1
|
Guo H, Wu H, Li J, Li R. Neurological and Cardiac Adverse Events in Cervical Cancer Treatment: A Case of Postoperative Sintilimab-Induced Encephalitis and Myocarditis. AMERICAN JOURNAL OF CASE REPORTS 2025; 26:e947730. [PMID: 40415286 PMCID: PMC12124153 DOI: 10.12659/ajcr.947730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/31/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have shown considerable promise in enhancing patient outcomes and improving survival rates, offering a new frontier in cancer treatment. As a result, their use in clinical practice has become more widespread. However, the adverse effects associated with ICIs can compromise treatment efficacy. Among these, immune-related neurological adverse events are relatively uncommon, with an incidence rate of approximately 17%. Central nervous system (CNS) symptoms, although less frequent (around 6%), are particularly concerning due to their higher risk compared to peripheral nervous system involvement. Additionally, in recent years, the incidence of cardiac toxicity has been increasing, often indicated by elevated cardiac biomarkers, but most cases are asymptomatic. CASE REPORT This report presents a case of a middle-aged woman with cervical cancer who developed both encephalitis and myocarditis during postoperative consolidation therapy with an immune checkpoint inhibitor. A thorough evaluation, including laboratory tests, imaging studies, and an assessment of the patient's medical history and clinical presentation, excluded infection and paraneoplastic encephalitis as potential causes. She was treated with high-dose corticosteroids and intravenous immunoglobulin (IVIG), resulting in gradual resolution of her central nervous system symptoms and normalization of cardiac biomarkers. CONCLUSIONS Although the incidences of immune-related encephalitis and myocarditis are generally low, they can be very severe. With the increasing use of ICIs in clinical practice, the incidence of immune-related neurological symptoms may rise. This highlights the need for increased vigilance in clinical applications, including early preventive measures and prompt diagnosis and treatment to mitigate the adverse effects of these therapies, thereby maximizing patient benefits.
Collapse
Affiliation(s)
- Haiyu Guo
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huanlei Wu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun Li
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ruichao Li
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
2
|
Li X, Wang H, Li X, Zeng M, He Z, Song L, Chen Z, Tang X, Wang A. An antibody-dependent cellular phagocytosis-related gene signature predicts survival and response to immunotherapy in stomach adenocarcinoma. Medicine (Baltimore) 2025; 104:e42079. [PMID: 40193680 PMCID: PMC11977745 DOI: 10.1097/md.0000000000042079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 04/09/2025] Open
Abstract
Antibody-dependent cellular phagocytosis (ADCP) is an immune biological process and plays a biological role in the clearance of tumor cells and the response to immune checkpoint inhibitors. However, the effects of ADCP on stomach adenocarcinoma (STAD) remain unclear. Clinical and genomic data were extracted from multiple datasets. The ADCP-related signature was established using Cox least absolute shrinkage and selection operator regression. Expression of the C5a receptor also known as complement component 5a receptor 1 in the tumor and adjacent-normal tissues was calculated using immunohistochemistry staining. Validation of the signature was conducted in the training and validation cohorts by Cox regression and log-rank tests. Furthermore, the immune infiltrates, the tumor immune dysfunction and exclusion score, and tumor mutation burden score were calculated using the corresponding algorithms, and Mann-Whitney U tests were used to evaluate the differences between groups. Seventy-three hub genes with predictive performance were identified to establish an ADCP-related signature. Accordingly, a 27-gene signature was established, C5a receptor also known as complement component 5a receptor 1, one of the signature genes, had higher expression in tumors than adjacent-normal samples, and its predictive performance was validated in the GSE84437 and The Cancer Genome Atlas cohorts. We found that the ADCP-related signature is an excellent prognostic predictor of STAD. Moreover, the molecular characteristics and some indices of response to immunotherapy differed between the high- and low-risk groups. We constructed a 27-gene signature that is associated with the prognosis and response to STAD-based immunotherapy and provide insights into the biological mechanisms underlying this predictive function.
Collapse
Affiliation(s)
- Xiaochuan Li
- Department of Colorectal and Anorectal Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| | - Hongjian Wang
- General Surgery Department of Yunfu People’s Hospital, Yunfu, China
| | - Xiaofeng Li
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Miaoen Zeng
- Department of Gastroenterology, Fogang County People’s Hospital, Qingyuan, China
| | - Zhuguang He
- Department of Oncology, Zhaoqing First People’s Hospital, Zhaoqing, China
| | - Linjie Song
- Department of Colorectal and Anorectal Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| | - Zhiming Chen
- Department of Integrated Traditional Chinese and Western Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Xinyue Tang
- Department of Oncology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Ang Wang
- Department of Oncology, Guangdong Second Provincial General Hospital, Guangzhou, China
| |
Collapse
|
3
|
Ioannidis K, Dimopoulos A, Decoene I, Guilliams M, Svitina H, Storozhuk L, de Oliveira‐Silva R, Basov S, Thanh NTK, Mourdikoudis S, Van Bael MJ, Smeets B, Sakellariou D, Papantoniou I. 4D Biofabrication of Magnetically Augmented Callus Assembloid Implants Enables Rapid Endochondral Ossification via Activation of Mechanosensitive Pathways. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413680. [PMID: 39998420 PMCID: PMC12005758 DOI: 10.1002/advs.202413680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/31/2025] [Indexed: 02/26/2025]
Abstract
The use of magnetic-driven strategies for non-contact manipulation of engineered living modules opens up new possibilities for tissue engineering. The integration of magnetic nanoparticles (MNPs) with cartilaginous microtissues enables model-driven 4D bottom-up biofabrication of remotely actuated assembloids, providing unique properties to mechanoresponsive tissues, particularly skeletal constructs. However, for clinical use, the long-term effects of magnetic stimulation on phenotype and in vivo functionality need further exploration. Magnetic-driven biofabrication includes both rapid processes, such as guided microtissue assembly, and slower biological processes, like extracellular matrix secretion. This work explores the interplay between magnetic fields and MNP-loaded cartilaginous microtissues through mathematical modeling and experimental approaches, investigating long-term stimulation effects on ECM maturation and chondrogenic hypertrophy. Transcriptomic analysis reveal that magnetic stimulation activated mechanosensitive pathways and catabolic processes, driving accelerated cartilage-to-bone transitions via endochondral ossification, outcomes not observed in non-stimulated controls. This study paves the way for pre-programmed, remotely actuated skeletal assembloids with superior bone-forming capacity for regenerating challenging bone fractures.
Collapse
Affiliation(s)
- Konstantinos Ioannidis
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| | - Andreas Dimopoulos
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| | - Isaak Decoene
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| | - Maya Guilliams
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
- MeBioS division, Biosystems DepartmentKU LeuvenKasteelpark, Arenberg 30Leuven3001Belgium
| | - Hanna Svitina
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| | - Liudmyla Storozhuk
- Healthcare Biomagnetics and Nanomaterials Laboratories, Department of Medical Physics and Biomedical EngineeringUniversity College London21 Albemarle StreetLondonW1S 4BSUK
- London Centre for NanotechnologyUniversity College London17‐19 Gordon StreetLondonWC1H 0AHUK
| | - Rodrigo de Oliveira‐Silva
- Membrane Separations, Adsorption, Catalysis, and Spectroscopy for Sustainable Solutions (cMACS), Department of Microbial and Molecular SystemsKU LeuvenCelestijnenlaan 200F, PB 2454Leuven3001Belgium
| | - Sergey Basov
- Quantum Solid State Physics, Department of Physics and AstronomyKU LeuvenCelestijnenlaan 200DLeuven3001Belgium
| | - Nguyen Thi Kim Thanh
- Healthcare Biomagnetics and Nanomaterials Laboratories, Department of Medical Physics and Biomedical EngineeringUniversity College London21 Albemarle StreetLondonW1S 4BSUK
- Biophysics Group, Department of Physics and AstronomyUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Stefanos Mourdikoudis
- CINBIO, Department of Physical Chemistry, Campus Universitario, Lagoas MarcosendeUniversidade de VigoVigo36310Spain
| | - Margriet J. Van Bael
- Quantum Solid State Physics, Department of Physics and AstronomyKU LeuvenCelestijnenlaan 200DLeuven3001Belgium
| | - Bart Smeets
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
- MeBioS division, Biosystems DepartmentKU LeuvenKasteelpark, Arenberg 30Leuven3001Belgium
| | - Dimitrios Sakellariou
- Membrane Separations, Adsorption, Catalysis, and Spectroscopy for Sustainable Solutions (cMACS), Department of Microbial and Molecular SystemsKU LeuvenCelestijnenlaan 200F, PB 2454Leuven3001Belgium
| | - Ioannis Papantoniou
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| |
Collapse
|
4
|
Chen P, Chen Z, Sui W, Han W. Recent advances in the mechanisms of PD-L1 expression in gastric cancer: a review. Biol Res 2025; 58:16. [PMID: 40091086 PMCID: PMC11912799 DOI: 10.1186/s40659-025-00597-3] [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: 05/04/2024] [Accepted: 03/07/2025] [Indexed: 03/19/2025] Open
Abstract
In the progression of gastric cancer (GC), various cell types in the tumor microenvironment (TME) exhibit upregulated expression of programmed death ligand 1 (PD-L1), leading to impaired T-cell function and evasion of immune surveillance. Infection with H. pylori and EBV leads to increased PD-L1 expression in various cell types within TME, resulting in immune suppression and facilitating immune escape of GC cells. In the TME, mesenchymal stem cells (MSCs), M1-like tumor-associated macrophages (MI-like TAM), and myeloid-derived suppressor cells (MDSCs) contribute to the upregulation of PD-L1 expression in GC cells. Conversely, mast cells, M2-like tumor-associated macrophages (M2-like TAM), and tumor-associated neutrophils (TANs) exhibit elevated levels of PD-L1 expression in response to the influence of GC cells. Together, these factors collectively contribute to the upregulation of PD-L1 expression in GC. This review aims to provide a comprehensive summary of the cellular expression patterns of PD-L1 in GC and the underlying molecular mechanisms. Understanding the complex regulatory pathways governing PD-L1 expression may offer novel insights for the development of effective immunotherapeutic interventions.
Collapse
Affiliation(s)
- Peifeng Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Avenue, Shushan District, Hefei, Anhui Province, 230022, China
| | - Zhangming Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Avenue, Shushan District, Hefei, Anhui Province, 230022, China
| | - Wannian Sui
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Avenue, Shushan District, Hefei, Anhui Province, 230022, China
| | - Wenxiu Han
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Avenue, Shushan District, Hefei, Anhui Province, 230022, China.
| |
Collapse
|
5
|
He X, Guan XY, Li Y. Clinical significance of the tumor microenvironment on immune tolerance in gastric cancer. Front Immunol 2025; 16:1532605. [PMID: 40028336 PMCID: PMC11868122 DOI: 10.3389/fimmu.2025.1532605] [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: 11/22/2024] [Accepted: 01/30/2025] [Indexed: 03/05/2025] Open
Abstract
In the realm of oncology, the tumor microenvironment (TME)-comprising extracellular matrix components, immune cells, fibroblasts, and endothelial cells-plays a pivotal role in tumorigenesis, progression, and response to therapeutic interventions. Initially, the TME exhibits tumor-suppressive properties that can inhibit malignant transformation. However, as the tumor progresses, various factors induce immune tolerance, resulting in TME behaving in a state that promotes tumor growth and metastasis in later stages. This state of immunosuppression is crucial as it enables TME to change from a role of killing tumor cells to a role of promoting tumor progression. Gastric cancer is a common malignant tumor of the gastrointestinal tract with an alarmingly high mortality rate. While chemotherapy has historically been the cornerstone of treatment, its efficacy in prolonging survival remains limited. The emergence of immunotherapy has opened new therapeutic pathways, yet the challenge of immune tolerance driven by the gastric cancer microenvironment complicates these efforts. This review aims to elucidate the intricate role of the TME in mediating immune tolerance in gastric cancer and to spotlight innovative strategies and clinical trials designed to enhance the efficacy of immunotherapeutic approaches. By providing a comprehensive theoretical framework, this review seeks to advance the understanding and application of immunotherapy in the treatment of gastric cancer, ultimately contributing to improved patient outcomes.
Collapse
Affiliation(s)
- Xiangyang He
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xin-Yuan Guan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Clinical Oncology, The University of Hongkong, Hong Kong, Hong Kong SAR, China
| | - Yan Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
6
|
Zhang Q, Yang C, Ma Z, Ye L, Wu Y, Zhong C, Shi Y, Zhu M. Sulforaphane impaired immune checkpoint blockade therapy through activating ΔNP63α/PD-L1 axis in gastric cancer. Mol Carcinog 2024; 63:1611-1620. [PMID: 38780147 DOI: 10.1002/mc.23749] [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: 03/07/2024] [Revised: 04/23/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
Sulforaphane (SFN) exerts anticancer effect on various cancers including gastric cancer. However, the regulatory effect of SFN on programmed death-ligand 1 (PD-L1) and checkpoint blockade therapy in gastric cancer have not been elucidated. Here we demonstrated that SFN suppressed gastric cancer cell growth both in vitro and in vivo study. SFN upregulated PD-L1 expression through activating ΔNP63α in gastric cancer cells. Further, we found that SFN impaired the anticancer effect of anti-PD-L1 monoclonal antibody (α-PD-L1 mab) on gastric cancer cells. These results uncover a novel PD-L1 regulatory mechanism and the double-edged role of SFN in gastric cancer intervention.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Public Health, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chenying Yang
- Yinzhou Center for Disease Control and Prevention, Ningbo, China
| | - Zhijian Ma
- Department of Nutrition, School of Acupuncture and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liangwen Ye
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yunfeng Wu
- Department of Nutrition, School of Acupuncture and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Caiyun Zhong
- Department of Nutrition and Food Safety, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ye Shi
- Department of Thoracic Surgery, Nanjing Chest Hospital, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Mingming Zhu
- Department of Nutrition, School of Acupuncture and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
7
|
Li LL, Yu AY, Zhu M, Ma LY, Cao MH, Liu WL, Qin XB, Gao C, Han ZX, Wang HM. Clinicopathological characteristics and prognosis of Epstein-Barr virus-associated gastric cancer. Arch Virol 2024; 169:114. [PMID: 38700535 DOI: 10.1007/s00705-024-06033-3] [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: 04/12/2023] [Accepted: 02/22/2024] [Indexed: 05/24/2024]
Abstract
OBJECTIVE Epstein-Barr virus (EBV)-associated gastric cancer (EBVaGC) is a distinct molecular subtype of gastric cancer (GC). At present, the clinical characteristics and prognostic implications of EBV infection and the potential clinical benefits of immune checkpoint blockade in GC remain to be clarified. Hence, this study was designed to analyze the clinical and pathological characteristics of GC patients with varying EBV infection states and compare their overall survival (OS). METHODS A retrospective study was performed on 1031 consecutive GC patients who underwent gastrectomy at the Affiliated Hospital of Xuzhou Medical University from February 2018 to November 2022. EBV-encoded RNA (EBER) in situ hybridization (ISH) was used for EBV assessment, and immunohistochemical staining was used for evaluation of human epidermal growth factor receptor 2 (HER2), programmed death ligand 1 (PD-L1), and Ki67 expression. EBVaGC was defined as tumors with EBV positivity. In addition, EBV-negative GC (EBVnGC) patients were matched with EBVaGC patients based on seven clinicopathological parameters (age, gender, anatomic subsite, tumor size, Lauren classification, degree of differentiation, and tumor-node-metastasis [TNM] stage). The correlations of clinical features with HER2, PD-L1, and Ki67 expression were evaluated statistically. The survival of patients was assessed through medical records, telephone, or WeChat communication, and prognostic analysis was performed using the logrank test as well as univariable and multivariable regression analysis. RESULTS Out of 1031 GC patients tested, 35 (3.4%) were diagnosed with EBVaGC. Notably, the EBVaGC group exhibited a distinct predominance of males and younger patients, significantly higher Ki67 and PD-L1 expression levels, and a lower prevalence of pericancerous nerve invasion than the EBVnGC group (P < 0.01). In the 35 EBVaGC cases, Ki67 expression was negatively correlated with age (P < 0.05), suggesting that a younger onset age was associated with higher Ki67 expression. In addition, PD-L1 expression was correlated with the degree of differentiation, T-stage, and clinical stage of the patient. Furthermore, PD-L1 expression was elevated in tumors with lower differentiation or at later stages (P < 0.05). Using univariate analysis, Ki67, PD-L1, and clinical stage were identified as significant factors influencing the overall survival (OS) of EBVaGC patients (P < 0.05). Moreover, multivariate survival analysis revealed that clinical stage and Ki67 expression were independent risk factors for the OS of the patients (P < 0.05), and the three-year OS rate of EBVaGC patients was 64.2%. CONCLUSION EBV-ISH is a practical and valuable method to identify EBVaGC. Owing to its unique etiological, pathological, and clinical characteristics, patients with EBVaGC might benefit from immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Lin-Lin Li
- Department of Pathology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
| | - Ao-Yang Yu
- Graduate School of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Mei Zhu
- Department of Oncology, Xuzhou Cancer Hospital, Xuzhou, 221005, Jiangsu, China
| | - Lu-Yao Ma
- Graduate School of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Meng-Han Cao
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
| | - Wen-Lou Liu
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
| | - Xiao-Bing Qin
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
| | - Chao Gao
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
| | - Zheng-Xiang Han
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China.
| | - Hong-Mei Wang
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China.
| |
Collapse
|
8
|
He XX, Du B, Wu T, Shen H. Prognostic analysis of related factors of adverse reactions to immunotherapy in advanced gastric cancer and establishment of a nomogram model. World J Gastrointest Oncol 2024; 16:1268-1280. [PMID: 38660670 PMCID: PMC11037037 DOI: 10.4251/wjgo.v16.i4.1268] [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: 12/14/2023] [Revised: 01/10/2024] [Accepted: 03/04/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Immunotherapy for advanced gastric cancer has attracted widespread attention in recent years. However, the adverse reactions of immunotherapy and its relationship with patient prognosis still need further study. In order to determine the association between adverse reaction factors and prognosis, the aim of this study was to conduct a systematic prognostic analysis. By comprehensively evaluating the clinical data of patients with advanced gastric cancer treated by immunotherapy, a nomogram model will be established to predict the survival status of patients more accurately. AIM To explore the characteristics and predictors of immune-related adverse reactions (irAEs) in advanced gastric cancer patients receiving immunotherapy with programmed death protein-1 (PD-1) inhibitors and to analyze the correlation between irAEs and patient prognosis. METHODS A total of 140 patients with advanced gastric cancer who were treated with PD-1 inhibitors in our hospital from June 2021 to October 2023 were selected. Patients were divided into the irAEs group and the non-irAEs group according to whether or not irAEs occurred. Clinical features, manifestations, and prognosis of irAEs in the two groups were collected and analyzed. A multivariate logistic regression model was used to analyze the related factors affecting the occurrence of irAEs, and the prediction model of irAEs was established. The receiver operating characteristic (ROC) curve was used to evaluate the ability of different indicators to predict irAEs. A Kaplan-Meier survival curve was used to analyze the correlation between irAEs and prognosis. The Cox proportional risk model was used to analyze the related factors affecting the prognosis of patients. RESULTS A total of 132 patients were followed up, of whom 63 (47.7%) developed irAEs. We looked at the two groups' clinical features and found that the two groups were statistically different in age ≥ 65 years, Ki-67 index, white blood cell count, neutrophil count, and regulatory T cell (Treg) count (all P < 0.05). Multivariate logistic regression analysis showed that Treg count was a protective factor affecting irAEs occurrence (P = 0.030). The ROC curve indicated that Treg + Ki-67 + age (≥ 65 years) combined could predict irAEs well (area under the curve = 0.753, 95% confidence interval: 0.623-0.848, P = 0.001). Results of the Kaplan-Meier survival curve showed that progression-free survival (PFS) was longer in the irAEs group than in the non-irAEs group (P = 0.001). Cox proportional hazard regression analysis suggested that the occurrence of irAEs was an independent factor for PFS (P = 0.006). CONCLUSION The number of Treg cells is a separate factor that affects irAEs in advanced gastric cancer patients receiving PD-1 inhibitor immunotherapy. irAEs can affect the patients' PFS and result in longer PFS. Treg + Ki-67 + age (≥ 65 years old) combined can better predict the occurrence of adverse reactions.
Collapse
Affiliation(s)
- Xu-Xu He
- Department of Surgery, Fudan University Affiliated Zhongshan Hospital (Qingpu Branch), Shanghai 201700, China
| | - Bang Du
- Department of Surgery, Anhui Provincial Red Cross Society Hospital, Hefei 230031, Anhui Province, China
| | - Tao Wu
- Department of General Surgery, West China Hospital of Sichuan University, Chengdu 610044, Sichuan Province, China
| | - Hao Shen
- Department of Surgery, Anhui Provincial Red Cross Society Hospital, Hefei 230031, Anhui Province, China
| |
Collapse
|
9
|
Fu M, Zhang X, Shen F, Ma J, Li Z. Prognostic value of peripheral blood neutrophil/lymphocyte ratio, platelet/lymphocyte ratio, pan-immune-inflammation value and systemic immune-inflammation index for the efficacy of immunotherapy in patients with advanced gastric cancer. Immunotherapy 2024. [PMID: 38578121 DOI: 10.2217/imt-2024-0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024] Open
Abstract
Aim: The study aimed to assess the value of pretreatment peripheral blood neutrophil/lymphocyte ratio (NLR), platelet/lymphocyte ratio (PLR), pan-immune-inflammation value (PIV) and systemic immune-inflammation index (SII) for predicting immunotherapy prognosis and efficacy in advanced gastric cancer (GC). Methods: A total of 84 advanced GC patients received immunotherapy were retrospectively collected. The optimal cut-off values were determined by receiver operating characteristic curves. The univariate and multivariate analysis investigated the effects of NLR, PLR, PIV and SII on patients prognosis. Results: NLR, PLR, PIV and SII had predictive value of efficacy. NLR ≥3.65 was an independent risk factor for worse outcomes. Conclusion: NLR, PLR, PIV and SII have predictive value of efficacy and NLR ≥3.65 suggests a poor prognosis following immunotherapy in advanced GC.
Collapse
Affiliation(s)
- Maodong Fu
- Department of Integrated Traditional Chinese & Western Medicine, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361015, People's Republic of China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, 361015, People's Republic of China
| | - Xiuping Zhang
- Department of Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361015, People's Republic of China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, 361015, People's Republic of China
| | - Feng Shen
- Department of Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361015, People's Republic of China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, 361015, People's Republic of China
| | - Jun Ma
- Department of Integrated Traditional Chinese & Western Medicine, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361015, People's Republic of China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, 361015, People's Republic of China
| | - Zhiyong Li
- Department of Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361015, People's Republic of China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, 361015, People's Republic of China
| |
Collapse
|
10
|
Ren X, Wang L, Liu L, Liu J. PTMs of PD-1/PD-L1 and PROTACs application for improving cancer immunotherapy. Front Immunol 2024; 15:1392546. [PMID: 38638430 PMCID: PMC11024247 DOI: 10.3389/fimmu.2024.1392546] [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: 02/28/2024] [Accepted: 03/22/2024] [Indexed: 04/20/2024] Open
Abstract
Immunotherapy has been developed, which harnesses and enhances the innate powers of the immune system to fight disease, particularly cancer. PD-1 (programmed death-1) and PD-L1 (programmed death ligand-1) are key components in the regulation of the immune system, particularly in the context of cancer immunotherapy. PD-1 and PD-L1 are regulated by PTMs, including phosphorylation, ubiquitination, deubiquitination, acetylation, palmitoylation and glycosylation. PROTACs (Proteolysis Targeting Chimeras) are a type of new drug design technology. They are specifically engineered molecules that target specific proteins within a cell for degradation. PROTACs have been designed and demonstrated their inhibitory activity against the PD-1/PD-L1 pathway, and showed their ability to degrade PD-1/PD-L1 proteins. In this review, we describe how PROTACs target PD-1 and PD-L1 proteins to improve the efficacy of immunotherapy. PROTACs could be a novel strategy to combine with radiotherapy, chemotherapy and immunotherapy for cancer patients.
Collapse
Affiliation(s)
- Xiaohui Ren
- Department of Respiratory Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lijuan Wang
- Department of Hospice Care, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Likun Liu
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Juan Liu
- Department of Special Needs Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
11
|
Wang X, Liu X, Dai H, Jia J. Association of lymphocyte subsets with the efficacy and prognosis of PD‑1 inhibitor therapy in advanced gastric cancer: results from a monocentric retrospective study. BMC Gastroenterol 2024; 24:113. [PMID: 38491354 PMCID: PMC10943815 DOI: 10.1186/s12876-024-03168-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 02/09/2024] [Indexed: 03/18/2024] Open
Abstract
PURPOSE This retrospective study aimed to investigate the changes in peripheral blood lymphocyte subsets before and after immunotherapy in patients with advanced gastric cancer and their relationship n with the therapeutic efficacy and clinical prognosis. METHODS Peripheral blood lymphocyte subsets, including CD4 + T cells, CD8 + T cells, CD4+/CD8 + ratio, NK cells, Treg cells, and B cells, were collected from 195 patients with advanced gastric cancer who were admitted to the First Hospital of Shanxi Medical University with immunotherapy from January 2020 to October 2021, at the time of diagnosis of advanced gastric cancer, before immunotherapy and after 3 cycles of immunotherapy. T-tests were used to examine the factors influencing the patients' peripheral blood lymphocyte subsets and the changes after immunotherapy. To examine the relationship between lymphocyte subsets and treatment outcomes, ROC curves were plotted using a logistic regression. Kaplan-Meier curve was drawn, and the Log Rank test was carried out to compare the differences in PFS between the different groups. Cox proportional hazards regression model was used to analyze the factors affecting PFS after calibration of other variables. RESULTS The proportion of peripheral blood lymphocyte subsets in patients with advanced gastric cancer was affected by age and PD-L1 level. Compared to the baseline, the treatment effective group had higher proportions of CD4 + T cells, a higher CD4+/CD8 + ratio, NK cells and Treg cells, and lower proportions of CD8 + T cells and B cells in the peripheral blood after three cycles of immunotherapy. In the treatment-naive group, there were no significant differences in the lymphocyte subsets. With cut-off values of 30.60% and 18.00%, baseline CD4 + T cell and NK cell ratios were independent predictors of immunotherapy efficacy and PFS. Treg cell ratio, gender, PD-L1 levels, and MMR status all predicted PFS independently. CONCLUSION The proportion of peripheral blood lymphocyte subsets was modified in patients who responded to PD-1 inhibitors. Different lymphocyte subpopulation levels can be used as biomarkers to predict immunotherapy efficacy and clinical prognosis in patients with advanced gastric cancer.
Collapse
Affiliation(s)
- Xinyan Wang
- The First Clinical Medical College of Shanxi Medical University, No.56, Xinjian South Road, Yingze District, Taiyuan, Shanxi, 030001, People's Republic of China
- Department of Oncology, The First Hospital of Shanxi Medical University, No.85, Jiefang South Road, Yingze District, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Xiaoling Liu
- Department of Special Medical, Shanxi Province Cancer Hospital, Taiyuan, Shanxi, 030013, China
| | - Huwei Dai
- The Second Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Junmei Jia
- Department of Oncology, The First Hospital of Shanxi Medical University, No.85, Jiefang South Road, Yingze District, Taiyuan, Shanxi, 030001, People's Republic of China.
| |
Collapse
|
12
|
Kang K, Li X, Peng Y, Zhou Y. Comprehensive Analysis of Disulfidptosis-Related LncRNAs in Molecular Classification, Immune Microenvironment Characterization and Prognosis of Gastric Cancer. Biomedicines 2023; 11:3165. [PMID: 38137387 PMCID: PMC10741100 DOI: 10.3390/biomedicines11123165] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Disulfidptosis is a novel form of programmed cell death that unveils promising avenues for the exploration of tumor treatment modalities. Gastric cancer (GC) is a malignant tumor characterized by high incidence and mortality rate. However, there has been no systematic study of disulfidptosis-related long noncoding RNAs (DRLs) signature in GC patients. METHODS The lncRNA expression profiles containing 412 GC samples were acquired from the Cancer Genome Atlas (TCGA) database. Differential expression analysis was performed alongside Pearson correlation analysis to identify DRLs. Prognostically significant DRLs were further screened using univariate COX regression analysis. Subsequently, LASSO regression and multifactorial COX regression analyses were employed to establish a risk signature composed of DRLs that exhibit independent prognostic significance. The predictive value of this risk signature was further validated in a test cohort. The ESTIMATE, CIBERSORT and ssGSEA methodologies were utilized to investigate the tumor immune microenvironment of GC populations with different DRLs profiles. Finally, the correlation between DRLs and various GC drug responses was explored. RESULTS We established a prognostic signature comprising 12 disulfidptosis-related lncRNAs (AC110491.1, AL355574.1, RHPN1-AS1, AOAH-IT1, AP001065.3, MEF2C-AS1, AC016394.2, LINC00705, LINC01952, PART1, TNFRSF10A-AS1, LINC01537). The Kaplan-Meier survival analysis revealed that patients in the high-risk group exhibited a poor prognosis. Both univariate and multivariate COX regression models demonstrated that the DRLs signature was an independent prognostic indicator in GC patients. Furthermore, the signature exhibited accurate predictions of survival at 1-, 3- and 5- years with the area under the curve (AUC) values of 0.708, 0.689 and 0.854, respectively. In addition, we also observed significant associations between the DRLs signature and various clinical variables, distinct immune landscape and drug sensitivity profiles in GC patients. The low-risk group patients may be more likely to benefit from immunotherapy and chemotherapy. CONCLUSIONS Our study investigated the role and potential clinical implications of DRLs in GC. The risk model constructed by DRLs demonstrated high accuracy in predicting the survival outcomes of GC and improving the treatment efficacy for GC patients.
Collapse
Affiliation(s)
- Kuo Kang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China;
- Hunan Key Laboratory of Precise Diagnosis and Treatment of Gastrointestinal Tumor, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xuanxuan Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China;
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yuanhao Peng
- National Health Council Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha 410078, China;
| | - Yangying Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China;
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
13
|
Mou P, Ge QH, Sheng R, Zhu TF, Liu Y, Ding K. Research progress on the immune microenvironment and immunotherapy in gastric cancer. Front Immunol 2023; 14:1291117. [PMID: 38077373 PMCID: PMC10701536 DOI: 10.3389/fimmu.2023.1291117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/24/2023] [Indexed: 12/18/2023] Open
Abstract
The tumor microenvironment, particularly the immune microenvironment, plays an indispensable role in the malignant progression and metastasis of gastric cancer (GC). As our understanding of the GC microenvironment continues to evolve, we are gaining deeper insights into the biological mechanisms at the single-cell level. This, in turn, has offered fresh perspectives on GC therapy. Encouragingly, there are various monotherapy and combination therapies in use, such as immune checkpoint inhibitors, adoptive cell transfer therapy, chimeric antigen receptor T cell therapy, antibody-drug conjugates, and cancer vaccines. In this paper, we review the current research progress regarding the GC microenvironment and summarize promising immunotherapy research and targeted therapies.
Collapse
Affiliation(s)
- Pei Mou
- Changzheng Hospital of Naval Medical University, Shanghai, China
| | - Qing-hua Ge
- Department of Otolaryngology, Changzheng Hospital of Naval Medical University, Shanghai, China
| | - Rong Sheng
- Department of Outpatient, Changzheng Hospital of Naval Medical University, Shanghai, China
| | - Teng-fei Zhu
- Department of Anesthesiology, Changzheng Hospital of Naval Medical University, Shanghai, China
| | - Ye Liu
- Department of Blood Transfusion, Changzheng Hospital of Naval Medical University, Shanghai, China
| | - Kai Ding
- Department of Gastroenterology, Changzheng Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
14
|
Yang X, Zhu Z, Liang T, Lei X. Comprehensive analysis of anoikis-related genes in prognosis and immune infiltration of gastric cancer based on bulk and single-cell RNA sequencing data. J Cancer Res Clin Oncol 2023; 149:13163-13173. [PMID: 37474682 DOI: 10.1007/s00432-023-05157-4] [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: 05/30/2023] [Accepted: 07/09/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Accumulating evidence suggests that anoikis resistance is a key process in cancer cell metastasis, making it an attractive therapeutic target. Therefore, anoikis may become a new treatment for gastric cancer. METHODS We used the univariate Cox regression method to screen gastric cancer-related anoikis genes, and a prognostic risk model was established. We analyzed differences between high- and low-risk groups in terms of tumor infiltrating immune cells, gene mutation signatures, and treatment of gastric cancer. Analysis of model associated genes at single-cell resolution was performed. RESULTS We filtered to 12 anoikis-related genes and built a prognostic risk model using seven of them, which performed well in multiple datasets. Patients with CCDC178 mutations had a worse prognosis. We also found that patients at low risk were more likely to benefit from chemotherapy and immunotherapy. ERBB2 was found to be highly expressed in epithelial cells and fibroblasts. Our analysis also indicated that gastric cancer samples with high infiltration of iCAFs had a worse prognosis. CONCLUSION Seven anoikis-related genes were selected to establish a risk model. The model can be used to predict the prognosis of patients and guide the drug treatment, which provides a new idea for the evaluation and treatment of gastric cancer patients.
Collapse
Affiliation(s)
- Xiaobo Yang
- Center for General Practice Medicine, Department of Nursing, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Shangtang Road 158, Hangzhou, Zhejiang, China, 310014
| | - Zheng Zhu
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Shangtang road 158, Hangzhou, Zhejiang, China, 310014
| | - Tianyu Liang
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Shangtang road 158, Hangzhou, Zhejiang, China, 310014.
| | - Xiaoju Lei
- Center for General Practice Medicine, Department of Nursing, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Shangtang Road 158, Hangzhou, Zhejiang, China, 310014.
| |
Collapse
|
15
|
Kang J, Han KM, Jung H, Kim H. Prognostic Significance of Programmed Cell Death Ligand 1 Expression in High-Grade Serous Ovarian Carcinoma: A Systematic Review and Meta-Analysis. Diagnostics (Basel) 2023; 13:3258. [PMID: 37892079 PMCID: PMC10606661 DOI: 10.3390/diagnostics13203258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/09/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
(1) Background: High-grade serous ovarian carcinoma (HGSOC) is an aggressive subtype of ovarian cancer. Recent advances have introduced prognostic markers and targeted therapies. Programmed cell death ligand 1 (PD-L1) has emerged as a potential biomarker for HGSOC, with implications for prognosis and targeted therapy eligibility; (2) Methods: A literature search was conducted on major databases, and extracted data were categorized and pooled. Subgroup analysis was performed for studies with high heterogeneity. (3) Results: Data from 18 eligible studies were categorized and pooled based on PD-L1 scoring methods, survival analysis types, and endpoints. The result showed an association between high PD-L1 expression and a favorable prognosis in progression-free survival (HR = 0.53, 95% CI = 0.35-0.78, p = 0.0015). Subgroup analyses showed similar associations in subgroups of neoadjuvant chemotherapy patients (HR = 0.6, 95% CI = 0.4-0.88, p = 0.009) and European studies (HR = 0.59, 95% CI = 0.42-0.82, p = 0.0017). In addition, subgroup analyses using data from studies using FDA-approved PD-L1 antibodies suggested a significant association between favorable prognosis and high PD-L1 expression in a subgroup including high and low stage data in overall survival data (HR = 0.46, 95% CI = 0.3-0.73, p = 0.0009). (4) Conclusions: This meta-analysis revealed a potential association between high PD-L1 expression and favorable prognosis. However, caution is warranted due to several limitations. Validation via large-scale studies, with mRNA analysis, whole tissue sections, and assessments using FDA-approved antibodies is needed.
Collapse
Affiliation(s)
| | | | | | - Hyunchul Kim
- Department of Pathology, CHA Ilsan Medical Center, 1205 Jungang-ro, Ilsandong-gu, Goyang-si 10414, Republic of Korea; (J.K.); (K.M.H.)
| |
Collapse
|
16
|
Chen G, Luo D, Qi X, Li D, Zheng J, Luo Y, Zhang C, Ren Q, Lu Y, Chan YT, Chen B, Wu J, Wang N, Feng Y. Characterization of cuproptosis in gastric cancer and relationship with clinical and drug reactions. Front Cell Dev Biol 2023; 11:1172895. [PMID: 37351275 PMCID: PMC10283039 DOI: 10.3389/fcell.2023.1172895] [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: 02/23/2023] [Accepted: 05/09/2023] [Indexed: 06/24/2023] Open
Abstract
Gastric cancer (GC) is the fifth most common cancer worldwide. Cuproptosis is associated with cell growth and death as well as tumorigenesis. Aiming to lucubrate the potential influence of CRGs in gastric cancer, we acquired datasets of gastric cancer patients from TCGA and GEO. The identification of molecular subtypes with CRGs expression was achieved through unsupervised learning-cluster analysis. To evaluate the application value of subtypes, the K-M survival analysis was conducted to evaluate the clinical prognostic characteristics. Subsequently, we performed Gene Set Variation Analysis (GSVA) and utilized ssGSEA to quantify the extent of immune infiltration. Further, the K-M survival analysis was used to identify the prognosis-related CRGs. Next, signature genes of diagnostic predictive value were screened using the least absolute shrinkage and selection operator (LASSO) algorithm from the expression matrix for TCGA, as well as the signature gene-related subtype was clustered by the "ConsensusClusterPlus" package. Finally, the immunological and drug sensitivity assessments of the signature gene-related subtypes were conducted. A total of 173 CRGs were identified, most of the CRGs undergo copy number variation in gastric cancer. Under different patient subtypes, immune cell levels differed significantly, and the subtype exhibiting high expression of the CRGs had a better prognosis. Furthermore, we selected 34 CRGs that were highly correlated with the prognosis of gastric cancer. By constructing a multivariate Cox proportional-hazards model and a hazard scoring system, we were able to categorize patients into high- and low-risk groups based on their hazard score. K-M analysis demonstrated a significant survival disadvantage in the high-risk group. Based on Lasso regression analysis, we screened 16 signature genes, a multivariate logistic regression model [cutoff: 0.149 (0.000, 0.974), AUC:0.987] and a prognosis network diagram was constructed and their prediction efficiency for gastric cancer prognostic diagnosis was well validated. According to the signature genes, the patients were separated to two signature subtypes. We found that patients with higher CRGs expression and better prognosis had lower levels of immune infiltration. Finally, according to the results of drug susceptibility analysis, docetaxel, 5-Fluorouracil, gemcitabin, and paclitaxel were found to be more sensitive to gastric cancer.
Collapse
Affiliation(s)
- Guoming Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Dongqiang Luo
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiangjun Qi
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Danyun Li
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiyuan Zheng
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yang Luo
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Qing Ren
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yuanjun Lu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yau-Tuen Chan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Junyu Wu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| |
Collapse
|