1
|
Cai Q, You S, Huang J, Gong C, Zhang W, Zhou A. Cost-effectiveness of trastuzumab deruxtecan as a second-line treatment for HER2-mutant advanced non-small cell lung cancer. Hum Vaccin Immunother 2025; 21:2468070. [PMID: 39989197 PMCID: PMC11853545 DOI: 10.1080/21645515.2025.2468070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/28/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025] Open
Abstract
The study of DESTINY-Lung01 and DESTINY-Lung02 demonstrated the favorable efficacy and optimal dosage of trastuzumab deruxtecan (T-DXd) in managing the human epidermal growth factor receptor 2 (HER2)-mutant non-small cell lung cancer (NSCLC) patients who had received previous treatment. The study sought to assess the cost-effectiveness of T-DXd in both the United States (US) and Chinese healthcare systems. Markov models were developed to evaluate the overall cost, incremental cost-effectiveness ratio (ICER), quality-adjusted life years (QALYs), and life years (LYs) of treatment with T-DXd compared with docetaxel, nivolumab, and pyrotinib for patients in the US and China. The level of willingness-to-pay (WTP) in the US and China is 150,000/QALYs and 32,517/QALYs, respectively. Sensitivity analyses were carried out to ensure the precision of the model. T-DXd yielded additional QALYs of 0.63 and 0.06 with an ICER of $338997.84 and $1437258.33 per QALY, respectively, in the US compared to the docetaxel and nivolumab regimens. And T-DXd yielded additional QALYs of 0.63, 0.06, and 0.13 with an ICER of $137959.45, $623805.93, and $515447.12 per QALY, respectively, in China compared to the docetaxel, nivolumab, and pyrotinib regimens. Sensitivity analysis showed that the cost of drugs is the most influential factor. T-DXd provides substantial therapeutic benefit for NSCLC patients with HER2 mutations who have had previous treatment but is not deemed cost-effective in either the US or China when compared to docetaxel, nivolumab, and pyrotinib. Price reduction is perhaps the main way to make T-DXd cost-effective.
Collapse
Affiliation(s)
- Qi Cai
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuhui You
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinglong Huang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Caifeng Gong
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wen Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Aiping Zhou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
2
|
Qiao Y, Xie D, Li Z, Cao S, Zhao D. Global research trends on biomarkers for cancer immunotherapy: Visualization and bibliometric analysis. Hum Vaccin Immunother 2025; 21:2435598. [PMID: 39773010 PMCID: PMC11730411 DOI: 10.1080/21645515.2024.2435598] [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: 09/05/2024] [Revised: 11/08/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
The global burden of cancer continues to grow, posing a significant public health challenge. Although cancer immunotherapy has shown significant efficacy, the response rate is not high. Therefore, the objective of our research was to identify the latest research trends and hotspots on biomarkers from 1993 to 2023. Data were collected from the database Web of Science core collection. Bibliometric analysis and visualization were conducted with CiteSpace(6.3.1), VOSviewer (v1.6.20), R-bibliometrix(v4.3.3), and Microsoft Excel(2019). A total of 2686 literatures were retrieved. The sheer annual volume of publications has shown a rapid upward trend since 2015. The United States has generated the most publications and Harvard University ranked as a leading institution. The global biomarker research on immune checkpoint inhibitors (ICIs) revealed regional differences and in-depth explorations should be promoted in developing countries. Although China has become the second largest country in terms of publication, the average citation per paper and the total link strength were both lower than the other countries. The research on biomarkers mainly concentrated upon the following aspects: PD-1/PD-L1, CTLA-4, gene expression, adverse events, total mutational burden (TMB), body mass index (BMI), gut microbiota, cd8(+)/cd4(+) t-cells, and blood-related biomarkers such as lactate dehydrogenase (LDH), neutrophil-lymphocyte ratio (NLR), cytokines. Furthermore, "artificial intelligence" and "machine learning" have become the most important research hotspot over the last 2 y, which will help us to identify useful biomarkers from complex big data and provide a basis for precise medicine for malignant tumors.
Collapse
Affiliation(s)
- Yuan Qiao
- Department of Clinical Pharmacy, Yan’an University Affiliated Hospital, Yan’an, Shaanxi, China
| | - Dong Xie
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhengxiang Li
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Shaohua Cao
- Department of Clinical Pharmacy, Yan’an University Affiliated Hospital, Yan’an, Shaanxi, China
| | - Dong Zhao
- Department of Clinical Laboratory, Yan’an University Affiliated Hospital, Yan’an, Shaanxi, China
| |
Collapse
|
3
|
Zhang S, Miao L, Tian X, Yang B, Luo B. Opportunities and challenges of immuno-oncology: A bibliometric analysis from 2014 to 2023. Hum Vaccin Immunother 2025; 21:2440203. [PMID: 39885669 PMCID: PMC11792843 DOI: 10.1080/21645515.2024.2440203] [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: 09/11/2024] [Revised: 11/22/2024] [Accepted: 12/06/2024] [Indexed: 02/01/2025] Open
Abstract
The emergence of immuno-oncology (IO) has led to revolutionary changes in the field of cancer treatment. Despite notable advancements in this field, a thorough exploration of its full depth and extent has yet to be performed. This study provides a comprehensive overview of publications pertaining to IO. Publications on IO from 2014 to 2023 were retrieved by searching the Web of Science Core Collection database (WoSCC). VOSviewer software and Citespace software were used for the visualized analysis. A total of 1,874 articles have been published in the IO domain. The number of publications and citations has been increasing annually. This study also examines the primary research directions within the field of IO. In conclusion, this study offers a comprehensive overview of the opportunities and challenges associated with IO, illuminating the current status of research and indicating potential future trajectories in this rapidly progressing field. This study provides a comprehensive survey of the current research status and hot spots within the field of IO. It will assist researchers in comprehending the current research emphasis and development trends in this field and offers guidance for future research directions.
Collapse
Affiliation(s)
- Siqi Zhang
- School of Clinical Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Department of Oncology, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, China
- Department of Oncology, Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| | - Lina Miao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoxia Tian
- School of Clinical Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Bingxu Yang
- School of Clinical Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Baoping Luo
- School of Clinical Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Department of Oncology, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, China
- Department of Oncology, Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
4
|
Fang W, Ma X, Liu B. Global research progress in antibody-drug conjugates for solid tumors: Bibliometrics and visualized analysis. Hum Vaccin Immunother 2025; 21:2472493. [PMID: 40013384 PMCID: PMC11869778 DOI: 10.1080/21645515.2025.2472493] [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: 09/22/2024] [Revised: 02/14/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025] Open
Abstract
Recently, the use of antibody-drug conjugates (ADCs) in the research and management of solid tumors has increased, making them a key focus in the field of oncology. In this study, we performed a comprehensive literature review of ADCs use in solid tumor treatment. We retrieved data from the Web of Science Core Collection (WoSCC). Following literature retrieval, we conducted a thorough bibliometric and knowledge-mapping analysis of the collected articles. There was a rapid growth in the number of annual publications in this field. The United States had the highest publication volumes and led ADC research for solid tumors. Additionally, The Dana-Farber Cancer Institute had the highest output, and G. Curigliano was identified as the most productive author. The journal "Cancers" led in the publishing of ADC research on solid tumors. Furthermore, key clustering terms such as "breast cancer," "targeted therapy," "bladder cancer," "ovarian cancer," "expression," and "drug delivery" emerged in this field as the research progressed. We identified six key themes by literature co-citation analysis, involving the research on the application of four ADCs in breast cancer, as well as the analysis of ADCs design, mechanisms, and strategies for reducing cytotoxicity. At the same time, based on the analysis of papers that have experienced a citation burst recently, we explored the future development trends of this field. Overall, our inaugural bibliometric analysis of ADCs for solid tumor research provides a systematic framework to guide future studies in this field. Therefore, facilitating and promoting further development in this area.
Collapse
Affiliation(s)
- Wenjun Fang
- Department of Pharmacy, The Affiliated Yancheng Maternity & Child Health Hospital of Yangzhou University, Yancheng, China
| | - Xueqing Ma
- Department of Dermatology, The Affiliated Yancheng Maternity & Child Health Hospital of Yangzhou University, Yancheng, China
| | - Ben Liu
- Yancheng No.1 People’s Hospital, Affiliated Hospital of Medical School, Nanjing University, Yancheng, China
- Pediatric Intensive Care Unit, The First People’s Hospital of Yancheng, Yancheng, China
| |
Collapse
|
5
|
Zhang Y, Fan W, Su F, Zhang X, Du Y, Li W, Gao Y, Hu W, Zhao J. Discussion on the mechanism of HER2 resistance in esophagogastric junction and gastric cancer in the era of immunotherapy. Hum Vaccin Immunother 2025; 21:2459458. [PMID: 39875210 PMCID: PMC11776468 DOI: 10.1080/21645515.2025.2459458] [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: 11/06/2024] [Revised: 01/11/2025] [Accepted: 01/24/2025] [Indexed: 01/30/2025] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) is a critical biomarker and therapeutic target in gastric/gastroesophageal junction (G/GEJ) cancers, despite the initial success of HER2-targeted therapies, such as trastuzumab, resistance to these drugs has emerged as a major impediment to effective long-term treatment. This review examines the mechanisms of drug resistance in HER2-positive G/GEJ cancer, the primary mechanisms of resistance explored include alterations in the HER2 receptor itself, such as mutations and changes in expression levels, as well as downstream signaling pathways, and interactions with the tumor microenvironment (TME). Furthermore, the review discusses the Novel therapeutic approaches, including the use of antibody-drug conjugates (ADCs) and combination therapies are assessed for their potential to enhance outcomes. By integrating recent research findings and clinical trials, this review aims to provide oncologists and researchers with insights into developing more effective treatments for patients with drug-resistant HER2-positive G/GEJ cancer.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Graduate School, Changzhi Medical College, Changzhi, Shanxi, China
| | - Wenxuan Fan
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Graduate School, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Fei Su
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Graduate School, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoling Zhang
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Yunyi Du
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Weiling Li
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Graduate School, Changzhi Medical College, Changzhi, Shanxi, China
| | - Yangjun Gao
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Wenqing Hu
- Department of Gastrointestinal Surgery, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Jun Zhao
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| |
Collapse
|
6
|
He Q, Liu X, Jiang L, Liu P, Xuan W, Wang Y, Meng R, Feng H, Lv S, Miao Q, Zheng D, Xu Y, Wang M. First-line treatments for KRAS-mutant non-small cell lung cancer: current state and future perspectives. Cancer Biol Ther 2025; 26:2441499. [PMID: 39681355 PMCID: PMC11651285 DOI: 10.1080/15384047.2024.2441499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/20/2024] [Accepted: 12/09/2024] [Indexed: 12/18/2024] Open
Abstract
KRAS mutations are common in non-small cell lung cancer (NSCLC) and are associated with patient prognosis; however, targeting KRAS has faced various difficulties. Currently, immunotherapy, chemotherapy, and chemoimmunotherapy play pivotal roles in the first-line treatment of KRAS-mutated NSCLC. Here, we summarize the current evidence on first-line therapies and compare the treatment outcomes and biomarkers for different regimens. KRAS inhibitors and other emerging alternative treatments are also discussed, as combining these drugs with immunotherapy may serve as a promising first-line treatment for KRAS-mutated NSCLC in the future. We hope that this review will assist in first-line treatment choices and shed light on the development of novel agents for KRAS-mutated NSCLC.
Collapse
Affiliation(s)
- Qi He
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoyan Liu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liyan Jiang
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Liu
- Department of Respiratory Medicine, Changsha Hospital Affiliated to Xiangya Medical College, Central South University (The First Hospital of Changsha), Changsha, China
| | - Weixia Xuan
- Department of Respiratory and Critical Care Medicine, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Yudong Wang
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Rui Meng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huijing Feng
- Department of Thoracic Oncology, Cancer Center, Shanxi Bethune Hospital, Taiyuan, Shanxi, China
| | - Shuang Lv
- Department of Internal Medicine-Oncology, Inner Mongolia People’s Hospital, Huhehot, Inner Mongolia, P.R. China
| | - Qian Miao
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fuzhou, China
| | - Di Zheng
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Yan Xu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengzhao Wang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
7
|
Rocha P, Bach R, Masfarré L, Hernandez S, Navarro-Gorro N, Rossell A, Villanueva X, Giner M, Sanchéz I, Galindo M, Del Rey-Vergara R, Iñañez A, Sanchéz-Espiridion B, Lu W, Acedo-Terrades A, Berenguer-Molins P, Sánchez-Font A, Chalela R, Curull V, Taus Á, Hardy-Werbin M, Sausen M, Georgiadis A, White J, Jackson JB, Moliner L, Clavé S, Bellosillo B, Rovira A, Wistuba I, Soto LMS, Perera-Bel J, Arriola E. Molecular and immunological features associated with long-term benefits in metastatic NSCLC patients undergoing immune checkpoint blockade. Oncoimmunology 2025; 14:2469377. [PMID: 39991958 PMCID: PMC11853546 DOI: 10.1080/2162402x.2025.2469377] [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] [Revised: 01/22/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025] Open
Abstract
INTRODUCTION Immunotherapy is firmly established as a treatment regimen in various solid tumors, driven by its exceptional benefits in a selected group of patients. Despite widespread adoption of immune checkpoint blockade (ICB) across diverse solid tumors, the quest for a clinically informative biomarker for long-term benefit remains unmet. METHODS A total of 49 patients with metastatic NSCLC treated with ICB were included. Long-term (LTR) and short-term responders (STR) were defined as those with a response to ICB lasting more than 24 months or less than 6 months, respectively. Longitudinal blood specimens were collected before ICB treatment initiation and early-on treatment. Plasma ctDNA next-generation sequencing panel (NGS) and serum proteomics were performed. GeoMx DSP on baseline tumor tissue was performed in a subset of patients. RESULTS Our analysis revealed specific characteristics of LTR compared with STR, namely higher PD-L1 in tumor cells (p = 0.005) and higher incidence of irAEs (p = 0.001). Genomic features associated with lack of benefit from ICB included co-occurring mutations in KRAS/STK11 and TP53/KMT2D (p < 0.05). At a baseline, LTR patients exhibited higher serum levels of proteins related with apoptosis (CASP8, PRKRA), chemotaxis, immune proteasome, processing of MHC class I (S100A4, PSMD9, RNF41) and immune homeostasis (HAVCR1, ARG1) (p < 0.05). Protein spatial profiling of tumor samples showed higher levels of proteins linked with the presence of immune cells (CD45), T cells (CD8), antigen presentation (HLA-DR) and immune regulation proteins (PD-L1, IDO1) within the tumor and tumor stroma component (p < 0.05) in LTR patients. Serum longitudinal analysis identified a set of proteins that presented distinct dynamics in LTR compared to STR, making them interesting candidates to evaluate as early predictors of treatment efficacy. CONCLUSIONS Our multimodal analysis of patients with metastatic NSCLC treated with ICB identified clinicopathological and immunological features associated with long-term benefits. The presence of preexisting antitumor immunity emerged as a strong predictor of long-term benefits, providing insights for potential biomarkers and therapeutic strategies for enhancing ICB outcomes in metastatic NSCLC.
Collapse
Affiliation(s)
- Pedro Rocha
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Rafael Bach
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Laura Masfarré
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Sharia Hernandez
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Adrià Rossell
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | | | - Mario Giner
- Pathology Department, Hospital del Mar, Barcelona, Spain
| | | | - Miguel Galindo
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | | | - Albert Iñañez
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Beatriz Sanchéz-Espiridion
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Wei Lu
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | - Victor Curull
- Pulmonology Department, Hospital del Mar, Barcelona, Spain
| | - Álvaro Taus
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | | | | | | | | | | | | | - Sergi Clavé
- Pathology Department, Hospital del Mar, Barcelona, Spain
| | - Beatriz Bellosillo
- Pathology Department, Hospital del Mar, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ana Rovira
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Luisa M Solis Soto
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Edurne Arriola
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|
8
|
Zhang T, Chen Y, Xiang Z. Machine learning-based integration develops a disulfidptosis-related lncRNA signature for improving outcomes in gastric cancer. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2025; 53:1-13. [PMID: 39701937 DOI: 10.1080/21691401.2024.2440415] [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: 05/29/2024] [Revised: 11/05/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024]
Abstract
Gastric cancer remains one of the deadliest cancers globally due to delayed detection and limited treatment options, underscoring the critical need for innovative prognostic methods. Disulfidptosis, a recently discovered programmed cell death triggered by disulphide stress, presents a fresh avenue for therapeutic exploration. This research examines disulfidptosis-related long noncoding RNAs (DRLs) in gastric cancer, with the goal of leveraging these lncRNAs as potential markers to enhance patient outcomes and treatment approaches. Comprehensive genomic and clinical data from stomach adenocarcinoma (STAD) were obtained from The Cancer Genome Atlas (TCGA). Employing least absolute shrinkage and selection operator (LASSO) regression analysis, a prognostic model was devised incorporating five key DRLs to forecast survival rates. The effectiveness of this model was validated using Kaplan-Meier survival plots, receiver operating characteristic (ROC) curves, and extensive functional enrichment studies. The importance of select lncRNAs and the expression variability of genes tied to disulfidptosis were validated via quantitative real-time PCR (qRT-PCR) and Western blot tests, establishing a solid foundation for their prognostic utility. Analyses of functional enrichment and tumour mutation burden highlighted the biological importance of these DRLs, connecting them to critical cancer pathways and immune responses. These discoveries broaden our comprehension of the molecular framework of gastric cancer and bolster the development of tailored treatment plans, highlighting the substantial role of DRLs in clinical prognosis and therapeutic intervention.
Collapse
Affiliation(s)
- Tianze Zhang
- Department of Gastrointestinal Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Yuqing Chen
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Zhiping Xiang
- Head and Neck Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
9
|
Wang T, Desai AA, Thurber GM, Tessier PM. Maximizing activity and selectivity of antibody-mediated effector functions using antibody mixtures. MAbs 2025; 17:2480666. [PMID: 40180622 PMCID: PMC11980503 DOI: 10.1080/19420862.2025.2480666] [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: 05/28/2024] [Revised: 02/17/2025] [Accepted: 03/12/2025] [Indexed: 04/05/2025] Open
Abstract
Fc-mediated effector functions are key for conferring potent antibody-mediated killing of cancer cells. However, it is difficult to achieve highly selective targeting of cancer cells while minimizing toxicity on healthy tissue because of the expression of most receptors, albeit at lower levels, on non-cancer cells. Previous attempts to increase the selectivity of antibody-mediated effector functions have sought to reduce binding affinity and/or increase avidity, which typically results in modest improvements in selectivity. To overcome this limitation, we report the use of mixtures of antibody variants that achieve high selectivity based on receptor level while maintaining high activity for cells with high receptor levels. We have studied mixtures of two variants of an anti-HER2 antibody (trastuzumab), one that is affinity-reduced and effector-competent and a second high-affinity variant that is effectorless. Notably, we observe that the high-affinity, effectorless antibody reduces effector function for cells with low receptor levels, including reduced antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP), while the high-avidity, effector-competent antibody mediates significant effector function for cells with high receptor levels. Moreover, replacing the effector-competent Fc region of the affinity-reduced antibody with high-affinity Fc domains that enhance effector function drives high activity while maintaining high selectivity for the antibody mixtures. These findings outline a general strategy for maximizing the therapeutic window by selectively targeting cancer cells based on receptor levels that could be applied to a wide range of applications involving antibody-mediated synapse formation, including antibody-drug conjugates and bispecific antibodies, such as T cell engagers.
Collapse
Affiliation(s)
- Tiexin Wang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Alec A. Desai
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Greg M. Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Peter M. Tessier
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
10
|
Bartsch YC, Webb NE, Burgess E, Kang J, Lauffenburger DA, Julg BD. Combinatorial Fc modifications for complementary antibody functionality. MAbs 2025; 17:2465391. [PMID: 39950649 PMCID: PMC11834420 DOI: 10.1080/19420862.2025.2465391] [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: 11/26/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) can be functionally enhanced via Fc engineering. To determine whether pairs of mAbs with different Fc modifications can be combined for functional complementarity, we investigated the in vitro activity of two HIV-1 mAb libraries, each equipped with 60 engineered Fc variants. Our findings demonstrate that the impact of Fc engineering on Fc functionality is dependent on the specific Fab clone. Notably, combinations of Fc variants of the same Fab specificity exhibited limited enhancement in functional breadth compared to combinations involving two distinct Fabs. This suggests that the strategic selection of complementary Fc modifications can enhance both functional activity and breadth. Furthermore, while some combinations of Fc variants displayed additive functional effects, others were detrimental, suggesting that the functional outcome of Fc mutations is not easily predicted. Collectively, these results provide preliminary evidence supporting the potential of complementary Fc modifications in mAb combinations. Future studies will be essential to identify the optimal Fc modifications that maximize in vivo efficacy.
Collapse
Affiliation(s)
- Yannic C. Bartsch
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, Massachusetts, USA
- Laboratory of Anti-Viral Antibody-Omics, TWINCORE-Institute for Experimental Infection Research, Helmholtz Center for Infection Research (HZI) and Medical School Hannover (MHH) and Cluster of Excellence RESIST (EXC 2155), Hannover, Germany
| | - Nicholas E. Webb
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Eleanor Burgess
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Jaewon Kang
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, Massachusetts, USA
| | | | - Boris D. Julg
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
11
|
Xu J, Wen J, Ji X, Chen J, Yang M, Hong M, Deng D. Preclinical development of a high affinity anti-exatecan monoclonal antibody and application in bioanalysis of antibody-exatecan conjugates. J Pharm Biomed Anal 2025; 262:116843. [PMID: 40209496 DOI: 10.1016/j.jpba.2025.116843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/11/2025] [Accepted: 03/24/2025] [Indexed: 04/12/2025]
Abstract
Exatecan, a topoisomerase I inhibitor, is currently utilized as a potent payload in antibody-drug conjugates, significantly enhances the efficacy and safety of these therapeutic agents. In the research of antibody-drug conjugates with exatecan as the payload conjugation, an anti-exatecan antibody serves as a crucial reagent for bioanalysis. In this study, BALB/c mice were immunized with bovine serum albumin conjugate exatecan (BSA-exatecan), and hybridoma technology was employed to screen seven hybridoma cell lines that stably express monoclonal antibodies. After evaluating their binding activity to exatecan, the cell line NO. 8B5-3H6 has been selected based on the EC50 value. The antibody was purified using protein A affinity chromatography, resulting in a mouse anti-exatecan monoclonal antibody with a purity exceeding 99 %. The binding profile with the exatecan demonstrated strong affinity, with an EC50 of 1.382. Bio-Layer Interferometry (BLI) analysis further confirmed the high affinity of this mouse anti-exatecan antibody with a KD of less than 1 pM. Subsequently a detection method was developed using the mouse anti-exatecan antibody as the coating reagent and mouse anti-human IgG Fab conjugate HRP as the detection reagent. The standard curve and quantification range of the method were established at 31.25 ng/mL to 4000 ng/mL. Validation of accuracy, precision, selectivity, stability, dilution linearity, hook effect, parallelism and specificity were performed in accordance with ICH M10 and FDA bioanalytical method validation guidelines, laying a solid foundation for subsequent toxicological and pharmacokinetic studies of antibody-drug conjugate.
Collapse
Affiliation(s)
- Junshuang Xu
- China Pharmaceutical University, No. 639, Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province 211198, China
| | - Jing Wen
- China Pharmaceutical University, No. 639, Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province 211198, China
| | - Xiaobo Ji
- Shanghai Junshi Biosciences Co., Ltd. (TopAlliance), East of Gate 7, Yunchuang Road, Wujiang District, Suzhou, Jiangsu Province, China
| | - Jieru Chen
- Shanghai Junshi Biosciences Co., Ltd. (TopAlliance), East of Gate 7, Yunchuang Road, Wujiang District, Suzhou, Jiangsu Province, China
| | - Meiyu Yang
- Shanghai Junshi Biosciences Co., Ltd. (TopAlliance), East of Gate 7, Yunchuang Road, Wujiang District, Suzhou, Jiangsu Province, China
| | - Min Hong
- Shanghai Junshi Biosciences Co., Ltd. (TopAlliance), East of Gate 7, Yunchuang Road, Wujiang District, Suzhou, Jiangsu Province, China.
| | - Dawei Deng
- China Pharmaceutical University, No. 639, Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province 211198, China
| |
Collapse
|
12
|
Pulido I, Luan Q, Pastor-Puente S, Gunder L, Wang Y, Ying C, Li J, Sun Y, Dai Y, Ascoli C, Abdelhady K, Massad M, Prince TL, Wang G, Foley KP, Ying W, Papautsky I, Carretero J, Shimamura T. Chaperone directed heterobifunctional molecules circumvent KRAS G12C inhibitor resistance. Cancer Lett 2025; 622:217691. [PMID: 40204148 DOI: 10.1016/j.canlet.2025.217691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
While KRASG12C inhibitors have shown promising results in clinical activity, acquired resistance remains a significant barrier to durable responses. Combination therapies have been explored to improve the efficacy of KRASG12C inhibitors; however, their use is often restricted due to toxicity and limitations in clinically amenable dosing schedules. Transcriptomic profiling and functional assays on acquired resistant models to adagrasib identified an enrichment of HSP90 client proteins in resistant phenotypes, suggesting a therapeutic vulnerability. To address the finding, RNK07421, a novel heterobifunctional molecule, was developed to simultaneously target KRASG12C and HSP90-client oncoproteins. Structural and biochemical analyses demonstrated that RNK07421 disrupts KRASG12C interactions by inducing a non-natural interface with HSP90, thereby impairing oncogenic signaling. In vitro, RNK07421 effectively suppressed ERK reactivation and reduced viability in KRASG12C-mutant cell lines exhibiting either intrinsic or acquired resistance. In vivo, RNK07421 significantly reduced tumor burden in xenograft models, outperforming both monotherapies and combination therapies. These findings highlight dual KRASG12C and HSP90 inhibition as a promising strategy to overcome resistance in KRASG12C-driven cancers.
Collapse
Affiliation(s)
- Ines Pulido
- Department of Surgery, Division of Cardiothoracic Surgery, University of Illinois Chicago, Chicago, IL, 60612, USA; University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Qiyue Luan
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Sara Pastor-Puente
- Department of Ophthalmology and Visual Science, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Laura Gunder
- Department of Surgery, Division of Cardiothoracic Surgery, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Yaya Wang
- Ranok Therapeutics, Waltham, MA, 02451, USA; Ranok Therapeutics, Hangzhou, 310020, China
| | - Chenghao Ying
- Ranok Therapeutics, Waltham, MA, 02451, USA; Ranok Therapeutics, Hangzhou, 310020, China
| | - Jinhua Li
- Ranok Therapeutics, Waltham, MA, 02451, USA; Ranok Therapeutics, Hangzhou, 310020, China
| | - Yuetong Sun
- Ranok Therapeutics, Waltham, MA, 02451, USA; Ranok Therapeutics, Hangzhou, 310020, China
| | - Yan Dai
- Ranok Therapeutics, Waltham, MA, 02451, USA; Ranok Therapeutics, Hangzhou, 310020, China
| | - Christian Ascoli
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Khaled Abdelhady
- Department of Surgery, Division of Cardiothoracic Surgery, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Malek Massad
- Department of Surgery, Division of Cardiothoracic Surgery, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Thomas L Prince
- Ranok Therapeutics, Waltham, MA, 02451, USA; Ranok Therapeutics, Hangzhou, 310020, China
| | - Guoqiang Wang
- Ranok Therapeutics, Waltham, MA, 02451, USA; Ranok Therapeutics, Hangzhou, 310020, China
| | - Kevin P Foley
- Ranok Therapeutics, Waltham, MA, 02451, USA; Ranok Therapeutics, Hangzhou, 310020, China
| | - Weiwen Ying
- Ranok Therapeutics, Waltham, MA, 02451, USA; Ranok Therapeutics, Hangzhou, 310020, China
| | - Ian Papautsky
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Julian Carretero
- Department of Physiology, Universitat de Valencia, Valencia, 46100, Spain
| | - Takeshi Shimamura
- Department of Surgery, Division of Cardiothoracic Surgery, University of Illinois Chicago, Chicago, IL, 60612, USA; University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
13
|
Jiang M, Ma S, Xuan Y, Chen K. Synthetic approaches and clinical application of KRAS inhibitors for cancer therapy. Eur J Med Chem 2025; 291:117626. [PMID: 40252381 DOI: 10.1016/j.ejmech.2025.117626] [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: 03/18/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/21/2025]
Abstract
Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations are among the most common oncogenic alterations in various cancers, including pancreatic, colorectal, and non-small cell lung cancer (NSCLC). Targeting KRAS has long been considered a difficult challenge due to its high affinity for guanosine triphosphate (GTP) and the lack of a druggable binding site. However, recent advancements in small-molecule inhibitor design have led to the development of targeted therapies aimed at KRAS mutations, particularly the KRASG12C mutation. Inhibitors such as Sotorasib and Adagrasib have shown promise in preclinical and clinical studies by irreversibly binding to the mutant KRAS protein, locking it in an inactive state and disrupting downstream signaling pathways critical for tumor growth and survival. These inhibitors have demonstrated clinical efficacy in treating patients with KRASG12C-mutated cancers, leading to tumor regression, prolonged progression-free survival, and improved patient outcomes. This review discusses the synthetic strategies employed to develop these KRAS inhibitor and also examines the clinical application of these inhibitors, highlighting the challenges and successes encountered during clinical trials. Ultimately, KRAS inhibitors represent a breakthrough in cancer therapy, offering a promising new treatment option for patients with KRAS-driven tumors.
Collapse
Affiliation(s)
- Min Jiang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Shaowei Ma
- Department of Interventional Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ying Xuan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Kuanbing Chen
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
14
|
Desai O, Rathore M, Boutros CS, Wright M, Bryson E, Curry K, Wang R. HER3: Unmasking a twist in the tale of a previously unsuccessful therapeutic pursuit targeting a key cancer survival pathway. Genes Dis 2025; 12:101354. [PMID: 40290122 PMCID: PMC12022662 DOI: 10.1016/j.gendis.2024.101354] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 04/30/2025] Open
Abstract
HER3, formally referred to as ERB-B2 receptor tyrosine kinase 3, is a member of the ErbB receptor tyrosine kinases (also known as EGFR) family. HER3 plays a significant pro-cancer role in various types of cancer due to its overexpression and abnormal activation, which initiates downstream signaling pathways crucial in cancer cell survival and progression. As a result, numerous monoclonal antibodies have been developed to block HER3 activation and subsequent signaling pathways. While pre-clinical investigations have effectively showcased significant anti-cancer effects of HER3-targeted therapies, these therapies have had little impact on cancer patient outcomes in the clinic, except for patients with rare NRG1 fusion mutations. This review offers a comprehensive description of the oncogenic functions of HER3, encompassing its structure and mediating signaling pathways. More importantly, it provides an in-depth exploration of past and ongoing clinical trials investigating HER3-targeted therapies for distinct types of cancer and discusses the tumor microenvironment and other critical determinants that may contribute to the observed suboptimal outcomes in most clinical studies using HER3-targeted therapies. Lastly, we suggest alternative approaches and the exploration of novel strategies to potentially improve the efficacy of targeting the pivotal oncogenic HER3 signaling pathway in future translational investigations.
Collapse
Affiliation(s)
- Omkar Desai
- Department of Surgery, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Moeez Rathore
- Department of Surgery, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Christina S. Boutros
- Department of Surgery, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Michel'le Wright
- Department of Surgery, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Elizabeth Bryson
- Department of Surgery, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Kimberly Curry
- Department of Surgery, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Rui Wang
- Department of Surgery, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
15
|
Xiao X, Huang L, Li M, Zhang Q. Intersection between lung cancer and neuroscience: Opportunities and challenges. Cancer Lett 2025; 621:217701. [PMID: 40194655 DOI: 10.1016/j.canlet.2025.217701] [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: 11/22/2024] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/09/2025]
Abstract
Lung cancer, which has the highest morbidity and mortality rates worldwide, involves intricate interactions with the nervous system. Research indicates that the nervous system not only plays a role in the origin of lung cancer, but also engages in complex interactions with cancer cells through neurons, neurotransmitters, and various neuroactive molecules during tumor proliferation, invasion, and metastasis, especially in brain metastases. Cancer and its therapies can remodel the nervous system. Despite advancements in immunotherapy and targeted therapies in recent years, drug resistance of lung cancer cells after treatment limits improvements in patient survival and prognosis. The emergence of neuroscience has created new opportunities for the treatment of lung cancer. However, it also presents challenges. This review emphasizes that a deeper understanding of the interactions between the nervous system and lung cancer, along with the identification of new therapeutic targets, may lead to significant advancements or even a revolution in treatment strategies for patients with lung cancer.
Collapse
Affiliation(s)
- Xiang Xiao
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, Jiangsu, 210009, PR China; The Fourth Clinical College of Nanjing Medical University, Nanjing, Jiangsu, 210009, PR China
| | - Lingli Huang
- The Fourth Clinical College of Nanjing Medical University, Nanjing, Jiangsu, 210009, PR China; Department of Pharmacy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, PR China
| | - Ming Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, Jiangsu, 210009, PR China; The Fourth Clinical College of Nanjing Medical University, Nanjing, Jiangsu, 210009, PR China.
| | - Quanli Zhang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, Jiangsu, 210009, PR China; The Fourth Clinical College of Nanjing Medical University, Nanjing, Jiangsu, 210009, PR China.
| |
Collapse
|
16
|
Hu Y, Du X, Yuan J, Gong X, Zhu Y, Li H, Lin X, Zheng F, Ran Y, Na Z, Hu H. A high-affinity antibody-drug conjugates Actuximab-MMAE for potent and selective targeting of CEACAM5-Positive tumors. Cancer Lett 2025; 620:217685. [PMID: 40158720 DOI: 10.1016/j.canlet.2025.217685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
Antibody-drug conjugates (ADCs) represent a promising class of anti-cancer therapy with an increasingly critical role in treating various tumors. They broaden the range of therapeutic targets, enabling the consideration of tumor-associated proteins that are overexpressed but lack well-defined mechanisms. Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) is a clinically relevant screening marker due to its tumor-specific overexpression, making it an attractive target for ADC development. However, the therapeutic potential of earlier anti-CEACAM5 ADCs has been limited by side effects and suboptimal drug-to-antibody ratios (DARs), restricting their clinical utility. In this study, we developed a novel anti-CEACAM5 ADC (named Actuximab-MMAE), characterized by high affinity, an optimized DAR, and potent tumor-selective cytotoxicity. Actuximab-MMAE demonstrated rapid and effective elimination of CEACAM5-positive tumors in vivo at low doses, while maintaining a favorable safety profile. These findings highlight Actuximab-MMAE as a promising therapeutic option for CEACAM5-overexpressing tumors, offering a new therapeutic method for targeted cancer therapy.
Collapse
Affiliation(s)
- Yuqi Hu
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xin Du
- Breast Cancer Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Jiayu Yuan
- Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Xizhao Gong
- Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Yue Zhu
- Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Hongde Li
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, 310018, China
| | - Xiaorong Lin
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, Shantou, 515000, China
| | - Fang Zheng
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yuliang Ran
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Zhenkun Na
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, 310018, China.
| | - Hai Hu
- Breast Cancer Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, China; Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, 310022, China; Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, 310018, China.
| |
Collapse
|
17
|
Goloudina A, Le Chevalier F, Authié P, Charneau P, Majlessi L. Shared neoantigens for cancer immunotherapy. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200978. [PMID: 40256120 PMCID: PMC12008704 DOI: 10.1016/j.omton.2025.200978] [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] [Indexed: 04/22/2025]
Abstract
Exploration of neoantigens holds the potential to be productive in immuno-oncotherapy. Among tumor-specific antigens, neoantigens result from genetic instability that gives rise to non-synonymous somatic mutations, highly specific to tumor cells. In addition to point mutations, gene rearrangements, indels leading to frameshifts, chromosomal translocations or inversions that may lead to fusion proteins, alternative mRNA splicing, and integration of genetic material of oncogenic viruses into the host genome provide consistent sources of neoantigens that are absent in healthy tissues. Out of these alterations, 2%-3% may generate T cell neoepitopes, possibly detectable by TCRs. Neoantigens are absent in healthy tissues and are thus at low risk of triggering autoimmunity. In addition, the host lymphocytes have not been rendered tolerant toward them and it is possible to induce immune responses against them. Here, we overview the two categories of neoantigens, i.e., private and shared, and their use in immuno-oncotherapy in selected pre-clinical and clinical studies. The vast majority of commonly occurring tumor-specific mutations are cancer causing and are permanently expressed by all malignant tumor cells, preventing the latter from escaping vaccine-induced anti-neoantigen immunity. The use of public neoantigens combined with efficient vaccine platforms can provide non-personalized "off-the-shelf" therapeutic vaccine candidates for broad-spectrum immunotherapy purposes.
Collapse
Affiliation(s)
- Anastasia Goloudina
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Fabien Le Chevalier
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Pierre Authié
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Pierre Charneau
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Laleh Majlessi
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| |
Collapse
|
18
|
Hemida N, El-Gamil DS, ElHady AK, Lin KC, Chang YH, Hilscher S, Schutkowski M, Ibrahim HS, Hamed MM, Chen SH, Chen CH, Abadi AH, Sippl W, Chen PJ, Cheng YS, Abdel-Halim M. Unlocking the potential of novel tetrahydro-β-carboline-based HDAC6 inhibitors for colorectal cancer therapy: Design, synthesis and biological evaluation. Bioorg Chem 2025; 160:108454. [PMID: 40252366 DOI: 10.1016/j.bioorg.2025.108454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/21/2025] [Accepted: 04/08/2025] [Indexed: 04/21/2025]
Abstract
Altered histone deacetylase 6 (HDAC6) expression and function have been linked to cancer progression, positioning it as a promising therapeutic target for cancer treatment. Herein, we introduce HDAC6 inhibitors based on the tetrahydro-β-carboline scaffold, with compound 18d exhibiting the strongest HDAC6 inhibitory potency, achieving an IC50 of 1.3 nM. Compound 18d exhibited significant growth inhibitory activity against an NCI panel of 60 human cancer cell lines with a minimal cytotoxic effect on non-tumor cells. In vitro mechanistic investigations were conducted in HCT-116 colorectal cancer cells where the capability of 18d to enhance the acetylation of α-tubulin (HDAC6 substrate) rather than nuclear H3 histone (HDAC1 substrate) confirmed selective inhibition of HDAC6 subtype. Additionally, compound 18d was observed to suppress the S phase and promote accumulation in the apoptotic sub-G1 phase, potentially through increasing cleaved caspase 3 and reducing Bcl-2 levels in HCT-116 cells. A wound healing assay also elicited the ability of 18d to hinder cell migration. Notably, 18d could suppress the phosphorylation of extracellular signal-regulated kinase (ERK)1/2, a crucial signaling pathway implicated in cancer cell proliferation, migration and apoptosis. Moreover, downregulation of the critical immune checkpoint protein programmed death-ligand 1 (PD-L1) revealed a potential role of 18d in augmenting immune response towards tumor cells. In summary, these findings highlight 18d's dual role in direct tumor growth suppression and immune system sensitization, highlighting a broader cancer therapeutic potential beyond conventional HDAC inhibition.
Collapse
Affiliation(s)
- Noreen Hemida
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Dalia S El-Gamil
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt; Department of Chemistry, Faculty of Pharmacy, Ahram Canadian University, Cairo 12451, Egypt
| | - Ahmed K ElHady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt; School of Life & Medical Sciences, University of Hertfordshire hosted by Global Academic Foundation, New Administrative Capital, Cairo, Egypt
| | - Kai-Chun Lin
- Institute of Plant Biology, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Yen-Hua Chang
- Institute of Plant Biology, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Sebastian Hilscher
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| | - Mike Schutkowski
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| | - Hany S Ibrahim
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| | - Mostafa M Hamed
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123 Saarbrücken, Germany
| | - Shun-Hua Chen
- School of Nursing, Fooyin University, Kaohsiung 831301, Taiwan
| | - Chun-Hong Chen
- Department of Medical Research, E-Da Hospital and Graduate Institute of Medicine, I-Shou University, Kaohsiung 824410, Taiwan
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Wolfgang Sippl
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| | - Po-Jen Chen
- Department of Medical Research, E-Da Hospital and Graduate Institute of Medicine, I-Shou University, Kaohsiung 824410, Taiwan; Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Yi-Sheng Cheng
- Institute of Plant Biology, College of Life Science, National Taiwan University, Taipei 10617, Taiwan; Department of Life Science, College of Life Science, National Taiwan University, Taipei 10617, Taiwan; Genome and Systems Biology Degree Program, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt.
| |
Collapse
|
19
|
Jin C, Emam M, Klauck SM, Ali NT, Salem R, Eldehna WM, Efferth T, Hegazy MEF, Dawood M. Targeting sensitive and multidrug resistant leukemia cells with a novel benzofuran-isatin conjugate. Eur J Pharmacol 2025; 997:177538. [PMID: 40122501 DOI: 10.1016/j.ejphar.2025.177538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Benzofuran-isatin conjugates are considered as promising compounds in cancer prevention and treatment. However, it is not known yet whether these compounds are useful to effectively treat multidrug-resistant tumors. In this study, we investigated the activity of G-5e, a novel benzofuran-isatin conjugate in a panel of cell lines exhibiting well-known drug resistance mechanisms (P-gp, BCRP, TP53, EGFR). P-glycoprotein overexpressing CEM/ADR5000 cell line displayed notable hypersensitivity (collateral sensitivity) to G-5e, which was mediated through autophagic cell death activation including downregulation of the autophagy suppressor RND2, upregulation of the autophagy inducer LC3B, and G0/G1 phase arrest during cell cycle progression. Independent of collateral sensitivity, transcriptomic analyses also revealed that G-5e caused downregulation of NF-κB and ERK1/2 pathways. Our findings highlight the potential of benzofuran-isatin conjugates to combat multidrug resistance and the role of RND2 for collateral sensitivity.
Collapse
Affiliation(s)
- Chunmei Jin
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences Chemistry, Johannes Gutenberg University, Staudinger Weg 5, 55128, Mainz, Germany
| | - Mahmoud Emam
- Phytochemistry and Plant Systematics Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Sabine M Klauck
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ) Heidelberg, National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Nadeen T Ali
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences Chemistry, Johannes Gutenberg University, Staudinger Weg 5, 55128, Mainz, Germany
| | - Rofaida Salem
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, P.O. Box 33516, Kafrelsheikh, Egypt
| | - Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, P.O. Box 33516, Kafrelsheikh, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia St., Alexandria, 21648, Egypt
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences Chemistry, Johannes Gutenberg University, Staudinger Weg 5, 55128, Mainz, Germany
| | - Mohamed-Elamir F Hegazy
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences Chemistry, Johannes Gutenberg University, Staudinger Weg 5, 55128, Mainz, Germany; Chemistry of Medicinal Plants Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza, 12622, Egypt
| | - Mona Dawood
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences Chemistry, Johannes Gutenberg University, Staudinger Weg 5, 55128, Mainz, Germany.
| |
Collapse
|
20
|
Hossain MT, Hossain MA. Targeting PI3K in cancer treatment: A comprehensive review with insights from clinical outcomes. Eur J Pharmacol 2025; 996:177432. [PMID: 40020984 DOI: 10.1016/j.ejphar.2025.177432] [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: 11/16/2024] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
The phosphoinositide 3-kinase (PI3K) pathway plays a crucial role in cancer, including cell growth, survival, metabolism, and metastasis. Its major role in tumor growth makes it a key target for cancer therapeutics, offering significant potential to slow tumor progression and enhance patient outcomes. Gain-of-function mutations, gene amplifications, and the loss of regulatory proteins like PTEN are frequently observed in malignancies, contributing to tumor development and resistance to conventional treatments such as chemotherapy and hormone therapy. As a result, PI3K inhibitors have received a lot of interest in cancer research. Several kinds of small-molecule PI3K inhibitors have been developed, including pan-PI3K inhibitors, isoform-specific inhibitors, and dual PI3K/mTOR inhibitors, each targeting a distinct component of the pathway. Some PI3K inhibitors such as idelalisib, copanlisib, duvelisib, alpelisib, and umbralisib have received FDA-approval, and are effective in the treatment of breast cancer and hematologic malignancies. Despite promising results in preclinical and clinical trials, the overall clinical success of PI3K inhibitors has been mixed. While some patients may get substantial advantages, a considerable number of them acquire resistance as a result of feedback activation of alternative pathways, adaptive tumor responses, and treatment-emergent mutations. The resistance mechanisms provide barriers to the sustained efficacy of PI3K-targeted treatments. This study reviews recent advancements in PI3K inhibitors, covering their clinical status, mechanism of action, resistance mechanisms, and strategies to overcome resistance.
Collapse
Affiliation(s)
- Md Takdir Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| | - Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| |
Collapse
|
21
|
Zhou G, Dong K, Xu X, Guo R, Li G, Wang J, Zhou L, Yuan S, Lou H, Li H, Dong H, Peng X. Fungal dimeric xanthones as anticancer agents by novelly stimulating sodium-calcium exchanger 1. Eur J Med Chem 2025; 290:117543. [PMID: 40153930 DOI: 10.1016/j.ejmech.2025.117543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025]
Abstract
Multitude of natural products have the ability to demonstrate inhibitory effects on cancer cells by regulating ion channels/transporters functions. Eighteen xanthone dimers (Xds), including five new dimers diaporxanthones H, I, J-L (1, 2, and 12-14), were isolated and characterized through co-culture and chemical conversion methods. ECD Cotton effect analyses and chemical communication method provided fundamental role in addressing the challenges of elucidating their absolute configurations. Structure-activity relationship (SAR) analysis showed that eight xanthone-xanthone Xds (2-7, 15 and 16) demonstrated marked cytotoxic effects against gastric cancer (GC) cell line AGS, with undetectable inhibition on human colon cancer cells. The anti-proliferative potency of Xds was 2-5 fold higher than positive control drug cisplatin. Mechanistic studies were conducted on a high-yield compound, 12-O-deacetyl-phomoxanthone A (4). Compound 4 activated Na+/Ca2+ exchanger 1 (NCX1), thereby causing an increase in cellular Ca2+ signaling and subsequent inhibition of the downstream PI3K/AKT/β-catenin pathway, ultimately leading to GC cell death. Like anti-GC, Xds also possessed anti-melanoma activity in vitro and in vivo. We demonstrate Xds have effective cytotoxic actions against GC and melanoma by targeting NCX1/Ca2+ signaling in cancer cells.
Collapse
Affiliation(s)
- Guolong Zhou
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Kemin Dong
- Department of Natural Medicinal Chemistry and Pharmacognosy, School of Pharmacy, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Xiaoyuan Xu
- Department of Natural Medicinal Chemistry and Pharmacognosy, School of Pharmacy, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Ruihong Guo
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Gang Li
- Department of Natural Medicinal Chemistry and Pharmacognosy, School of Pharmacy, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Jianxin Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Liyong Zhou
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Shuangzhi Yuan
- Key Laboratory of Chemical Biology of Ministry of Education, Department of Natural Product Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, People's Republic of China
| | - Hongxiang Lou
- Department of Natural Medicinal Chemistry and Pharmacognosy, School of Pharmacy, Qingdao University, Qingdao, 266071, People's Republic of China; Key Laboratory of Chemical Biology of Ministry of Education, Department of Natural Product Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, People's Republic of China
| | - Hongmei Li
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, People's Republic of China
| | - Hui Dong
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Xiaoping Peng
- Department of Natural Medicinal Chemistry and Pharmacognosy, School of Pharmacy, Qingdao University, Qingdao, 266071, People's Republic of China.
| |
Collapse
|
22
|
Iizuka A, Kanda M, Sato Y, Shimizu D, Umeda S, Tanaka H, Hattori N, Hayashi M, Tanaka C, Kodera Y. Association of NR0B1 with Malignant Phenotypes in Esophageal Squamous Cell Carcinoma Through Modulation of p53-Independent Cell-Cycle Regulation. Ann Surg Oncol 2025; 32:4464-4475. [PMID: 40080367 DOI: 10.1245/s10434-025-17109-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 02/17/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Transcriptome analysis of primary tumor tissues from esophageal squamous cell carcinoma (ESCC) patients with early postoperative distant metastasis identified nuclear receptor subfamily 0, group B, member 1 (NR0B1) as a novel gene associated with the malignant phenotypes of ESCC. This study aimed to elucidate the oncological functions of NR0B1 in ESCC and assess the significance of its tissue expression. METHODS We investigated the effects of NR0B1 knockdown on the proliferation, migration, and adhesion capacities, in vivo tumor growth, and intracellular signaling pathways of ESCC cell lines. The correlation between tissue NR0B1 expression at both the mRNA and protein levels and postoperative prognosis was analyzed by using two independent cohorts. RESULTS Silencing NR0B1 significantly inhibited the proliferation, migration, and adhesion capacities of ESCC cell lines and decreased tumor growth in mouse cell line derived xenograft models. Knockdown of NR0B1 results in the upregulation of cell cycle regulators p21 and p27, alongside the downregulation of TK1, cyclin E1, CDK2 and CDT1, in a manner independent of p53. Although elevated tissue NR0B1 expression did not show a significant association with TNM stage, it was identified as an independent prognostic factor at both the mRNA and protein levels across two distinct patient cohorts. CONCLUSIONS NR0B1 plays a critical role in the malignant phenotype of ESCC by modulating cell cycle regulators. Tissue NR0B1 expression may serve as a valuable biomarker for assessing prognostic risk in ESCC patients.
Collapse
Affiliation(s)
- Akimitsu Iizuka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Yusuke Sato
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Dai Shimizu
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinichi Umeda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Haruyoshi Tanaka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norifumi Hattori
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masamichi Hayashi
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Chie Tanaka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
23
|
Subbiah V, Othus M, Palma J, Cuglievan B, Kurzrock R. Designing Clinical Trials for Patients With Rare Cancers: Connecting the Zebras. Am Soc Clin Oncol Educ Book 2025; 45:e100051. [PMID: 40228175 DOI: 10.1200/edbk-25-100051] [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: 04/16/2025]
Abstract
The field of rare cancer research is rapidly transforming, marked by significant progress in clinical trials and treatment strategies. Rare cancers, as defined by the National Cancer Institute, occur in fewer than 150 cases per million people each year, yet they collectively represent a significant portion of all cancer diagnoses. Because of their infrequency, these cancers pose distinct challenges for clinical trials, including limited patient populations, geographical dispersion, and a general lack of awareness of treatment options. Economic limitations further complicate drug development, making initiatives such as the Orphan Drug Act essential for incentivizing research. The advent of next-generation sequencing (NGS) and precision medicine has been instrumental in identifying actionable genetic alterations in parallel with an explosion in the development of genomically targeted therapies, immunotherapies, and antibody drug conjugates. Advances in clinical NGS, precision medicine, and tumor-agnostic therapies have become central to the progress in rare cancer research. The development and approval of tumor-agnostic drugs, such as BRAF, NTRK, and RET inhibitors, and immunotherapy for mismatch repair deficient/microsatellite instability-high status cancers highlight the potential of personalized treatments across diverse cancer types and across the age spectrum. Collaborative trials from cooperative groups including SWOG DART, ASCO TAPUR, NCI-MATCH, pediatric COG-match, DRUP, IMPRESS, and innovative registrational basket and platform trials (eg, VE-Basket, ROAR, LIBRETTO-001, ARROW), along with patient advocacy group-run trials like TRACK, are enhancing access to clinical trials. In addition, artificial intelligence has the potential to improve the trial matching process. An integrated approach, combining these innovations in collaboration with multiple stakeholders, is crucial for advancing rare cancer research, offering hope for better patient outcomes and quality of life.
Collapse
Affiliation(s)
| | - Megan Othus
- SWOG Cancer Research Network Statistical Center, Seattle, WA
- Division of Public Health, Fred Hutchinson Cancer Center, Seattle, WA
| | - Jim Palma
- TargetCancer Foundation, Rare Cancer Patient Advocacy Group, Cambridge, MA
| | - Branko Cuglievan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Razelle Kurzrock
- Genomic Sciences and Precision Medicine Center, and Medical College of Wisconsin Cancer Center, Milwaukee, WI
- WIN Consortium, Paris, France
- University of Nebraska, Lincoln, NE
| |
Collapse
|
24
|
Nakazato Y, Hirano K, Mitsuma T, Arimasu Y, Hirokawa T, Chiba T, Fujiwara M, Tanaka R, Kondo H, Kamma H. Regulatory SNP of TERT promoter accompanied by C228T and BRAFV 600E is an exacerbating factor of papillary thyroid carcinoma. Oncol Lett 2025; 29:267. [PMID: 40235685 PMCID: PMC11997643 DOI: 10.3892/ol.2025.15013] [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: 05/15/2024] [Accepted: 10/09/2024] [Indexed: 04/17/2025] Open
Abstract
Despite the increased incidence of thyroid cancer due to enhanced precision of ultrasound technology and extensive utilization of puncture aspiration cytology, the mortality rate remains low, raising concerns about overdiagnosis. Papillary thyroid carcinoma (PTC) is the most common type, primarily diagnosed through cell nuclei examination. Recent advancements in identifying genetic mutations and tumor classification have refined diagnostic methods. Point mutations in the telomerase reverse transcriptase promoter (TERTp), specifically -124 C >T (C228T) and -146 C >T (C250T), and the regulatory single nucleotide polymorphism -245 T >C, C allele of rs2853669 (TrSNP) are potential thyroid cancer biomarkers. The present study tested the hypothesis that the coexistence of BRAF mutations in driver genes upstream of the MAPK pathway and late mutations unrelated to signaling, such as point mutations in TERTp, increases tumor virulence. A total of 133 patients with PTC who underwent surgery between January 2014 and November 2021 were included in the study. Blood and tumor tissue samples were collected, and DNA was extracted for genetic mutation analysis using PCR and Sanger sequencing. The TrSNP analysis of blood and surgical tissue samples showed a 97.7% agreement rate. TrSNP was detected in 70 of 133 patients (52.6%) and was significantly associated with tumor size, particularly in tumors >2.0 cm. TERTp point mutations were identified in 29 of 133 patients (21.8%), with C228T strongly associated with tumor size, particularly in tumors >4.0 cm, and extraglandular invasion. BRAF V600E was detected in 82 patients (61.7%) but showed no significant association with clinicopathological parameters. However, the coexistence of BRAF V600E with C228T and TrSNP affected tumor size and progression. The findings indicated that TrSNPs, along with C228T and BRAF V600E, may serve as potential molecular markers to predict PTC growth or exacerbation. Notably, coexistence of C228T and TrSNP is a preoperative indicator of disease progression.
Collapse
Affiliation(s)
- Yoko Nakazato
- Department of General Thoracic and Thyroid Surgery, Kyorin University, School of Medicine, Tokyo 181-8611, Japan
| | - Koichi Hirano
- Department of General Thoracic and Thyroid Surgery, Kyorin University, School of Medicine, Tokyo 181-8611, Japan
| | - Tomoya Mitsuma
- Department of General Thoracic and Thyroid Surgery, Kyorin University, School of Medicine, Tokyo 181-8611, Japan
| | - Yu Arimasu
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine Research Institute, Tokyo 162-8655, Japan
| | | | - Tomohiro Chiba
- Department of Pathology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 112-0012, Japan
| | - Masachika Fujiwara
- Department of Pathology, Kyorin University School of Medicine, Tokyo 181-8611, Japan
| | - Ryota Tanaka
- Department of General Thoracic and Thyroid Surgery, Kyorin University, School of Medicine, Tokyo 181-8611, Japan
| | - Haruhiko Kondo
- Department of General Thoracic and Thyroid Surgery, Kyorin University, School of Medicine, Tokyo 181-8611, Japan
| | - Hiroshi Kamma
- Nasu Institute of Medical Sciences, Kamma Memorial Hospital, Nasushiobara, Tochigi 325-0046, Japan
| |
Collapse
|
25
|
Bokhary RY. Prevalence of HER2 expression and its association with clinicopathological parameters in gastric and gastroesophageal junction adenocarcinoma: A 10?year experience of an academic center. Mol Clin Oncol 2025; 22:49. [PMID: 40242368 PMCID: PMC12001012 DOI: 10.3892/mco.2025.2844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
HER2 overexpression is a marker for targeted therapy in adenocarcinoma of the gastroesophageal junction (GEJ) and stomach. The present study aimed to evaluate the frequency of HER2 overexpression with reference to clinicopathological characteristics in subjects from King Abdulaziz University Hospital, Jeddah, Saudi Arabia over a 10-year period. A retrospective cross-sectional study was conducted on all biopsy and resection specimens diagnosed with either gastric cancer (GC) or GEJ adenocarcinomas from patients between January 2014 and December 2023 that had a final pathology report. Demographic characteristics of 122 patients, including age and sex, were collected, along with pathological details such as tumor grade, histological subtype and HER2 status. χ2 test was used to analyze the association between collected clinicopathological characteristics and HER2 status of the tumor. Most patients were aged 40-60 years. Males constituted 66% of the patients, and the ethnic distribution between Saudi and non-Saudi was almost equal. The most common subtype of cancer was the intestinal type (49%), and the majority of cases were poorly differentiated (64%). HER2 status was assessed in only 61% of cases, with 13.5% showing gene amplification. There was no significant association found between HER2 status and clinicopathological features.
Collapse
Affiliation(s)
- Rana Y. Bokhary
- Department of Pathology, Faculty of Medicine, King Abdulaziz University & King Abdulaziz University Hospital, Jeddah 21589, Saudi Arabia
| |
Collapse
|
26
|
Guo Y, Zhai J, Yang Y, Wei Q, Li S, Huo R, Tong G, Xu E, Chen Y, Han S, Chen D. Long-term survival of an ALK fusion lung adenocarcinoma patient with high mutation burden and microsatellite instability high: a case report. Anticancer Drugs 2025; 36:427-431. [PMID: 39908227 DOI: 10.1097/cad.0000000000001693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
Immune checkpoint blockage (ICB) therapy has shown minimal effectiveness in anaplastic lymphoma kinase (ALK)-positive nonsmall cell lung cancer (NSCLC) regardless of Programmed death-ligand 1 expression. ALK fusion accompanied by mismatch repair deficiency or microsatellite instability-high (MMRd/MSI-H) and high tumor mutation burden (TMB-H) are extremely rare in NSCLC, and the efficacy of ALK inhibitors or ICB-based therapies is unclear. Here, we report the case of a 60-year-old female patient with metastatic lung adenocarcinoma accompanied by EML4-ALK fusion, TMB-H, MMRd/MSI-H, and pathogenic mutations in TP53, MLH1, and STK11. The patient experienced progression on initial iruplinalkib and subsequent alectinib therapy within 5 months. After the failure of third-line therapy with cisplatin-pemetrexed combined with bevacizumab, she received sintilimab plus anlotinib which led to a progression-free survival of 6.5 months. She received sintilimab combined with albumin-paclitaxel plus carboplatin and achieved partial response after 6 months. She developed adverse events after one cycle of sintilimab plus albumin-paclitaxel treatment. Then she was continued with sintilimab plus anlotinib as a maintenance therapy due to intolerance to chemotherapy. After progression on ICB-based therapy, the patient was treated with lorlatinib and still under follow-up with overall survival of more than 3 years. Our findings highlight the therapeutic potential of ICB-based regimens in patients with MSI-H and ALK-rearranged NSCLC.
Collapse
Affiliation(s)
- Yanrong Guo
- Department of Respiratory Diseases, Shanxi Province Cancer Hospital, Taiyuan
| | - Jinfang Zhai
- Department of Respiratory Diseases, Shanxi Province Cancer Hospital, Taiyuan
| | - Yanli Yang
- Department of Respiratory Diseases, Shanxi Province Cancer Hospital, Taiyuan
| | - Qin Wei
- Department of Medicine, Xiamen Spacegen Co., Ltd, Xiamen
| | - Shengshu Li
- Department of Respiratory Diseases, Shanxi Province Cancer Hospital, Taiyuan
| | - Rujie Huo
- Department of Respiratory Diseases, Shanxi Province Cancer Hospital, Taiyuan
| | - Guoping Tong
- Department of Respiratory Diseases, Shanxi Province Cancer Hospital, Taiyuan
| | - Enwei Xu
- Department of Pathology, Shanxi Province Cancer Hospital, Taiyuan, China
| | - Yan Chen
- Department of Medicine, Xiamen Spacegen Co., Ltd, Xiamen
| | - Songyan Han
- Department of Respiratory Diseases, Shanxi Province Cancer Hospital, Taiyuan
| | - Deyi Chen
- Department of Medicine, Xiamen Spacegen Co., Ltd, Xiamen
| |
Collapse
|
27
|
Ma L, Mao JH, Barcellos-Hoff MH. Systemic inflammation in response to radiation drives the genesis of an immunosuppressed tumor microenvironment. Neoplasia 2025; 64:101164. [PMID: 40184664 PMCID: PMC11999686 DOI: 10.1016/j.neo.2025.101164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
The composition of the tumor immune microenvironment has become a major determinant of response to therapy, particularly immunotherapy. Clinically, a tumor microenvironment lacking lymphocytes, so-called "cold" tumors, are considered poor candidates for immune checkpoint inhibition. In this review, we describe the diversity of the tumor immune microenvironment in breast cancer and how radiation exposure alters carcinogenesis. We review the development and use of a radiation-genetic mammary chimera model to clarify the mechanism by which radiation acts. Using the chimera model, we demonstrate that systemic inflammation elicited by a low dose of radiation is key to the construction of an immunosuppressive tumor microenvironment, resulting in aggressive, rapidly growing tumors lacking lymphocytes. Our experimental studies inform the non-mutagenic mechanisms by which radiation affects cancer and provide insight into the genesis of cold tumors.
Collapse
Affiliation(s)
- Lin Ma
- Department of Stomatology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, China
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Mary Helen Barcellos-Hoff
- Department of Radiation Oncology, School of Medicine, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94143 USA.
| |
Collapse
|
28
|
Dong D, Yu X, Liu H, Xu J, Guo J, Guo W, Li X, Wang F, Zhang D, Liu K, Sun Y. Study of immunosenescence in the occurrence and immunotherapy of gastrointestinal malignancies. Semin Cancer Biol 2025; 111:16-35. [PMID: 39929408 DOI: 10.1016/j.semcancer.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/18/2025] [Accepted: 01/26/2025] [Indexed: 02/25/2025]
Abstract
In human beings heterogenous, pervasive and lethal malignancies of different parts of the gastrointestinal (GI) tract viz., tumours of the oesophagus, stomach, small intestine, colon, and rectum, represent gastrointestinal malignancies. Primary treatment modality for gastric cancer includes chemotherapy, surgical interventions, radiotherapy, monoclonal antibodies and inhibitors of angiogenesis. However, there is a need to improve upon the existing treatment modality due to associated adverse events and the development of resistance towards treatment. Additionally, age has been found to contribute to increasing the incidence of tumours due to immunosenescence-associated immunosuppression. Immunosenescence is the natural process of ageing, wherein immune cells as well as organs begin to deteriorate resulting in a dysfunctional or malfunctioning immune system. Accretion of senescent cells in immunosenescence results in the creation of a persistent inflammatory environment or inflammaging, marked with elevated expression of pro-inflammatory and immunosuppressive cytokines and chemokines. Perturbation in the T-cell pools and persistent stimulation by the antigens facilitate premature senility of the immune cells, and senile immune cells exacerbate inflammaging conditions and the inefficiency of the immune system to identify the tumour antigen. Collectively, these conditions contribute positively towards tumour generation, growth and eventually proliferation. Thus, activating the immune cells to distinguish the tumour cells from normal cells and invade them seems to be a logical strategy for the treatment of cancer. Consequently, various approaches to immunotherapy, viz., programmed death ligand-1 (PD-1) inhibitors, Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors etc are being extensively evaluated for their efficiency in gastric cancer. In fact, PD-1 inhibitors have been sanctioned as late late-line therapy modality for gastric cancer. The present review will focus on deciphering the link between the immune system and gastric cancer, and the alterations in the immune system that incur during the development of gastrointestinal malignancies. Also, the mechanism of evasion by tumour cells and immune checkpoints involved along with different approaches of immunotherapy being evaluated in different clinical trials will be discussed.
Collapse
Affiliation(s)
- Daosong Dong
- Department of Pain, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Xue Yu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Molecular Pathology and Epidemiology of Gastric Cancer in the Universities of Liaoning Province, Shenyang, Liaoning 110001, China
| | - Haoran Liu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Jingjing Xu
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Jiayan Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Wei Guo
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiang Li
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Fei Wang
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Dongyong Zhang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Kaiwei Liu
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Yanbin Sun
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
29
|
Gou M, Zhang H, Qian N, Zhang Y, Sun Z, Li G, Wang Z, Dai G. Deep learning radiomics analysis for prediction of survival in patients with unresectable gastric cancer receiving immunotherapy. Eur J Radiol Open 2025; 14:100626. [PMID: 39807092 PMCID: PMC11728962 DOI: 10.1016/j.ejro.2024.100626] [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: 10/28/2024] [Revised: 12/03/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
Objective Immunotherapy has become an option for the first-line therapy of advanced gastric cancer (GC), with improved survival. Our study aimed to investigate unresectable GC from an imaging perspective combined with clinicopathological variables to identify patients who were most likely to benefit from immunotherapy. Method Patients with unresectable GC who were consecutively treated with immunotherapy at two different medical centers of Chinese PLA General Hospital were included and divided into the training and validation cohorts, respectively. A deep learning neural network, using a multimodal ensemble approach based on CT imaging data before immunotherapy, was trained in the training cohort to predict survival, and an internal validation cohort was constructed to select the optimal ensemble model. Data from another cohort were used for external validation. The area under the receiver operating characteristic curve was analyzed to evaluate performance in predicting survival. Detailed clinicopathological data and peripheral blood prior to immunotherapy were collected for each patient. Univariate and multivariable logistic regression analysis of imaging models and clinicopathological variables was also applied to identify the independent predictors of survival. A nomogram based on multivariable logistic regression was constructed. Result A total of 79 GC patients in the training cohort and 97 patients in the external validation cohort were enrolled in this study. A multi-model ensemble approach was applied to train a model to predict the 1-year survival of GC patients. Compared to individual models, the ensemble model showed improvement in performance metrics in both the internal and external validation cohorts. There was a significant difference in overall survival (OS) among patients with different imaging models based on the optimum cutoff score of 0.5 (HR = 0.20, 95 % CI: 0.10-0.37, P < 0.001). Multivariate Cox regression analysis revealed that the imaging models, PD-L1 expression, and lung immune prognostic index were independent prognostic factors for OS. We combined these variables and built a nomogram. The calibration curves showed that the C-index of the nomogram was 0.85 and 0.78 in the training and validation cohorts. Conclusion The deep learning model in combination with several clinical factors showed predictive value for survival in patients with unresectable GC receiving immunotherapy.
Collapse
Affiliation(s)
- Miaomiao Gou
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Hongtao Zhang
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Niansong Qian
- Department of Thoracic Oncology, The Eighth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Yong Zhang
- Department of Medical Oncology, The Second Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Zeyu Sun
- R&D Center, Keya Medical Technology Co., Ltd, Beijing, PR China
| | - Guang Li
- R&D Center, Keya Medical Technology Co., Ltd, Beijing, PR China
| | - Zhikuan Wang
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Guanghai Dai
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| |
Collapse
|
30
|
Noaman I, Gilmour J, Baird A. Asymptomatic oesophageal stent fracture 21 months after insertion. Radiol Case Rep 2025; 20:3031-3036. [PMID: 40230711 PMCID: PMC11994300 DOI: 10.1016/j.radcr.2025.03.020] [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: 11/27/2024] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 04/16/2025] Open
Abstract
Oesophageal stenting is frequently used as a measure of palliative treatment in oesophageal cancer for the relief of dysphagia . With the advent of new modalities of immunotherapy used concurrently with conventional chemotherapy, the survival of patients with oesophageal cancer being treated with palliative intent has lengthened considerably. Consequently, there is higher likelihood of these patients experiencing stent related complications and surviving with them for longer periods of time. In this case report, we discuss a patient who was diagnosed with stent fracture 21 months after initial stent insertion.
Collapse
Affiliation(s)
- Islam Noaman
- Southeast of Scotland Radiology Training Program, Edinburgh, Scotland, United Kingdom
- Ain Shams University, Cairo, Egypt
| | - John Gilmour
- Clinical Radiology Department, Western General Hospital, Edinburgh, Scotland
| | - Andrew Baird
- Clinical Radiology Department, Western General Hospital, Edinburgh, Scotland
| |
Collapse
|
31
|
Li J, Wang D, Su N, Wang M, Wang Y. Immunotherapy benefits PD‑L1‑positive gastric‑type endocervical adenocarcinoma: A multicenter, retrospective study. Mol Clin Oncol 2025; 22:46. [PMID: 40236834 PMCID: PMC11995453 DOI: 10.3892/mco.2025.2841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 01/28/2025] [Indexed: 04/17/2025] Open
Abstract
Gastric-type endocervical adenocarcinoma (GEA) usually exhibits notable aggressiveness and resistance to current therapies. A high expression of programmed death-ligand 1 (PD-L1) was previous reported in GEA and indicated it might benefit from immunotherapy targeting programmed cell death protein 1 (PD-1)/PD-L1. In the present study, the efficacy of immunotherapy in a panel of patients with GEA was explored, aiming to provide the first-hand evidence on this topic. A total of 44 pathologically diagnosed patients with GEA were recruited from the First Affiliated Hospital of Zhengzhou University and the Cancer Hospital of Zhengzhou University. The clinical and pathological information including age, tumor stage, treatments and prognosis were retrieved from our medical records system. Kaplan-Meier analysis was conducted to evaluate the role of immunotherapy on patients' overall survival (OS) and progression-free survival (PFS). According to the treatments, patients with GEA were divided into two groups: The immunotherapy group (n=19) and the non-immunotherapy group (n=25, the control group). In the immunotherapy group, 9 patients received PD-1/PD-L1 inhibitors as part of their primary treatment, while the remaining 10 received it after tumor recurrence/metastasis. Compared with the control group, the use of immunotherapy during primary treatment significantly extended PFS (median PFS: 14 vs. 6 months, P=0.004) and OS (median OS: 24 vs. 16 months, P=0.019). However, in the 10 patients who initiated immunotherapy after tumor recurrence/metastasis, the survival benefits were only observed for OS (median OS: 33.5 vs. 16 months, P=0.013) but not PFS. Furthermore, the efficacy of immunotherapy was more significant in patients with PD-L1-positive GEA than those PD-L1-negative cases, which improved both the PFS (median PFS: 17 vs. 7 months, P=0.002) and OS (median OS: 36 vs. 16 months, P<0.001). This is the first study, to the best of our knowledge, reporting the efficacy of immunotherapy for GEA. It was demonstrated that the earlier use of PD-1/PD-L1 inhibitors was significantly associated with an improved prognosis, and PD-L1 status could predict the response of immunotherapy. These preliminary findings warrant further validations in the future.
Collapse
Affiliation(s)
- Jing Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Dian Wang
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Ning Su
- Department of Gynecologic Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450000, P.R. China
| | - Min Wang
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Yulu Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
32
|
Glushko T, Costello J, Chima R, McGettigan M, Kim R, Jeong D, Qayyum A. Molecular signatures of intrahepatic cholangiocarcinoma: role in targeted therapy selection. Eur J Radiol 2025; 187:112056. [PMID: 40222184 DOI: 10.1016/j.ejrad.2025.112056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 02/08/2025] [Accepted: 03/17/2025] [Indexed: 04/15/2025]
Abstract
Cholangiocarcinoma is a highly lethal disease with a 5-year overall survival rate of 7-20%. A minority of patients present with resectable disease, and relapse rates remain high. Emerging data from next generation sequencing analysis have identified various actionable mutations which drive the different disease courses opening door to precision medicine and targeted therapies. This review focuses on the clinical significance of genetic alterations as well as the role of systemic therapies, immunotherapy and targeted therapies for intrahepatic cholangiocarcinoma.
Collapse
Affiliation(s)
- Tetiana Glushko
- Moffitt Cancer Center, Department of Radiology, 2902 USF Magnolia Drive, Tampa, FL 33612, United States.
| | - James Costello
- Moffitt Cancer Center, Department of Radiology, 2902 USF Magnolia Drive, Tampa, FL 33612, United States
| | - Ranjit Chima
- Moffitt Cancer Center, Department of Radiology, 2902 USF Magnolia Drive, Tampa, FL 33612, United States
| | - Melissa McGettigan
- Moffitt Cancer Center, Department of Radiology, 2902 USF Magnolia Drive, Tampa, FL 33612, United States
| | - Richard Kim
- Moffitt Cancer Center, Department of Radiology, 2902 USF Magnolia Drive, Tampa, FL 33612, United States
| | - Daniel Jeong
- Moffitt Cancer Center, Department of Radiology, 2902 USF Magnolia Drive, Tampa, FL 33612, United States
| | - Aliya Qayyum
- Moffitt Cancer Center, Department of Radiology, 2902 USF Magnolia Drive, Tampa, FL 33612, United States
| |
Collapse
|
33
|
Shao YF, Baca Y, Hinton A, Xiu J, VanderWalde A, Hadfield M, Park SJ, Darabi S, Sato T, Moser JC. Immune Profiling of Uveal Melanoma Liver Metastases and Response to Checkpoint Inhibitors. J Immunother 2025; 48:189-195. [PMID: 40231356 PMCID: PMC12052074 DOI: 10.1097/cji.0000000000000558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/13/2025] [Indexed: 04/16/2025]
Abstract
Responses to immune checkpoint inhibitors (ICIs) differ significantly between uveal melanoma (UM) and cutaneous melanoma (CM) patients. We investigated the immune profile of metastatic UM(mUM) patients and identified markers that are predictive of improved survival. Metastatic liver samples from 189 UM patients and 48 CM patients were analyzed at genomic and transcriptional levels, and survival analysis was performed on a subgroup of 76 ICI-treated mUM patients. UM liver metastases seem to preserve the genomic and immune characteristics of primary UM (pUM), with a low prevalence of ICI markers and high mutation rates of GNA11, GNAQ, BAP1, and SF3B. Compared with mCM, UM liver metastasis showed lower infiltration of several immune cells, but a greater proportion of M2 macrophages. Compared with UM liver metastases, CM liver metastases showed higher expression of G2M checkpoint and EF2 target genes. Among the mUM and mCM samples, expression of G2M and E2F pathway genes was highest in tumors with high TMB values and T-cell inflamed scores. A longer median overall survival (OS) was observed in mUM patients with higher expression of LAG3, HLA class I, or HLA class II; which may represent a small proportion of immune hot tumors. Expression patterns of G2M checkpoint and E2F targets suggest a possible contribution to immunotherapy response.
Collapse
Affiliation(s)
- Yusra F. Shao
- Karmanos Cancer Institute and Wayne State University, Detroit, MI
| | - Yasmine Baca
- Medical Affairs, Caris Life Sciences, Phoenix, AZ
| | | | - Joanne Xiu
- Medical Affairs, Caris Life Sciences, Phoenix, AZ
| | | | | | - Soo J. Park
- Moores Cancer Center, University of California San Diego, La Jolla, CA
| | | | - Takami Sato
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | | |
Collapse
|
34
|
Reiss KA, Soares KC, Torphy RJ, Gyawali B. Treatment Innovations in Pancreatic Cancer: Putting Patient Priorities First. Am Soc Clin Oncol Educ Book 2025; 45:e473204. [PMID: 40173379 DOI: 10.1200/edbk-25-473204] [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: 04/04/2025]
Abstract
Pancreatic adenocarcinoma remains one of the most aggressive and difficult-to-treat solid tumor malignancies, with a high mortality-to-incidence ratio. Globally, pancreatic cancer ranks 12th in terms of incidence but sixth for mortality signifying its aggressive behavior and limited treatment options. While the mortality rates for many other solid tumors have substantially improved over the past few decades, temporal trends in pancreatic cancer mortality rates are quite sobering. In the United States, from 2000 to 2020, the mortality rates from pancreatic cancer have increased, whereas at the same time, mortality rates from other cancers, such as lung, colorectal, or kidney, have fallen appreciably. Is this for lack of treatment innovation? How do we improve survival for patients with pancreatic cancer? In this chapter, we discuss the recent advances and future directions with targeted therapies and immunotherapies in the treatment of pancreatic cancer, and provide the reasons for both optimism and caution for the future of systemic treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Kim A Reiss
- Division of Hematology/Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Kevin C Soares
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Robert J Torphy
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Bishal Gyawali
- Department of Oncology, Queen's University, Kingston, Canada
- Division of Cancer Care and Epidemiology, Queen's University, Kingston, Canada
- Department of Public Health Sciences, Queen's University, Kingston, Canada
| |
Collapse
|
35
|
Kang M, Wang WS, Chang Z. Antibiotic Use at the End of Life: Current Practice and Ways to Optimize. Am J Hosp Palliat Care 2025; 42:610-615. [PMID: 39030663 PMCID: PMC11915761 DOI: 10.1177/10499091241266986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2024] Open
Abstract
Infections are common complications in end of life (EOL). However, clinicians have minimal guidance regarding antibiotic decision-making in EOL care, leading to the overuse of antibiotics. While symptom relief is frequently cited as a major reason for antibiotic use in EOL, antibiotics have not been shown to provide significant improvement in symptoms outside of urinary tract infections. In addition, when prognosis is expected to be in the range of days to weeks, antibiotics have not been shown to provide significant survival benefit. Antibiotics can be beneficial in EOL care in appropriate scenarios, but the current widespread use of antibiotics in EOL requires reevaluation. There needs to be broader efforts to think about antibiotics like other invasive medical procedures in which benefits and risks are weighed, recognizing that not all patients in EOL who receive antibiotics will benefit. In addition, during care planning process, discussing and documenting antibiotic preferences will be beneficial. Non-antibiotic symptom management should be incorporated to plan of care when infection is suspected. Ultimately, the use of antibiotics at EOL should be for the clear benefit for the recipient and should be guided by the type of infection and its clinical course, patients' primary disease and its prognosis, and the preferences of patients or surrogate decision makers.
Collapse
Affiliation(s)
- Minji Kang
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Winnie S. Wang
- Division of General Internal Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Zieanna Chang
- Division of General Internal Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
36
|
Shang Y, He Y, Zhang X, He W, Hua H, Ye F, Zhou X, Li Y, Zhong W, Wu G, Jiang W. Optimization of Immunotherapy Strategies Based on Spatiotemporal Heterogeneity of Tumour-Associated Tissue-Resident Memory T Cells. Immunology 2025; 175:123-133. [PMID: 40114407 PMCID: PMC12052439 DOI: 10.1111/imm.13924] [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: 11/18/2024] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
Tissue-resident memory T cells (TRMs) reside in peripheral tissues and provide rapid immune defence against local infection and tumours. Tumour-associated TRMs share common tissue-resident features and formation mechanisms, representing some unique subsets of tumour-infiltrating lymphocytes (TILs). However, differences in the tumour microenvironment(TME) and tumour evolution stage result in TRMs exhibiting temporal and spatial heterogeneity of phenotype and function not only at different stages, before and after treatment, but also between tumours originating from different tissues, primary and metastatic cancer, and tumour and adjacent normal tissue. The infiltration of TRMs is often associated with immunotherapy response and favourable prognosis; however, due to different definitions, it has been shown that some subtypes of TRMs can also have a negative impact. Therefore, it is crucial to precisely characterise the TRM subpopulations that can influence the therapeutic efficacy and clinical prognosis of various solid tumours. Here, we review the spatiotemporal heterogeneity of tumour-associated TRMs, as well as the differences in their impact on clinical outcomes. We also explore the relationship between TRMs and immune checkpoint blockade (ICB) and TIL therapy, providing insights into potential new targets and strategies for immunotherapy.
Collapse
Affiliation(s)
- Yile Shang
- Department of Colorectal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- College of MedicineZhejiang UniversityHangzhouChina
| | - Yinjun He
- College of MedicineZhejiang UniversityHangzhouChina
| | - Xiang Zhang
- Department of Colorectal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Wenguang He
- Department of Radiology, First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Hanju Hua
- Department of Colorectal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Feng Ye
- Department of Colorectal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Xile Zhou
- Department of Colorectal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Yandong Li
- Department of Colorectal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Weixiang Zhong
- Department of Pathology, First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Guosheng Wu
- Department of Colorectal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Weiqin Jiang
- Department of Colorectal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
37
|
Baez-Navarro X, Groenendijk FH, Oudijk L, von der Thüsen J, Fusco N, Curigliano G, van Deurzen CHM. HER2-low across solid tumours: different incidences and definitions. Pathology 2025; 57:403-414. [PMID: 40221332 DOI: 10.1016/j.pathol.2025.02.003] [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: 11/05/2024] [Revised: 01/22/2025] [Accepted: 02/10/2025] [Indexed: 04/14/2025]
Abstract
Antibody-drug conjugates, particularly trastuzumab deruxtecan (T-DXd), have emerged as effective therapies for various solid tumours. Clinical trials show that T-DXd improves survival in both HER2-positive and HER2-low breast cancer patients. Additionally, it improves survival in HER2-positive gastro-oesophageal cancer and elicits objective responses in HER2-low tumours. Responses have also been noted in lung and gynaecological cancers with HER2 expression, although subgroup analyses for HER2-low cases are lacking. This review assesses HER2 protein expression levels and gene amplification across solid tumours where T-DXd shows potential benefits. We focus on the accuracy and limitations of HER2 testing methods, particularly for identifying HER2-low cancer. A semi-systematic approach was employed, searching EMBASE, Medline, Cochrane, and PubMed databases. We calculated median incidences of HER2-positive, HER2-low, and HER2-0 by immunohistochemistry (IHC), and HER2 amplification by in situ hybridisation (ISH). A total of 144 studies were included, covering breast (n=57), gastro-oesophageal (n=33), lung (n=17), gynaecological (n=24), and various other carcinomas (n=13). The median incidences of HER2-low were 52%, 16%, 58%, and 17% in breast, gastro-oesophageal, endometrial, and ovarian cancers, respectively, with unknown incidences in lung and cervical cancers. Factors influencing HER2-low detection include tumour heterogeneity, antibody clones, observer variability, and lack of validated scoring criteria. Given the significant proportion of HER2-low cases, many patients could benefit from T-DXd, but limitations in detection accuracy necessitate further research and standardisation in diagnostic methods and criteria to advance the clinical utility of T-DXd for HER2-low tumours.
Collapse
Affiliation(s)
- Ximena Baez-Navarro
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands.
| | | | - Lindsey Oudijk
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jan von der Thüsen
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Nicola Fusco
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | | |
Collapse
|
38
|
Shen K, Yuan S, Su N, Tang F, Rehim S, Wang H, Guo H, Zhang Y, Wu Y, Wang H. Monotherapy and combination therapy using antibody‑drug conjugates for platinum‑resistant ovarian cancer. Oncol Rep 2025; 53:68. [PMID: 40242965 PMCID: PMC12046379 DOI: 10.3892/or.2025.8901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Platinum‑resistant ovarian cancer (PROC) is a significant clinical challenge due to the limited number of treatment options and poor outcomes. Moreover, cytotoxic drugs have an unsatisfactory therapeutic efficacy, high toxicity and side effects. An antibody‑drug conjugate (ADC) is a novel cancer therapeutic strategy that combines an antibody, a linker and a payload. ADCs precisely target the tumor cells by binding to the antigen on the surface of tumor cells, thus accurately delivering the cytotoxic drugs and minimizing systemic toxicity. The approval of mirvetuximab soravtansine by the US Food and Drug Administration for treating folate receptor alpha‑positive, platinum‑resistant epithelial ovarian cancer has promoted studies on the use of ADCs in ovarian cancer. A phase III clinical trial showed that mirvetuximab soravtansine achieved an objective remission rate of 42.3% in platinum‑resistant, FRα‑positive ovarian cancer, compared with 15.9% using chemotherapy, demonstrating its immense potential for ADC development. The present review summarizes the research progress on the use of ADCs in PROC as a monotherapy and combination therapy and considers the future development direction of ADCs in PROC.
Collapse
Affiliation(s)
- Ke Shen
- Department of Gynecology and Obstetrics, 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
| | - Shuang Yuan
- Department of Gynecology and Obstetrics, 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
| | - Ning Su
- Department of Gynecology and Obstetrics, 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
- Department of Gynecologic Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Furong Tang
- Department of Gynecology and Obstetrics, 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
| | - Shamsnur Rehim
- Department of Gynecology and Obstetrics, 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
| | - Han Wang
- Department of Gynecology and Obstetrics, 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
| | - Huihui Guo
- Department of Gynecology and Obstetrics, 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
| | - Yu Zhang
- Department of Gynecology and Obstetrics, 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
| | - Yufeng Wu
- Department of Gynecology and Obstetrics, 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
| | - Hongjing Wang
- Department of Gynecology and Obstetrics, 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
|
39
|
Wei J, Qin Y, Zhang L, Gong X. Combined protein and transcriptomics identifies DCTPP1 as a putative biomarkers for predicting immunotherapy responsiveness in gastric cancer patients. Anticancer Drugs 2025; 36:415-426. [PMID: 39908235 DOI: 10.1097/cad.0000000000001704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
This study aimed to screen the changes after overexpression of dCTP pyrophosphatase 1 (DCTPP1) in human gastric adenocarcinoma cells (AGS) cells by proteome and transcriptome sequencing. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis were performed to explore the functional significance of the differentially expressed DCTPP1 in gastric cancer (GC). Cell Counting Kit-8 (CCK-8) assay was used to detect the proliferation of cells. Western blot was used to detect the expression of proteins. A total of 28 genes that were significantly associated with DCTPP1 overexpression and had prognostic value were screened by Cox regression analysis. The results of gene set enrichment analysis showed that the genomes of patients with subtype A exhibited significant enrichment in pathways such as DNA repair, pyrimidine synthesis, and glucose metabolism. The tumor immune dysfunction and exclusion and The Cancer Immunome Atlas databases showed that patients with type A GC were better candidates for immunotherapy than patients with type B GC. Furthermore, the CCK-8 assay indicated significantly enhanced proliferative activity after overexpressing DCTPP1 in AGS cells, corroborating the findings from the bioinformatic analysis. The data suggest a potential association between DCTPP1 expression and both the prognosis of GC patients and the efficacy of immunotherapy. These findings offer valuable insights for the potential optimization of therapeutic strategies in gastric cancer.
Collapse
Affiliation(s)
- Jun Wei
- Department of Gastroenterology, General Practice, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical University
| | | | - Luwen Zhang
- School of Clinic Medicine, Bengbu Medical University, Bengbu, Anhui Province, China
| | - Xiaobing Gong
- Department of Gastroenterology, General Practice, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province
| |
Collapse
|
40
|
Zhang M, Yang Q, Lou J, Hu Y, Shi Y. A new strategy to HER2-specific antibody discovery through artificial intelligence-powered phage display screening based on the Trastuzumab framework. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167772. [PMID: 40056877 DOI: 10.1016/j.bbadis.2025.167772] [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: 11/21/2024] [Revised: 02/23/2025] [Accepted: 02/28/2025] [Indexed: 03/10/2025]
Abstract
Human epidermal growth factor receptor 2 (HER2) is a recognized drug target, and it serves as a critical target for various cancer treatments, necessitating the discovery of more antibodies for therapeutic and detection purposes. Here, we have developed an innovative workflow for antibody generation through Artificial Intelligence-powered Phage Display Screening (AIPDS). This workflow integrates artificial intelligence-driven antibody CDRH3 sequence design, high-throughput DNA synthesis and phage display screening. We applied AIPDS workflow to generate promising antibodies against the human epidermal growth factor receptor 2 (HER2), offering a template for streamlined antibody generation. Seven novel antibodies stood out, demonstrating promising efficacy in various functional assays. Notably, DYHER2-02 demonstrates strong performance across all experimental tests. In summary, our study introduces a novel methodology to generate new antibody variants of an existing antibody using an AI-assisted phage display approach. These new antibody variants hold potential applications in research, diagnosis, and therapeutic applications.
Collapse
Affiliation(s)
- Mancang Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Qiangzhen Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Jiangrong Lou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Yang Hu
- United Research Center for Next Generation DNA Synthesis of SJTU-Dynegene, Shanghai 201108, People's Republic of China
| | - Yongyong Shi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China; Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China.
| |
Collapse
|
41
|
Gao X, Bian T, Wei X, Xu M, Ren J, Zhao Y, Zheng Y, Li X. Exploration of mechanism of Bufalin and Bufalin derivatives in hepatocellular carcinoma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156686. [PMID: 40220402 DOI: 10.1016/j.phymed.2025.156686] [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: 01/14/2025] [Revised: 03/11/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Bufalin, a promising candidate for the treatment of hepatocellular carcinoma (HCC), has garnered interest from researchers in exploring its pharmacological mechanisms. The traditional single-target research method makes it difficult to elucidate the molecular mechanism systematically, and there are still limitations. PURPOSE To explore the synergistic mechanisms of Bufalin and its derivatives in the treatment of HCC through quantitative and integrated analysis of big data, this study provides a reference for clinical screening of anti-liver cancer drugs and accelerates the research and development of new drugs. METHODS The literature on the anti-liver cancer activity of Bufalin was searched, the research trends and hotspots were analyzed, and the hot targets were identified. Potential targets and signaling pathways were predicted and compared with established targets. To evaluate the affinity of Bufalin and its derivatives with key targets and validate the results of network pharmacology. The structure-activity relationships and pathways were reviewed, and key pharmacophore and potential pathways were identified. RESULTS Analysis of 302 literatures showed that Na+/K+-ATPase was a hot target in the anti-liver cancer mechanism of Bufalin. 76 potential targets are primarily associated with pathways such as PI3K/AKT/mTOR, Hedgehog, and AMPK/mTOR, which play roles in regulating key targets like phosphoinositide 3-kinase (PIK3), adenosine monophosphate-activated protein kinase (AMPK), and epidermal growth factor receptor (EGFR). Bufalin derivatives have a higher affinity with key targets. The cdocker interaction energy (CIE) of BF211, compound 1, and compound 2 with PI3K were -48.0722 kcal/mol, -50.8791 kcal/mol, and -59.7326 kcal/mol, respectively, which were significantly lower than Bufalin (-26.0859 kcal/mol). CONCLUSIONS Bufalin and its derivatives have significant anti-HCC activity and show good potential for drug development. Their mechanism of action involves a complex network of AMPK targets and PI3K/AKT/mTOR signaling pathways. In the future, specific inhibitors should be developed against these targets and pathways as a new therapeutic strategy for HCC. Among them, compounds 2, 7, and 8 have a higher affinity for potential targets than Bufalin and its other derivatives and have higher research value. In addition, α-pyrone is the key pharmacophore of Bufalin, and structural modification of the C3 position can significantly regulate its cytotoxicity and solubility. Therefore, optimization of the structure of the C3 position is expected to reduce the toxicity and improve the water solubility of Bufalin, overcoming the limitations of its clinical application and providing a safer and more effective solution for the treatment of HCC.
Collapse
Affiliation(s)
- Xinchen Gao
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Tianrun Bian
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xinyu Wei
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Mengsi Xu
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiali Ren
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ying Zhao
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yanchao Zheng
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Xiankuan Li
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
42
|
Slotman E, Pape M, van Laarhoven HWM, Pouw RE, van der Linden YM, Verhoeven RHA, Siesling S, Fransen HP, Raijmakers NJH. Considerations to forgo systemic treatment in patients with advanced esophageal or gastric cancer: A real-world evidence study. Int J Cancer 2025; 156:1950-1960. [PMID: 39786196 PMCID: PMC11924308 DOI: 10.1002/ijc.35314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 01/12/2025]
Abstract
The majority of patients with advanced esophageal or gastric cancer do not start palliative systemic treatment. To gain insight into the considerations underlying the decision not to start systemic treatment, we analyzed characteristics of patients starting and not starting systemic treatment, reasons for not starting systemic treatment, and receipt of local palliative treatments on a nationwide scale. Patients diagnosed with advanced esophageal or gastric cancer between 2015 and 2021 were included (n = 10,948). Survival was compared using propensity score matching on patient and disease characteristics. Most patients did not start systemic treatment (esophageal cancer 59%; gastric cancer 64%). These patients were generally older, more often female, had more comorbidities and a worse performance status. The main reason for not starting systemic treatment was patient or family preference (35%). Among patients who did not start systemic treatment, 47% (esophageal) and 19% (gastric), received local palliative treatment, most commonly radiotherapy. Patients who did not start systemic treatment had worse median overall survival compared to patients who did start (esophageal cancer 2.9 months vs. 8.9 months; gastric cancer 2.2 vs. 8.2 months). These findings indicate that patient condition and disease burden are important aspects in systemic treatment decisions. However, patient or family preference was the main reason for not starting systemic treatment, highlighting that their priorities also strongly influence the decision. Systemic treatment did show to be associated with improved overall survival in matched patients, and therefore adequately weighing treatment risks and benefits based on real world data against patient preferences is of utmost importance.
Collapse
Affiliation(s)
- Ellis Slotman
- Department of Research and DevelopmentNetherlands Comprehensive Cancer OrganisationUtrechtThe Netherlands
- Department of Health Technology and Services ResearchUniversity of Twente, Technical Medical CentreEnschedeThe Netherlands
| | - Marieke Pape
- Department of Research and DevelopmentNetherlands Comprehensive Cancer OrganisationUtrechtThe Netherlands
- Medical OncologyAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
- Cancer Treatment and Quality of LifeCancer Center AmsterdamAmsterdamThe Netherlands
| | - Hanneke W. M. van Laarhoven
- Medical OncologyAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
- Cancer Treatment and Quality of LifeCancer Center AmsterdamAmsterdamThe Netherlands
| | - Roos E. Pouw
- Cancer Treatment and Quality of LifeCancer Center AmsterdamAmsterdamThe Netherlands
- Department of Gastroenterology and HepatologyAmsterdam University Medical Centers, location VUmcAmsterdamThe Netherlands
| | - Yvette M. van der Linden
- Centre of Expertise in Palliative CareLeiden University Medical CentreLeidenThe Netherlands
- Department of RadiotherapyLeiden University Medical CentreLeidenThe Netherlands
| | - Rob H. A. Verhoeven
- Department of Research and DevelopmentNetherlands Comprehensive Cancer OrganisationUtrechtThe Netherlands
- Medical OncologyAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
| | - Sabine Siesling
- Department of Research and DevelopmentNetherlands Comprehensive Cancer OrganisationUtrechtThe Netherlands
- Department of Health Technology and Services ResearchUniversity of Twente, Technical Medical CentreEnschedeThe Netherlands
| | - Heidi P. Fransen
- Department of Research and DevelopmentNetherlands Comprehensive Cancer OrganisationUtrechtThe Netherlands
| | - Natasja J. H. Raijmakers
- Department of Research and DevelopmentNetherlands Comprehensive Cancer OrganisationUtrechtThe Netherlands
| |
Collapse
|
43
|
Parry CW, Pellicano F, Schüttelkopf AW, Beyer KS, Bower J, Bryson A, Cameron K, Cerutti NM, Clark JP, Davidson SC, Davies K, Drysdale MJ, Engelman J, Estevan-Barber A, Gohlke A, Gray CH, Guthy DA, Hong M, Hopkins A, Hutchinson LD, Konczal J, Maira M, McArthur D, Mezna M, McKinnon H, Nepravishta R, Ostermann N, Pasquali CC, Pollock K, Pugliese A, Rooney N, Schmiedeberg N, Shaw P, Velez-Vega C, West C, West R, Zecri F, Taylor JB. Reversible Small Molecule Multivariant Ras Inhibitors Display Tunable Affinity for the Active and Inactive Forms of Ras. J Med Chem 2025; 68:9129-9161. [PMID: 40162713 DOI: 10.1021/acs.jmedchem.4c02929] [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: 04/02/2025]
Abstract
Activating mutations of Ras are one of the most prevalent drivers of cancer and are often associated with poor clinical outcomes. Despite FDA approval for two irreversible inhibitors that target the inactive state of KRasG12C, significant unmet clinical need still exists, and the susceptibility of non-G12C mutants to inactive-state inhibition remains unclear. Here we report the discovery of a novel series of reversible inhibitors that bind in an enlarged version of the switch I-II pocket with nanomolar affinities. Dependent on chemotype these can either preferentially bind to the inactive or active state or bind both with similar affinity. The active-state binders inhibit the Raf interaction for wild-type Ras, and a broad range of oncogenic KRas mutants with nanomolar potency. A subseries of these molecules displays cellular inhibition of Ras-Raf binding, as well as decreased phosphorylation of the downstream protein ERK, demonstrating that potent multivariant Ras inhibitors can be accessed from this novel pocket.
Collapse
Affiliation(s)
- Charles W Parry
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Francesca Pellicano
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Alexander W Schüttelkopf
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Kim S Beyer
- Novartis Institute for BioMedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Justin Bower
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Amy Bryson
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Kenneth Cameron
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Nichole M Cerutti
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Jonathan P Clark
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Stuart C Davidson
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Keneth Davies
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Martin J Drysdale
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Jeffrey Engelman
- Novartis Institutes for BioMedical Research, Inc., 181 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Anna Estevan-Barber
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Andrea Gohlke
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Christopher H Gray
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Daniel A Guthy
- Novartis Institute for BioMedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Min Hong
- NCI RAS Initiative, Frederick National Laboratory for Cancer Research, P.O. Box B, Frederick, Maryland 21702, United States
| | - Alana Hopkins
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Luke D Hutchinson
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Jennifer Konczal
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Michel Maira
- Novartis Institute for BioMedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Duncan McArthur
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Mokdad Mezna
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Heather McKinnon
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Ridvan Nepravishta
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Nils Ostermann
- Novartis Institute for BioMedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Camila C Pasquali
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Katie Pollock
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Angelo Pugliese
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Nicholas Rooney
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Niko Schmiedeberg
- Novartis Institute for BioMedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Paul Shaw
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Camilo Velez-Vega
- Novartis Institutes for BioMedical Research, Inc., 181 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Christopher West
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Ryan West
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Frederic Zecri
- Novartis Institutes for BioMedical Research, Inc., 181 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - John B Taylor
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| |
Collapse
|
44
|
Ladekarl M, Mørk ML, Albertsen ES, Nielsen D, Lassen U, Mau-Sørensen M, Nielsen CM, Jakobsen A, von der Maase H. Twenty-one-year report from the Danish Health Authority Expert Advisory Panel for review of treatment of 10 000 cancer patients. Oncologist 2025; 30:oyaf059. [PMID: 40338216 DOI: 10.1093/oncolo/oyaf059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 02/12/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Patients with hard-to-treat or rare cancers and those not responding to standard-of-care (SoC) treatment have unmet needs. Limited access to novel drugs is an increasing additional challenge. In 2003, the Danish government adopted a Health Act to ensure that treatment of patients with life-threatening disease could be reevaluated by independent experts. The Danish Health Authority (DHA) set up an Expert Advisory Panel to provide advice on possibilities for further treatment of patients, including treatment not approved nationally. A few years later, clinical units were established that could offer unestablished treatment to patients by referral from the Panel. The treatment was first reimbursed by the Government and later by regional authorities. MATERIALS AND METHODS We present the structure, workflow, and impact of the Health Act for 21 years for patients with cancer. Annual reports from the DHA were the primary data source. RESULTS 11 034 cases from 9603 cancer patients were evaluated by the Panel from 2003 to 2023, representing a median of 372 unique cases yearly. In 53%, the Panel advised on further treatment in Denmark, and of these, 56% were recommended nationally nonapproved treatment, 21% SoC treatment or workup, and 19% clinical trial participation. In 4.5% of cases, advice was given on treatment abroad. A significant decline in admissions to the Panel from a peak of 1167 patients in 2008 to 3-400 yearly from 2012 to 2017 followed the conversion of nonapproved treatments to SoC practice. A shift in drug reimbursement, independent of Panel advise, reduced the clinical impact and explained the further decline observed in admissions lately to only 51 patients in 2023. CONCLUSIONS This unique national scheme provided early access to treatment for patients with no further SoC options and facilitated the introduction of new cancer treatments, initiation of clinical trials, and establishment of trial units in the country. The scheme may be adapted to other countries with a public healthcare system. Results of the current report indicate that impact is dependent on delivering clinical units and reimbursement associated with the recommended treatment.
Collapse
Affiliation(s)
- Morten Ladekarl
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, 9000 Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, 9220 Aalborg, Denmark
| | | | | | - Dorte Nielsen
- Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, 2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
| | - Ulrik Lassen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
- Department of Oncology, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen, Denmark
| | - Morten Mau-Sørensen
- Department of Oncology, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen, Denmark
| | | | - Anders Jakobsen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark
| | - Hans von der Maase
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
- Department of Oncology, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen, Denmark
| |
Collapse
|
45
|
Taniguchi H, Nishikawa K, Haneji T, Izawa N, Imamura H, Yamaguchi H. Prospective Cohort Study of Trastuzumab Biosimilar CT-P6 in HER2-Positive Gastric Cancer: Japanese Real-World Outcomes. Oncol Ther 2025:10.1007/s40487-025-00341-7. [PMID: 40338476 DOI: 10.1007/s40487-025-00341-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/09/2025] [Indexed: 05/09/2025] Open
Abstract
INTRODUCTION CT-P6, the first trastuzumab biosimilar, was approved on the basis of data limited to human epidermal growth factor receptor-2 (HER2)-positive early breast cancer. Usage for other indications was granted by extrapolation, and post-approval clinical studies were conducted to confirm the effect of CT-P6 in HER2-positive gastric cancer. METHODS After approval in Japan in 2018, a prospective post-marketing surveillance was conducted in 171 patients (130 male, 41 female) with HER2-positive unresectable advanced or recurrent gastric cancer. The safety and efficacy of CT-P6 were evaluated over 1 year. RESULTS CT-P6 was primarily combined with fluoropyrimidine and/or platinum agents. Adverse events occurred in 151 patients (88.3%), with 55 patients (32.2%) experiencing grade 3 or higher. Infusion reactions occurred in 12.3%. Four cardiac disorders were reported: two of grade 1 cardiac dysfunction and two of severe ischemic heart disease. Interstitial lung disease was reported in four patients (2.3%). The objective response rate was 34.4%, and the disease control rate was 82.4%. The progression-free survival (PFS) was 7.4 months. Significant risk factors for PFS included gastroesophageal junction, ≥ 3 metastases, no gastrectomy, prior chemotherapy, and no platinum agent. CONCLUSIONS In this cohort study, CT-P6 demonstrated sufficient efficacy and no new safety concerns in HER2-positive advanced gastric cancer, serving as a cost-effective alternative to originator trastuzumab. TRIAL REGISTRATION Japan Registry of Clinical Trials, Trial ID: jRCT2071230094 (November 2023).
Collapse
Affiliation(s)
- Hiroya Taniguchi
- Department of Clinical Oncology, Aichi Cancer Center Hospital, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi, 464-8681, Japan
| | - Kiyohiro Nishikawa
- Quality and Pharmacovigilance Division, Pharmaceuticals Group, Nippon Kayaku Co., Ltd., 2-1-1 Marunouchi, Chiyoda-ku, Tokyo, 100-0005, Japan.
- Asajes Ventures, 3-11-5 Nihonbashi Honcho, Chuo-ku, Tokyo, 103-0023, Japan.
| | - Tatsuo Haneji
- Quality and Pharmacovigilance Division, Pharmaceuticals Group, Nippon Kayaku Co., Ltd., 2-1-1 Marunouchi, Chiyoda-ku, Tokyo, 100-0005, Japan
| | - Naoki Izawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Hiroshi Imamura
- Department of Digestive Surgery, Toyonaka Municipal Hospital, 4-14-1 Shibaharacho, Toyonaka, Osaka, 560-8565, Japan
| | - Hironori Yamaguchi
- Department of Surgery, Division of Gastroenterological, General and Transplant Surgery, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| |
Collapse
|
46
|
Kodama H, Narita Y, Nakamura M, Takahashi M, Mizukami T, Ando T, Mitani S, Komori A, Hosokawa A, Moriwaki T, Sugiyama K, Taguri M, Orihara S, Kagamu H, Yamaguchi T, Nishikawa H, Muro K. A multicenter, prospective, observational study of nivolumab readministration for advanced gastric cancer (NIVO RETURNS). Future Oncol 2025:1-7. [PMID: 40336472 DOI: 10.1080/14796694.2025.2500918] [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: 04/07/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
Combination treatment with an anti-programmed cell death-1 (PD-1) antibody, an immune checkpoint inhibitor (ICI), and chemotherapy is the standard treatment for patients with HER2-negative advanced gastric/esophagogastric cancer (AGC). ICI re-administration has been reported to have a clinical benefit for patients with lung cancer or melanoma. However, data on patients with AGC have not yet been collected. We plan to conduct a prospective, multicenter, observational NIVO RETURNS study to evaluate the efficacy and safety of nivolumab monotherapy re-administration in patients with AGC refractory to initial anti-PD-1 or anti-programmed cell death ligand-1 (PD-L1) antibody treatment. Patients who have achieved clinical benefits (complete response, partial response, or stable disease for ≥ 6 months) from prior treatment, including anti-PD-1/PD-L1 therapy, will be included. The primary endpoint will be the objective response rate to nivolumab re-administration. We anticipate that our findings will contribute to the improvement of survival outcomes as later-line treatment for AGC.Clinical trial registration: UMIN000050515, UMIN000051044.
Collapse
Affiliation(s)
- Hiroyuki Kodama
- Cancer Chemotherapy Center, Osaka Medical and Pharmaceutical University, Osaka, Japan
- Department of Gastroenterology, Osaka Medical and Pharmaceutical University, Osaka, Japan
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Yukiya Narita
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Michio Nakamura
- Department of Gastroenterology, Sapporo City General Hospital, Sapporo, Japan
| | - Masanobu Takahashi
- Department of Medical Oncology, Tohoku University Hospital, Sendai, Japan
| | | | - Takayuki Ando
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Seiichiro Mitani
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Azusa Komori
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Ayumu Hosokawa
- Department of Clinical Oncology, University of Miyazaki Hospital, Miyazaki, Japan
| | - Toshikazu Moriwaki
- Department of Gastroenterology and Hepatology, Kurashiki Central Hospital, Kurashiki, Japan
| | - Keiji Sugiyama
- Department of Medical Oncology, NHO Nagoya Medical Center, Nagoya, Japan
| | - Masataka Taguri
- Department of Health Data Science, Tokyo Medical University, Tokyo, Japan
| | - Shunichiro Orihara
- Department of Health Data Science, Tokyo Medical University, Tokyo, Japan
| | - Hiroshi Kagamu
- Division of Respiratory Medicine, Saitama Medical University International Medical Center, Saitama, Japan
| | - Toshifumi Yamaguchi
- Cancer Chemotherapy Center, Osaka Medical and Pharmaceutical University, Osaka, Japan
- Department of Gastroenterology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Hiroki Nishikawa
- Department of Gastroenterology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| |
Collapse
|
47
|
Li BY, Li HL, Zeng FE, Luan XY, Liu BQ, Wang ZZ, Zhang L, Dong XZ. Identification of PD-L1-related biomarkers for selecting gastric adenocarcinoma patients for PD-1/PD-L1 inhibitor therapy. Discov Oncol 2025; 16:689. [PMID: 40338384 DOI: 10.1007/s12672-025-02515-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 04/25/2025] [Indexed: 05/09/2025] Open
Abstract
PD-1/PD-L1 inhibitors have been used to treat gastric cancer, and PD-L1 expression has been identified as a biomarker for predicting the effectiveness of immunotherapy in the treatment of gastric cancer. However, PD-L1 expression prediction for immunotherapy response is inaccurate, and improved response biomarkers are required. Thus, it is important to identify additional biomarkers that can predict the responses to PD-1/PD-L1 monoclonal antibodies in gastric cancer. In this study, GO and KEGG enrichment analysis of 142 DEGs co-expressed with PD-L1 were performed, and 41 genes were identified based on the intersection of the mRNA-significant GO term network and the mRNA-significant signalling pathway network. Further intersection analysis of the 41 candidate genes and 137 positive immunotherapy response genes indicated that BATF2 significantly affects the overall survival of GC patients. The transcription factor prediction for BATF2 identified additional potential predictors and therapeutic targets for GC. STAT and IRF family members were predicted to be transcription factors for BATF2. In addition, BATF2 knockdown significantly promoted GC cell growth, and PD-L1 expression was upregulated in si-BATF2-treated MKN-45 cells. Thus, BATF2 may serve as a biomarker for predicting the efficacy of PD-L1 blockade therapy in GC. BATF2 acts as a tumour suppressor gene during the development of GC. BATF2 is closely related to PD-L1 expression in GC, and high BATF2 expression positively correlates with low PD-L1 expression. BATF2 can be used as a potential biomarker and therapeutic target for responding to anti-PD-1 and anti-PD-L1 immunotherapies in GC.
Collapse
Affiliation(s)
- Bo-Ya Li
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing, China
| | - Hui-Ling Li
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing, China
| | - Fei-Er Zeng
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, UK
| | - Xuan-Yu Luan
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bi-Qing Liu
- Department of Pharmacy, Children's Hospital Affiliated to Capital Institute of Paediatrics, Beijing, China
| | - Zhi-Zhou Wang
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing, China
| | - Lan Zhang
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing, China.
| | - Xian-Zhe Dong
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing, China.
| |
Collapse
|
48
|
Hua Y, Gao Y, Luo S, Song G, Tian X, Wang C, Lv S, Zhang X, Shao G. Programmed cell death 1 inhibitor combined with chemotherapy compared to chemotherapy alone as first-line treatment in advanced gastric cancer patients: A real-world study. Int Immunopharmacol 2025; 154:114487. [PMID: 40179586 DOI: 10.1016/j.intimp.2025.114487] [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: 08/22/2024] [Revised: 10/13/2024] [Accepted: 03/11/2025] [Indexed: 04/05/2025]
Abstract
OBJECTIVE Previous trials have revealed better treatment efficacy of programmed cell death 1 (PD-1) inhibitors combined with chemotherapy as first-line treatments than chemotherapy alone in advanced gastric cancer (GC) patients, but real-world evidence is still lacking. Hence, this real-world study aimed to investigate the efficacy and safety of PD-1 inhibitors plus chemotherapy as first-line treatments compared with chemotherapy alone in advanced GC patients. METHODS In total, 102 advanced GC patients were allocated into a combination group (receiving chemotherapy combined with a PD-1 inhibitor as a first-line treatment) (n = 48) or a chemotherapy group (receiving chemotherapy only as a first-line treatment) (n = 54) according to their actual treatment regimens. RESULTS The objective response rate (ORR) was greater in the combination group than in the chemotherapy group (25.0 % versus 9.3 %, P = 0.033), whereas the disease control rate (DCR) was not different between the groups (83.3 % versus 66.7 %, P = 0.054). Progression-free survival (PFS) was prolonged in the combination group than in the chemotherapy group (P = 0.018). The median (95 % confidence interval) PFS was 19.7 (12.2-27.2) months in the combination group and 16.5 (7.3-25.7) months in the chemotherapy group. Multivariate logistic regression analyses revealed that PD-1 inhibitors combined with chemotherapy were independently associated with an increased ORR (odds ratio: 4.180, P = 0.024), increased DCR (odds ratio: 2.928, P = 0.049), and prolonged PFS (hazard ratio: 0.388, P = 0.030). No difference was found in total or each specific grade III-IV adverse reaction between the groups (all P > 0.05). CONCLUSION Treatment with a PD-1 inhibitor plus chemotherapy as a first-line treatment shows better treatment efficacy with similar safety to that of chemotherapy alone in advanced GC patients.
Collapse
Affiliation(s)
- Yunqi Hua
- Department of Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia 014040, China
| | - Yuqian Gao
- Department of Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia 014040, China
| | - Shuang Luo
- Department of Graduate School, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014030, China
| | - Ge Song
- Department of Graduate School, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014030, China
| | - Xiaoling Tian
- Department of Graduate School, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014030, China
| | - Chenlin Wang
- Department of Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia 014040, China
| | - Shuang Lv
- Department of Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia 014040, China
| | - Xinyi Zhang
- Department of Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia 014040, China
| | - Guo Shao
- Center for Translational Medicine, the Third People's Hospital of Longgang District & Longgang Institute of Medical Imaging, Shantou University Medical College, Shenzhen 518100, China.
| |
Collapse
|
49
|
Yan Z, Wang C, Wu J, Wang J, Ma T. TIM-3 teams up with PD-1 in cancer immunotherapy: mechanisms and perspectives. MOLECULAR BIOMEDICINE 2025; 6:27. [PMID: 40332725 DOI: 10.1186/s43556-025-00267-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 04/13/2025] [Accepted: 04/18/2025] [Indexed: 05/08/2025] Open
Abstract
Immunotherapy using immune checkpoint inhibitors (ICIs) has become a prominent strategy for cancer treatment over the past ten years. However, the efficacy of ICIs remains limited, with certain cancers exhibiting resistance to these therapeutic approaches. Consequently, several immune checkpoint proteins are presently being thoroughly screened and assessed in both preclinical and clinical studies. Among these candidates, T cell immunoglobulin and mucin-domain containing-3 (TIM-3) is considered a promising target. TIM-3 exhibits multiple immunosuppressive effects on various types of immune cells. Given its differential expression levels at distinct stages of T cell dysfunction in the tumor microenvironment (TME), TIM-3, along with programmed cell death protein 1 (PD-1), serves as indicators of T cell exhaustion. Moreover, it is crucial to carefully evaluate the impact of TIM-3 and PD-1 expression in cancer cells on the efficacy of immunotherapy. To increase the effectiveness of anti-TIM-3 and anti-PD-1 therapies, it is proposed to combine the inhibition of TIM-3, PD-1, and programmed death-ligand 1 (PD-L1). The efficacy of TIM-3 inhibition in conjunction with PD-1/PD-L1 inhibitors is being evaluated in a number of ongoing clinical trials for patients with various cancers. This study systematically investigates the fundamental biology of TIM-3 and PD-1, as well as the detailed mechanisms through which TIM-3 and PD-1/PD-L1 axis contribute to cancer immune evasion. Additionally, this article provides a thorough analysis of ongoing clinical trials evaluating the synergistic effects of combining PD-1/PD-L1 and TIM-3 inhibitors in anti-cancer treatment, along with an overview of the current status of TIM-3 and PD-1 antibodies.
Collapse
Affiliation(s)
- Zhuohong Yan
- Department of Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Chunmao Wang
- Department of Thoracic Surgery, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Jinghong Wu
- Department of Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Jinghui Wang
- Department of Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Teng Ma
- Department of Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
| |
Collapse
|
50
|
Alchaikh Hassan R, Patel A, Dasanu CA. Selecting optimal therapy for advanced non-functional pancreatic neuroendocrine tumors: latest research and therapeutic advances. Expert Opin Pharmacother 2025:1-5. [PMID: 40316435 DOI: 10.1080/14656566.2025.2501142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Affiliation(s)
| | - Arti Patel
- Department of Internal Medicine, Eisenhower Health, Rancho Mirage, CA, USA
| | - Constantin A Dasanu
- Lucy Curci Cancer Center, Eisenhower Health, Rancho Mirage, CA, USA
- Department of Medical Oncology and Hematology, University of California in San Diego Health System, San Diego, CA, USA
| |
Collapse
|