding of MOMP activity in immune modulation, providing valuable insights for more effective immunotherapeutic strategies across diverse tumours.
Collapse
Affiliation(s)
- Qingshan Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
- Department of Breast OncologySun Yat‐sen University Cancer CenterGuangzhouChina
| | - Fenglin Gao
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Junwan Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
- Biotherapy Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Kaiming Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
- Department of Breast OncologySun Yat‐sen University Cancer CenterGuangzhouChina
| | - Tian Du
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
- Department of Breast OncologySun Yat‐sen University Cancer CenterGuangzhouChina
| | - Yuhong Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Ruizhao Cai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
- Department of Breast OncologySun Yat‐sen University Cancer CenterGuangzhouChina
| | - Dechang Zhao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
- Department of Breast OncologySun Yat‐sen University Cancer CenterGuangzhouChina
| | - Rong Deng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Jun Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
- Department of Breast OncologySun Yat‐sen University Cancer CenterGuangzhouChina
| |
Collapse
|
45
|
Lu DD, Yuan L, Wang ZZ, Zhao JJ, Du YH, Ning N, Chen GQ, Huang SC, Yang Y, Zhang Z, Nan Y. To explore the mechanism of Yigong San anti-gastric cancer and immune regulation. World J Gastrointest Oncol 2024; 16:1965-1994. [PMID: 38764819 PMCID: PMC11099436 DOI: 10.4251/wjgo.v16.i5.1965] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/21/2024] [Accepted: 02/20/2024] [Indexed: 05/09/2024] Open
Abstract
BACKGROUND Yigong San (YGS) is a representative prescription for the treatment of digestive disorders, which has been used in clinic for more than 1000 years. However, the mechanism of its anti-gastric cancer and regulate immunity are still remains unclear. AIM To explore the mechanism of YGS anti-gastric cancer and immune regulation. METHODS Firstly, collect the active ingredients and targets of YGS, and the differentially expressed genes of gastric cancer. Secondly, constructed a protein-protein interaction network between the targets of drugs and diseases, and screened hub genes. Then the clinical relevance, mutation and repair, tumor microenvironment and drug sensitivity of the hub gene were analyzed. Finally, molecular docking was used to verify the binding ability of YGS active ingredient and hub genes. RESULTS Firstly, obtained 55 common targets of gastric cancer and YGS. The Kyoto Encyclopedia of Genes and Genomes screened the microtubule-associated protein kinase signaling axis as the key pathway and IL6, EGFR, MMP2, MMP9 and TGFB1 as the hub genes. The 5 hub genes were involved in gastric carcinogenesis, staging, typing and prognosis, and their mutations promote gastric cancer progression. Finally, molecular docking results confirmed that the components of YGS can effectively bind to therapeutic targets. CONCLUSION YGS has the effect of anti-gastric cancer and immune regulation.
Collapse
Affiliation(s)
- Dou-Dou Lu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Zhao-Zhao Wang
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Jian-Jun Zhao
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yu-Hua Du
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Na Ning
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Guo-Qing Chen
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Shi-Cong Huang
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yi Yang
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Zhe Zhang
- Department of Chinese Medical Gastrointestinal, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yi Nan
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| |
Collapse
|
46
|
Yu KX, Yuan WJ, Wang HZ, Li YX. Extracellular matrix stiffness and tumor-associated macrophage polarization: new fields affecting immune exclusion. Cancer Immunol Immunother 2024; 73:115. [PMID: 38693304 PMCID: PMC11063025 DOI: 10.1007/s00262-024-03675-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 03/12/2024] [Indexed: 05/03/2024]
Abstract
In the malignant progression of tumors, there is deposition and cross-linking of collagen, as well as an increase in hyaluronic acid content, which can lead to an increase in extracellular matrix stiffness. Recent research evidence have shown that the extracellular matrix plays an important role in angiogenesis, cell proliferation, migration, immunosuppression, apoptosis, metabolism, and resistance to chemotherapeutic by the alterations toward both secretion and degradation. The clinical importance of tumor-associated macrophage is increasingly recognized, and macrophage polarization plays a central role in a series of tumor immune processes through internal signal cascade, thus regulating tumor progression. Immunotherapy has gradually become a reliable potential treatment strategy for conventional chemotherapy resistance and advanced cancer patients, but the presence of immune exclusion has become a major obstacle to treatment effectiveness, and the reasons for their resistance to these approaches remain uncertain. Currently, there is a lack of exact mechanism on the regulation of extracellular matrix stiffness and tumor-associated macrophage polarization on immune exclusion. An in-depth understanding of the relationship between extracellular matrix stiffness, tumor-associated macrophage polarization, and immune exclusion will help reveal new therapeutic targets and guide the development of clinical treatment methods for advanced cancer patients. This review summarized the different pathways and potential molecular mechanisms of extracellular matrix stiffness and tumor-associated macrophage polarization involved in immune exclusion and provided available strategies to address immune exclusion.
Collapse
Affiliation(s)
- Ke-Xun Yu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Wei-Jie Yuan
- Department of Gastrointestinal Surgery, Xiangya Hospital of Central South University, Changsha, China
| | - Hui-Zhen Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yong-Xiang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
47
|
Wang W, Fan J, Zhang C, Huang Y, Chen Y, Fu S, Wu J. Targeted modulation of gut and intra-tumor microbiota to improve the quality of immune checkpoint inhibitor responses. Microbiol Res 2024; 282:127668. [PMID: 38430889 DOI: 10.1016/j.micres.2024.127668] [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: 08/08/2023] [Revised: 01/22/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024]
Abstract
Immune checkpoint inhibitor (ICI) therapies, such as those blocking the interaction of PD-1 with its ligands, can restore the immune-killing function of T cells. However, ICI therapy is clinically beneficial in only a small number of patients, and it is difficult to predict post-treatment outcomes, thereby limiting its widespread clinical use. Research suggests that gut microbiota can regulate the host immune system and affect cancer progression and treatment. Moreover, the effectiveness of immunotherapy is related to the composition of the patient's gut microbiota; different gut microbial strains can either activate or inhibit the immune response. However, the importance of the microbial composition within the tumor has not been explored until recently. This study describes recent advances in the crosstalk between microbes in tumors and gut microbiota, which can modulate the tumor microbiome by directly translocating into the tumor and altering the tumor microenvironment. This study focused on the potential manipulation of the tumor and gut microbiota using fecal microbiota transplantation (FMT), probiotics, antimicrobials, prebiotics, and postbiotics to enrich immune-boosting bacteria while decreasing unfavorable bacteria to proactively improve the efficacy of ICI treatments. In addition, the use of genetic technologies and nanomaterials to modify microorganisms can largely optimize tumor immunotherapy and advance personalized and precise cancer treatment.
Collapse
Affiliation(s)
- WeiZhou Wang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - JunYing Fan
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chi Zhang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yuan Huang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yue Chen
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China; Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - ShaoZhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China.
| | - JingBo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China; Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan 646000, China.
| |
Collapse
|
48
|
Sun H, Chang Z, Li H, Tang Y, Liu Y, Qiao L, Feng G, Huang R, Han D, Yin DT. Multi-omics analysis-based macrophage differentiation-associated papillary thyroid cancer patient classifier. Transl Oncol 2024; 43:101889. [PMID: 38382228 PMCID: PMC10900934 DOI: 10.1016/j.tranon.2024.101889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/02/2024] [Accepted: 01/21/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND The reclassification of Papillary Thyroid Carcinoma (PTC) is an area of research that warrants attention. The connection between thyroid cancer, inflammation, and immune responses necessitates considering the mechanisms of differential prognosis of thyroid tumors from an immunological perspective. Given the high adaptability of macrophages to environmental stimuli, focusing on the differentiation characteristics of macrophages might offer a novel approach to address the issues related to PTC subtyping. METHODS Single-cell RNA sequencing data of medullary cells infiltrated by papillary thyroid carcinoma obtained from public databases was subjected to dimensionality reduction clustering analysis. The RunUMAP and FindAllMarkers functions were utilized to identify the gene expression matrix of different clusters. Cell differentiation trajectory analysis was conducted using the Monocle R package. A complex regulatory network for the classification of Immune status and Macrophage differentiation-associated Papillary Thyroid Cancer Classification (IMPTCC) was constructed through quantitative multi-omics analysis. Immunohistochemistry (IHC) staining was utilized for pathological histology validation. RESULTS Through the integration of single-cell RNA and bulk sequencing data combined with multi-omics analysis, we identified crucial transcription factors, immune cells/immune functions, and signaling pathways. Based on this, regulatory networks for three IMPTCC clusters were established. CONCLUSION Based on the co-expression network analysis results, we identified three subtypes of IMPTCC: Immune-Suppressive Macrophage differentiation-associated Papillary Thyroid Carcinoma Classification (ISMPTCC), Immune-Neutral Macrophage differentiation-associated Papillary Thyroid Carcinoma Classification (INMPTCC), and Immune-Activated Macrophage differentiation-associated Papillary Thyroid Carcinoma Classification (IAMPTCC). Each subtype exhibits distinct metabolic, immune, and regulatory characteristics corresponding to different states of macrophage differentiation.
Collapse
Affiliation(s)
- Hanlin Sun
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Zhengyan Chang
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Hongqiang Li
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Yifeng Tang
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Yihao Liu
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Lixue Qiao
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Guicheng Feng
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Runzhi Huang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, PR China.
| | - Dongyan Han
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, PR China.
| | - De-Tao Yin
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, PR China; Engineering Research Center of Multidisciplinary Diagnosis and Treatment of Thyroid Cancer of Henan Province, Zhengzhou 450052, Henan, PR China; Key Medicine Laboratory of Thyroid Cancer of Henan Province, Zhengzhou 450052, Henan, PR China.
| |
Collapse
|
49
|
Huang Q, Ge Y, He Y, Wu J, Tong Y, Shang H, Liu X, Ba X, Xia D, Peng E, Chen Z, Tang K. The Application of Nanoparticles Targeting Cancer-Associated Fibroblasts. Int J Nanomedicine 2024; 19:3333-3365. [PMID: 38617796 PMCID: PMC11012801 DOI: 10.2147/ijn.s447350] [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: 11/12/2023] [Accepted: 03/23/2024] [Indexed: 04/16/2024] Open
Abstract
Cancer-associated fibroblasts (CAF) are the most abundant stromal cells in the tumor microenvironment (TME), especially in solid tumors. It has been confirmed that it can not only interact with tumor cells to promote cancer progression and metastasis, but also affect the infiltration and function of immune cells to induce chemotherapy and immunotherapy resistance. So, targeting CAF has been considered an important method in cancer treatment. The rapid development of nanotechnology provides a good perspective to improve the efficiency of targeting CAF. At present, more and more researches have focused on the application of nanoparticles (NPs) in targeting CAF. These studies explored the effects of different types of NPs on CAF and the multifunctional nanomedicines that can eliminate CAF are able to enhance the EPR effect which facilitate the anti-tumor effect of themselves. There also exist amounts of studies focusing on using NPs to inhibit the activation and function of CAF to improve the therapeutic efficacy. The application of NPs targeting CAF needs to be based on an understanding of CAF biology. Therefore, in this review, we first summarized the latest progress of CAF biology, then discussed the types of CAF-targeting NPs and the main strategies in the current. The aim is to elucidate the application of NPs in targeting CAF and provide new insights for engineering nanomedicine to enhance immune response in cancer treatment.
Collapse
Affiliation(s)
- Qiu Huang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yue Ge
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Jian Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yonghua Tong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Haojie Shang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Xiao Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Ding Xia
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Ejun Peng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| |
Collapse
|
50
|
Hong H, Shi X, Ou W, Ou P. Prognostic biomarker CPEB3 and its associations with immune infiltration in clear cell renal cell carcinoma. Biomed Rep 2024; 20:63. [PMID: 38476610 PMCID: PMC10928475 DOI: 10.3892/br.2024.1751] [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: 09/01/2023] [Accepted: 01/17/2024] [Indexed: 03/14/2024] Open
Abstract
The role and underlying mechanism of cytoplasmic polyadenylation element binding protein 3 (CPEB3) in clear cell renal cell carcinoma [ccRCC progression remain poorly characterized. The present study was designed to evaluate the role of CPEB3 in ccRCC and its clinical associations. The overall response rate of first-line therapies (ICIs combined with VEGFR-TKIs or ICI combination) for ccRCC] is 42.0-59.3%, so a number of patients with ccRCC do not benefit from these therapies. To avoid immunosurveillance and immune killing, tumor cells decrease immunogenicity and recruit immunosuppressive cells such as regulatory T cells (Tregs). Tregs inhibit the development of anti-tumor immunity, thereby hindering immune surveillance of cancer and preventing effective anti-tumor immune response in tumor-bearing hosts. The present study analyzed clinical specimens from patients ccRCC and then examined the role of CPEB3 in ccRCC via bioinformatics analysis. CPEB3 expression was significantly reduced in ccRCC compared with normal tissue and low CPEB3 expression was associated with poor overall survival. Moreover, CPEB3 expression was an independent predictor of survival. CPEB3 expression was positively associated with immune biomarkers [CD274, programmed cell death 1 ligand 2, Hepatitis a virus cellular receptor 2, Chemokine (C-X-C motif) ligand (CXCL)9, CXCL10, Inducible T cell costimulatory, CD40, CD80 and CD38] that improve the outcome of anti-tumor immune responses. CPEB3 expression in ccRCC also affected the status of 24 types of infiltrating immune cell, of which Tregs were the most significantly negatively correlated cell type. CPEB3 may serve as a prognostic biomarker in ccRCC and its mechanism may be related to the regulation of Tregs.
Collapse
Affiliation(s)
- Hualan Hong
- Department of Medical Oncology, Cancer Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350000, P.R. China
- Department of Medical Oncology, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350000, P.R. China
| | - Xi Shi
- Department of Medical Oncology, Cancer Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350000, P.R. China
- Department of Medical Oncology, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350000, P.R. China
| | - Wenyong Ou
- Department of Surgery 1, Longyan People Hospital, Longyan, Fujian 364000, P.R. China
| | - Pengju Ou
- Department of Medical Oncology, Cancer Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350000, P.R. China
- Department of Medical Affairs, Guangzhou Lupeng Pharmaceutical Co., Ltd. Guangzhou, Guangdong 510000, P.R. China
| |
Collapse
|