1
|
Liu W, Yang X, Zhou Y, Huang Z, Huang J. Gut microbiota in melanoma: Effects and pathogeneses. Microbiol Res 2025; 296:128144. [PMID: 40120565 DOI: 10.1016/j.micres.2025.128144] [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: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
The gut microbiota exhibits intricate connections with the body's immune system and holds significant implications for various diseases and cancers. Currently, accumulating evidence suggests a correlation between the composition of the gut microbiota and the development, treatment, and prognosis of melanoma. However, the underlying pathogenesis remains incompletely elucidated. In this comprehensive review, we present an in-depth review of the role played by gut microbiota in melanoma tumorigenesis, growth, metastasis, treatment response, and prognosis. Furthermore, we discuss the potential utility of gut microbiota as a promising prognostic marker. Lastly, we summarize three routes through which gut microbiota influences melanoma: immunity, aging, and the endocrine system. By modulating innate and adaptive immunity in patients with melanoma across different age groups and genders, the gut microbiota plays a crucial role in anti-tumor immune regulation from tumorigenesis to prognosis management, thereby impacting tumor growth and metastasis. This review also addresses current study limitations while highlighting future research prospects.
Collapse
Affiliation(s)
- Wenwen Liu
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xin Yang
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yuwei Zhou
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ziru Huang
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jian Huang
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, China; School of Healthcare Technology, Chengdu Neusoft University, Chengdu, Sichuan, China.
| |
Collapse
|
2
|
Sun Y, Liu JQ, Chen WJ, Tang WF, Zhou YL, Liu BJ, Wei Y, Dong JC. Astragaloside III inhibits MAPK-mediated M2 tumor-associated macrophages to suppress the progression of lung Cancer cells via Akt/mTOR signaling pathway. Int Immunopharmacol 2025; 154:114546. [PMID: 40184811 DOI: 10.1016/j.intimp.2025.114546] [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: 12/28/2024] [Revised: 02/25/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025]
Abstract
Tumor-associated macrophages (TAMs) play a key role in facilitating a range of cancerous processes by modulating the tumor microenvironment thus being a target for cancer treatment. Astragaloside III (AS-III), a compound derived from Astragalus triterpenoid saponins, has demonstrated immunomodulatory and anticancer properties, but the underlying mechanism remains unclear. Here, we demonstrated that AS-III suppressed metastasis, angiogenesis and induced apoptosis of lung cancer in vitro and in vivo by inhibiting macrophage M2 polarization and inducing M1 phenotype transformation. This was achieved through the inhibition of the MAPK signaling pathway. Furthermore, the tumor inhibitory effects of AS-III were found to be mediated by the Akt/mTOR pathway. Overall, these results highlight the role of AS-III in modifying the TAMs in TME, offering fresh perspectives on tumor immunotherapy by means of targeting macrophage.
Collapse
Affiliation(s)
- Yan Sun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Jia-Qi Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Wen-Jing Chen
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Wei-Feng Tang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yao-Long Zhou
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Bao-Jun Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China.
| | - Jing-Cheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Zhao Z, Yang C, Li J. Pemetrexed and platinum with or without pembrolizumab for advanced non-small-cell lung cancer (NSCLC): a systematic review and meta-analysis. Clin Transl Oncol 2025; 27:2024-2036. [PMID: 39402420 DOI: 10.1007/s12094-024-03751-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/01/2024] [Indexed: 04/19/2025]
Abstract
OBJECTIVE This meta-analysis aimed to evaluate the efficacy and safety of combining pemetrexed and platinum with or without pembrolizumab for the treatment of advanced non-small-cell lung cancer (NSCLC). METHODS A systematic search of PubMed, Embase, Cochrane Library, and Web Of Science databases was conducted to identify studies comparing pemetrexed and platinum with or without pembrolizumab in advanced NSCLC. Raw data were extracted from eligible studies to calculate Hazard Ratios (HR) for Progression-Free Survival (PFS) and Overall Survival (OS), as well as rates of adverse events of all grades and those of Grade 3 or higher. RESULTS Eight studies with 1639 patients occurred advanced NSCLC included. The group receiving pembrolizumab in combination with pemetrexed and platinum showed significant benefits in terms of OS (HR 0.63; 95% CI 0.54-0.73; p < 0.00001) and PFS (HR:0.64; 95% CI 0.48-0.85; p = 0.002) compared to the group receiving pemetrexed and platinum alone. However, this benefit was accompanied by a higher incidence of Grade 3 or higher adverse events (OR: 1.55; 95% CI 1.24-1.95; p = 0.0001). CONCLUSION The combination of pemetrexed and platinum with pembrolizumab is recommended as a first-line treatment option for advanced NSCLC due to its significant efficacy benefits. However, the increased risk of Grade 3 or higher adverse events suggests the need for careful consideration and assessment when considering this regimen for second-line or subsequent therapy.
Collapse
Affiliation(s)
- Zichen Zhao
- Lianyungang Clinical Medical College of Nanjing Medical University, Lianyungang, China
| | - Chuchu Yang
- Lianyungang Clinical Medical College of Nanjing Medical University, Lianyungang, China
| | - Jiashu Li
- Lianyungang Clinical Medical College of Nanjing Medical University, Lianyungang, China.
| |
Collapse
|
4
|
Yabuki Y, Mitsuhashi A, Ogino H, Yoshida A, Nguyen NT, Yoneda H, Ozaki R, Tsukazaki Y, Morita Y, Nokihara H, Sato S, Shinohara T, Hanibuchi M, Nishioka Y. Hypoxia-inducible factor-targeting therapy augmented the sensitivity to programmed death ligand-1 blockade by enhancing interferon-γ-induced chemokines in tumor cells. Int J Cancer 2025; 156:1814-1825. [PMID: 39686841 DOI: 10.1002/ijc.35301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 10/06/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024]
Abstract
Immune checkpoint inhibitors (ICIs) targeting programmed death ligand-1 (PD-L1) provide clinical benefits for various advanced malignancies. However, the predictive factors that determine sensitivity to ICIs have not been fully elucidated. We focused on tumor-derived CXCL10/11 as a pivotal factor that determines the response to PD-L1 blockade by regulating T cell accumulation and tumor angiogenesis. We previously reported that CXCL10/11 was upregulated by interferon (IFN)-γ in ICI-sensitive tumor cells but not in ICI-resistant cells, including mouse Lewis lung carcinoma (LLC). In the present study, gene silencing of tumor-derived CXCL10/11 induced resistance to PD-L1 blockade in AB1-HA mesothelioma cell-bearing mice. To identify the mechanisms underlying ICI resistance, we performed a microarray analysis to compare the IFN-γ-inducible genes between ICI-sensitive AB1-HA and ICI-resistant LLC in vitro. A pathway analysis based on microarray data indicated that hypoxia-inducible factor (HIF) 1A is the key signal that inhibits CXCL10/11 expression. We revealed that the HIF1A inhibitors echinomycin (EC) and YC-1 upregulated CXCL10/11 genes induced by IFN-γ in tumor cells in vitro. In addition, combination therapy with PD-L1 blockade and EC demonstrated synergistic antitumor effects in LLC-bearing mice. Combination therapy enhanced tumor infiltration of CD8 T cells and suppressed tumor angiogenesis. The present study suggests that HIF1A signaling in tumor cells dominates ICI resistance via the downregulation of tumor-derived CXCL10/11.
Collapse
Affiliation(s)
- Yohei Yabuki
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Atsushi Mitsuhashi
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hirokazu Ogino
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Aito Yoshida
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Na Thi Nguyen
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hiroto Yoneda
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Ryohiko Ozaki
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yuki Tsukazaki
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yutaka Morita
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hiroshi Nokihara
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Seidai Sato
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Tsutomu Shinohara
- Department of Community Medicine for Respirology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Masaki Hanibuchi
- Department of Community Medicine for Respirology, Hematology and Metabolism, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yasuhiko Nishioka
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
- Department of Community Medicine for Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
5
|
Wang J, Chen Q, Shan Q, Liang T, Forde P, Zheng L. Clinical development of immuno-oncology therapeutics. Cancer Lett 2025; 617:217616. [PMID: 40054657 PMCID: PMC11930610 DOI: 10.1016/j.canlet.2025.217616] [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: 11/05/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
Immuno-oncology (IO) is one of the fastest growing therapeutic areas within oncology. IO agents work indirectly via the host's adaptive and innate immune system to recognize and eradicate tumor cells. Despite checkpoint inhibitors being only introduced to the market since 2011, they have become the second most approved product category. Current Food and Drug Administration (FDA)-approved classes of IO agents include: immune checkpoint inhibitors (ICIs), chimeric antigen receptor T-cell therapy (CAR-T), bi-specific T-cell engager (BiTE) antibody therapy, T-cell receptor (TCR) engineered T cell therapy, tumor-infiltrating lymphocyte (TIL) therapy, cytokine therapy, cancer vaccine therapy, and oncolytic virus therapy. Cancer immunotherapy has made progress in multiple cancer types including melanoma, non-small cell lung cancer (NSCLC), renal cell carcinoma (RCC), and urothelial carcinoma; however, several cancers remain refractory to immunotherapy. Future directions of IO include exploration in the neoadjuvant/perioperative setting, combination strategies, and optimizing patient selection through improved biomarkers.
Collapse
Affiliation(s)
- Jianxin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Qi Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Qiang Shan
- Department of General Surgery, Haining People's Hospital, Haining, 314400, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Patrick Forde
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA; The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Mays Cancer Center at the University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Lei Zheng
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA; The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Mays Cancer Center at the University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
6
|
Hua K, Hong H, Wang X. Biomarker-guided adaptive enrichment design with threshold detection for clinical trials with time-to-event outcome. J Biopharm Stat 2025:1-18. [PMID: 40253620 DOI: 10.1080/10543406.2025.2489291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 03/14/2025] [Indexed: 04/22/2025]
Abstract
Biomarker-guided designs are increasingly used to evaluate personalized treatments based on patients' biomarker status in Phase II and III clinical trials. With adaptive enrichment, these designs can improve the efficiency of evaluating the treatment effect in biomarker-positive patients by increasing their proportion in the randomized trial. While time-to-event outcomes are often used as the primary endpoint to measure treatment effects for a new therapy in severe diseases like cancer and cardiovascular diseases, there is limited research on biomarker-guided adaptive enrichment trials in this context. Such trials almost always adopt hazard ratio methods for statistical measurement of treatment effects. In contrast, restricted mean survival time (RMST) has gained popularity for analyzing time-to-event outcomes because it offers more straightforward interpretations of treatment effects and does not require the proportional hazard assumption. This paper proposes a two-stage biomarker-guided adaptive RMST design with threshold detection and patient enrichment. We develop sophisticated methods for identifying the optimal biomarker threshold and biomarker-positive subgroup, treatment effect estimators, and approaches for type I error rate, power analysis, and sample size calculation. We present a numerical example of re-designing an oncology trial. An extensive simulation study is conducted to evaluate the performance of the proposed design.
Collapse
Affiliation(s)
- Kaiyuan Hua
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Hwanhee Hong
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Xiaofei Wang
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
7
|
Yao S, Elakad O, Yang XH, Altaf AR, Yu WT, Bohnenberger H, Peng LG. MTHFD2 marks pemetrexed resistance in pulmonary adenocarcinoma with EGFR wild type. Discov Oncol 2025; 16:581. [PMID: 40253662 PMCID: PMC12009792 DOI: 10.1007/s12672-025-02355-z] [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: 10/12/2024] [Accepted: 04/09/2025] [Indexed: 04/22/2025] Open
Abstract
PURPOSE Lung cancer is the leading cause of cancer-related deaths worldwide. Patients with an amplification of the MTHFD2 gene have a particularly poor prognosis. MTHFD2 signaling has been associated with migration, metastasis, and proliferation of lung cancer cells mediated through ERK signaling. Although the enzymatic activity of the MTHFD2 protein is well understood, little is known about its larger role in chemoresistance. METHODS Seventy-nine of non-small cell lung cancer (NSCLC) samples with clinical follow-up were subjected to immunohistochemical staining for MTHFD2 and sequenced using next generation sequencing (NGS) to determine EGFR status. MTHFD2 gene was knocked down in two NSCLC cell lines with wild type EGFR gene (HCC44 and H1993) where MTHFD2 signaling and chemotherapy resistance against pemetrexed were evaluated. RESULTS MTHFD2 expression data revealed a strong prognosis relevance in adenocarcinoma (LUAD). Immunoblotting of cell lines showed a MTHFD2 dependent and cell type specific ERK signaling in EGFR wild type cells. MTHFD2 expression induced proliferation of NSCLC cells and their resistance against pemetrexed. Knocking down the MTHFD2 gene induced cycle arrest, however, it did not activate apoptosis signaling within HCC44 cell line. CONCLUSIONS MTHFD2 expression is strongly associated with prognosis in LUAD patients, as well as with increased cellular proliferation and resistance to pemetrexed in LUAD patients with wild-type EGFR. These findings suggest that MTHFD2 could serve as a valuable biomarker for predicting treatment outcomes in LUAD. Further studies are needed to fully explore the clinical implications and potential combination therapies targeting MTHFD2 in LUAD.
Collapse
Affiliation(s)
- Sha Yao
- Department of Pathology, The 3rd Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Omar Elakad
- Institute of Pathology, University Medical Center, 37079, Göttingen, Germany
| | - Xiang Hui Yang
- Department of Oncology, Changsha Central Hospital, University of South China, Changsha, 410004, China
| | - Adnan Raza Altaf
- College of Engineering, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wen Tao Yu
- Department of Pathology, The 3rd Xiangya Hospital, Central South University, Changsha, 410013, China
| | | | - Luo Gen Peng
- Department of Oncology, Changsha Central Hospital, University of South China, Changsha, 410004, China.
| |
Collapse
|
8
|
Liu J, Liu J, Chen H, Zhang Q, Zhang P, Chen Z, Lu X, Xu Y. Prognostic value of combined nutritional and inflammatory markers in NSCLC patients receiving ICIs. Discov Oncol 2025; 16:571. [PMID: 40253541 PMCID: PMC12009260 DOI: 10.1007/s12672-025-02391-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 04/14/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND In the treatment of non-small cell lung cancer (NSCLC), immune checkpoint inhibitors (ICIs) have markedly improved patient survival, yet some patients do not benefit. The existing prognostic factors are limited, highlighting the development of reliable and convenient predictive indicators. METHODS A retrospective analysis was performed on 219 NSCLC patients treated with ICIs from June 2019 to January 2024. The nutritional risk screening (NRS 2002) and the neutrophil-to-lymphocyte ratio (NLR) were employed to evaluate the patients' nutritional status and inflammatory response, aiming to investigate the correlation between these markers and treatment outcomes. RESULTS The median follow-up duration for the overall population was 29 (IQR: 25.96-32.04) months. The analysis showed that the median progression-free survival (mPFS) and median overall survival (mOS) in the high nutritional risk group (NRS2002 ≥ 3, accounting for 23.74%) were significantly lower than those in the low nutritional risk group (NRS2002 < 3, accounting for 76.26%) (mPFS: 2.5 vs 16 months; mOS: 8 vs 16 months, both P < 0.001). Similarly, patients with high NLR values (> 4.92) had significantly shorter OS and PFS than those with low NLR (≤ 4.92) values (mOS: 7 vs 18 months; mPFS: 3 vs 17 months, both P < 0.001). Multivariate Cox analysis revealed that a high NRS 2002 score (HR = 2.76, 95% CI 1.68-4.54, P < 0.001) and high NLR (HR = 2.77, 95% CI 1.65-4.64, P < 0.001) were independent predictors of poor prognosis. Risk stratification was performed using a combined scoring system of NRS 2002 and NLR (0 points-low risk, 1 point-moderate risk, 2 points-high risk), and it was found that as the risk score increased, OS and PFS significantly decreased (mOS: 8 [2.61-13.39] vs 16 [13.04-18.96] vs NA [NA-NA] months; mPFS: 2.5 [0.99-4.02] vs 8.5 [5.47-11.53] vs 16 [11.41-20.59] months, respectively, both P < 0.001). The utility of the combined NLR and NRS2002 scoring model was assessed using a time-dependent receiver operating characteristic (ROC) curve, with results indicating that at 12 months, the AUC value of the combined scoring model was 0.81 (CI 0.72-0.90). At 24 and 36 months, the AUC values were 0.73 (CI 0.66-0.80) and 0.70 (CI 0.64-0.76), respectively. Moreover, the nomogram model exhibited high predictive accuracy in predicting survival prognosis, with AUC values of 0.84 (CI 0.77-0.91), 0.85 (CI 0.79-0.91), and 0.78 (CI 0.69-0.88) at 12, 24, and 36 months, respectively. CONCLUSION The combined NRS 2002 and NLR scoring can serve as an effective prognostic tool for NSCLC patients receiving ICIs treatment. This scoring system helps clinicians more accurately identify patients who will benefit from immunotherapy, thereby facilitating more personalized treatment plans. Further validation of this scoring system's applicability and reliability is warranted in future multicenter, large-sample prospective studies.
Collapse
Affiliation(s)
- Jianying Liu
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Jiaxin Liu
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Haiyan Chen
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Qingwei Zhang
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Peihong Zhang
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Zhisheng Chen
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Xuefeng Lu
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China.
| | - Yijiao Xu
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China.
| |
Collapse
|
9
|
Wu Y, Li R, Tan F, Cao J, Bi N. Efficacy of immunotherapy remained in patients with recurrent/metastatic non-small-cell lung cancer after surgery with or without postoperative thoracic radiotherapy: a bi-center retrospective study. Thorac Cancer 2025. [PMID: 40244829 DOI: 10.1111/1759-7714.15384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/04/2024] [Accepted: 05/09/2024] [Indexed: 04/19/2025] Open
Abstract
PURPOSE Since mediastinal lymph node dissection and radiotherapy (RT) have potential unclear impacts on pulmonary lymphatic system, this study aimed to assess the effectiveness of immune checkpoint inhibitors (ICIs) in recurrent/metastatic non-small-cell lung cancer (NSCLC) patients who previously received radical surgery with or without thoracic RT. METHODS Clinical data of patients who underwent pulmonary lobectomy with systematic lymphadenectomy (2000.1.1-2021.7.2) and received immunotherapy after progression were retrospectively analyzed. Efficacy was mainly evaluated based on progression-free survival (PFS) from the start of the ICIs. Toxicity was defined as treatment discontinuation due to immune-related adverse effects (irAEs). RESULTS Ninety-five patients were enrolled in the final cohort and 30 (31.6%) patients received thoracic RT before ICI treatment. ICIs were administered as a first-line systematic treatment in 52.6% of patients. The median follow-up time was 14.7 months (95% confidence interval [CI] 13.3-18.7 months). The median PFS was 12.3 months (95% CI 8.5-36.6 months). Six (6.3%) patients had treatment suspended due to irAEs. Patients who received RT had comparable median PFS with the non-RT group (17.0 months vs. 11.1 months, p = 0.16). Similar toxicity rates were observed. Similar mPFS were reported in the stage III subgroup (RT vs. non-RT, 8.10 vs. 8.45 months, p = 0.86) or the subgroup treated by ICIs as primary systematic therapy (RT vs. non-RT, 13.6 vs. 16.1 months, p = 0.45). CONCLUSIONS ICIs remained effective in recurrent/metastatic NSCLC patients with radical surgery and RT did not significantly compromise therapeutic effects.
Collapse
Affiliation(s)
- Yuqi Wu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Renda Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianzhong Cao
- Department of Radiation Oncology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Nan Bi
- Department of Radiation 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
|
10
|
Arango-Argoty G, Bikiel DE, Sun GJ, Kipkogei E, Smith KM, Carrasco Pro S, Choe EY, Jacob E. AI-driven predictive biomarker discovery with contrastive learning to improve clinical trial outcomes. Cancer Cell 2025:S1535-6108(25)00130-8. [PMID: 40250446 DOI: 10.1016/j.ccell.2025.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/20/2024] [Accepted: 03/26/2025] [Indexed: 04/20/2025]
Abstract
Modern clinical trials can capture tens of thousands of clinicogenomic measurements per individual. Discovering predictive biomarkers, as opposed to prognostic markers, remains challenging. To address this, we present a neural network framework based on contrastive learning-the Predictive Biomarker Modeling Framework (PBMF)-that explores potential predictive biomarkers in an automated, systematic, and unbiased manner. Applied retrospectively to real clinicogenomic datasets, particularly for immuno-oncology (IO) trials, our algorithm identifies biomarkers of IO-treated individuals who survive longer than those treated with other therapies. We demonstrate how our framework retrospectively contributes to a phase 3 clinical trial by uncovering a predictive, interpretable biomarker based solely on early study data. Patients identified with this predictive biomarker show a 15% improvement in survival risk compared to those in the original trial. The PBMF offers a general-purpose, rapid, and robust approach to inform biomarker strategy, providing actionable outcomes for clinical decision-making.
Collapse
Affiliation(s)
| | - Damian E Bikiel
- Oncology Data Science, Oncology R&D, AstraZeneca, Waltham, MA, USA
| | - Gerald J Sun
- Oncology Data Science, Oncology R&D, AstraZeneca, Waltham, MA, USA
| | - Elly Kipkogei
- Oncology Data Science, Oncology R&D, AstraZeneca, Waltham, MA, USA
| | - Kaitlin M Smith
- Oncology Data Science, Oncology R&D, AstraZeneca, Waltham, MA, USA
| | | | - Elizabeth Y Choe
- Oncology Data Science, Oncology R&D, AstraZeneca, Waltham, MA, USA
| | - Etai Jacob
- Oncology Data Science, Oncology R&D, AstraZeneca, Waltham, MA, USA.
| |
Collapse
|
11
|
West M, Yoshida K, Yu J, Lemaire V. Predicting survival in prospective clinical trials using weakly-supervised QSP. NPJ Precis Oncol 2025; 9:106. [PMID: 40229450 PMCID: PMC11997190 DOI: 10.1038/s41698-025-00898-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 03/24/2025] [Indexed: 04/16/2025] Open
Abstract
Quantitative systems pharmacology (QSP) models of cancer immunity provide mechanistic insights into cellular dynamics and drug effects that are difficult to study clinically. However, their inability to predict patient survival mechanistically limits their utility in anti-cancer drug development. To overcome this, we link virtual patients from a QSP model to real clinical trial patients. Using data from atezolizumab trials in non-small cell lung cancer, we show that tumor-based linkage effectively captures survival outcomes. By treating linked survival and censoring as weak supervision labels, we trained survival models using only QSP model covariates, without clinical covariates. Our approach also predicts survival for treatments not included in training data. Specifically, we accurately estimated survival hazard ratios (HR) for chemotherapy monotherapy and atezolizumab plus chemotherapy combination. The predicted HR of 0.70 (95% prediction interval [PI] 0.55-0.86) closely matches the observed HR of 0.79 (95% PI 0.64-0.98) from the IMpower130 trial.
Collapse
Affiliation(s)
- Matthew West
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Modeling and Simulation, Clinical Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Kenta Yoshida
- Modeling and Simulation, Clinical Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Jiajie Yu
- Clinical Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Vincent Lemaire
- Modeling and Simulation, Clinical Pharmacology, Genentech Inc., South San Francisco, CA, USA.
| |
Collapse
|
12
|
Trovarelli G, Rizzo A, Zinnarello FD, Cerchiaro M, Angelini A, Pala E, Ruggieri P. Modern Treatment of Skeletal Metastases: Multidisciplinarity and the Concept of Oligometastasis in the Recent Literature. Curr Oncol 2025; 32:226. [PMID: 40277781 DOI: 10.3390/curroncol32040226] [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: 03/11/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Bone metastases are a major concern in cancer management since they significantly contribute to morbidity and mortality. Metastatic lesions, commonly arising from breast, prostate, lung, and kidney cancers, affect approximately 25% of cancer patients, leading to severe complications such as pain, fractures, and neurological deficits. This narrative review explores contemporary approaches to bone metastases, emphasizing a multidisciplinary strategy and the evolving concept of oligometastatic disease. Oligometastases, defined by limited metastatic spread (1-5 lesions), offer a potential window for curative treatment through aggressive interventions, including stereotactic ablative radiotherapy and resection surgery. Tumor boards, integrating systemic therapies with local interventions, are crucial to optimize treatment. Despite promising results, gaps remain in defining optimal treatment sequences and refining patient selection criteria. Future research should focus on personalized approaches, leveraging biomarkers and advanced imaging to enhance outcomes and the quality of life in patients with bone metastases.
Collapse
Affiliation(s)
- Giulia Trovarelli
- Department of Orthopedics and Orthopedic Oncology, University of Padua, 35122 Padua, Italy
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, 35122 Padua, Italy
| | - Arianna Rizzo
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Felicia Deborah Zinnarello
- Department of Orthopedics and Orthopedic Oncology, University of Padua, 35122 Padua, Italy
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, 35122 Padua, Italy
| | - Mariachiara Cerchiaro
- Department of Orthopedics and Orthopedic Oncology, University of Padua, 35122 Padua, Italy
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, 35122 Padua, Italy
| | - Andrea Angelini
- Department of Orthopedics and Orthopedic Oncology, University of Padua, 35122 Padua, Italy
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, 35122 Padua, Italy
| | - Elisa Pala
- Department of Orthopedics and Orthopedic Oncology, University of Padua, 35122 Padua, Italy
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, 35122 Padua, Italy
| | - Pietro Ruggieri
- Department of Orthopedics and Orthopedic Oncology, University of Padua, 35122 Padua, Italy
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, 35122 Padua, Italy
| |
Collapse
|
13
|
Nam C, Huang G, Zheng Y, Zhao H, Pan Y, Hu B, Wenger T, Van HT, Xu LY, Li EM, Koeffler HP, Ge K, Dou Y, Sinha UK, Park YM, Lin DC. The MLL3/GRHL2 complex regulates malignant transformation and anti-tumor immunity in squamous cancer. J Exp Med 2025; 222:e20240758. [PMID: 39964485 PMCID: PMC11834937 DOI: 10.1084/jem.20240758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 10/28/2024] [Accepted: 01/06/2025] [Indexed: 02/20/2025] Open
Abstract
Upper aerodigestive squamous cell carcinoma (UASCC) presents significant challenges in clinical management due to its aggressive nature. Here, we elucidate the role of MLL3 mutations as early, clonal genomic events in UASCC tumorigenesis, highlighting their role as foundational drivers of cancer development. Utilizing CRISPR-edited, cross-species organoid modeling, we demonstrate that loss of MLL3 contributes to early squamous neoplastic evolution. Furthermore, we identify an MLL3/GRHL2 protein complex that regulates the UASCC epigenome, particularly impacting immune response pathways. Notably, a novel MLL3/GRHL2-IRF1 axis promotes the expression of Th1 chemokines, enhancing anti-tumor immunity by facilitating T cell infiltration into the tumor microenvironment. Consequently, MLL3 regulates the in vivo efficacy of immune checkpoint blockade (ICB) therapy, corroborated by the strong association between MLL3 expression and human patients' clinical response to ICB therapy. Our work underscores the significance of MLL3 in UASCC pathogenesis and highlights the interplay between MLL3/GRHL2 and immune response pathways as potential therapeutic targets for UASCC treatment.
Collapse
Affiliation(s)
- Chehyun Nam
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Guowei Huang
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, China
| | - Yueyuan Zheng
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hua Zhao
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Yuhao Pan
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Boyan Hu
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Talia Wenger
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Hieu T. Van
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Li-Yan Xu
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, China
| | - En-Min Li
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, China
| | | | - Kai Ge
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yali Dou
- Department of Medicine, Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA, USA
| | - Uttam K. Sinha
- Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Young Min Park
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, South Korea
| | - De-Chen Lin
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
14
|
Hughes DJ, Chand G, Johnson J, Tegala R, Bailey D, Adamson K, Edmonds S, Meszaros LK, Moore AEB, Manickavasagar T, Ndagire S, Gennatas S, Georgiou A, Ghosh S, Josephs D, Karapanagiotou E, McLean E, Ting HH, Spicer J, Goh V, Cook GJR. PD-L1 imaging with [ 99mTc]NM-01 SPECT/CT is associated with metabolic response to pembrolizumab with/without chemotherapy in advanced lung cancer. Br J Cancer 2025:10.1038/s41416-025-02991-w. [PMID: 40188291 DOI: 10.1038/s41416-025-02991-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/15/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) immunohistochemistry is a predictive biomarker for anti-PD-(L)1 therapy in non-small cell lung cancer (NSCLC). It is not a reliable predictor of clinical benefit with non-invasive imaging providing a potential solution. We present the PECan study, the aim of which to assess the relationship of [99mTc]-labeled anti-PD-L1 single-domain antibody (NM-01) single-photon emission computed tomography (SPECT)/CT with metabolic response to anti-PD-(L)1. METHODS PD-L1 tumour proportion score (TPS) measured using SP263 assay. [99mTc]NM-01 SPECT/CT and [18F]FDG PET/CT performed before and 9-weeks following pembrolizumab with/without chemotherapy in patients with advanced NSCLC. Tumor (T) to blood pool (BP) maximum region of interest (ROImax) measurements performed in primary and metastatic lesions using SPECT/CT images. RESULTS Fifteen patients were included (median age 63 years, 9 male). Intertumoural heterogeneity evident in 10(67%) patients. Mean [99mTc]NM-01 T:BP demonstrated moderate correlation with PD-L1 TPS (r = 0.45, p < 0.05). Depth of [18F]FDG PET/CT metabolic response at 9-weeks (n = 13), correlated strongly with baseline [99mTc]NM-01 T:BP (r = -0.73, p < 0.05), but only moderately with PD-L1 TPS (r = -0.46, p = 0.06). CONCLUSION [99mTc]NM-01 SPECT/CT allows non-invasive quantification of PD-L1 in primary tumour and metastases in NSCLC. [99mTc]NM-01 uptake moderately correlates with PD-L1 immunohistochemistry, determines heterogeneity, and is associated with early metabolic response to anti-PD-1 pembrolizumab. CLINICAL TRIALS REGISTRATION PD-L1 Expression in Cancer (PECan) study (NCT04436406), registered 18 June 2020 https://clinicaltrials.gov/ct2/show/NCT04436406.
Collapse
Affiliation(s)
- Daniel Johnathan Hughes
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- King's College London & Guy's and St. Thomas' PET Centre, St Thomas' Hospital, London, UK
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Gitasha Chand
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- Nanomab Technology (UK) Limited, Borehamwood, Hertfordshire, UK
| | - Jessica Johnson
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Ronan Tegala
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Damion Bailey
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Kathryn Adamson
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Scott Edmonds
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | | | - Amelia Elizabeth Broomfield Moore
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
| | - Thubeena Manickavasagar
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Department of Radiology, Guy's and St. Thomas' NHS Foundation Trust, St Thomas' Hospital, London, UK
| | - Susan Ndagire
- King's Health Partners Cancer Biobank, Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, Great Maze Pond, London, UK
| | - Spyridon Gennatas
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Alexandros Georgiou
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Sharmistha Ghosh
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Debra Josephs
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Eleni Karapanagiotou
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Emma McLean
- Department of Histopathology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Hong Hoi Ting
- Nanomab Technology (UK) Limited, Borehamwood, Hertfordshire, UK
| | - James Spicer
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Vicky Goh
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- Department of Radiology, Guy's and St. Thomas' NHS Foundation Trust, St Thomas' Hospital, London, UK
| | - Gary J R Cook
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK.
- King's College London & Guy's and St. Thomas' PET Centre, St Thomas' Hospital, London, UK.
| |
Collapse
|
15
|
Xie D, Yu J, He C, Jiang H, Qiu Y, Fu L, Kong L, Xu H. Predicting the immune therapy response of advanced non-small cell lung cancer based on primary tumor and lymph node radiomics features. Front Med (Lausanne) 2025; 12:1541376. [PMID: 40248083 PMCID: PMC12003267 DOI: 10.3389/fmed.2025.1541376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/20/2025] [Indexed: 04/19/2025] Open
Abstract
Objective To identify imaging biomarkers of primary tumors and lymph nodes in patients with stage III-IV non-small cell lung cancer (NSCLC) and assess their predictive ability for treatment response (response vs. non-response) to immune checkpoint inhibitors (ICIs) after 6 months. Methods Retrospective analysis of 83 NSCLC patients treated with ICIs. Quantitative imaging features of the maximum primary lung tumors and lymph nodes on contrast-enhanced CT imaging were extracted at baseline (time point 0, TP0) and after 2-3 cycles of immunotherapy (time point 1, TP1). Delta-radiomics features (delta-RFs) were defined as the net changes in radiomics features (RFs) between TP0 and TP1. Interobserver interclass coefficient (ICC) and Pearson correlation analyses were applied for feature selection, and logistic regression (LR) was used to build a model for predicting treatment response. Results Four and five important delta-RFs were selected to construct the nodal and tumor models, respectively. Δ Tumor diameter was used for constructing the clinical prediction model. The predictive efficacy of the nodal model for the treatment response status was higher than that of the tumor and clinical models. In the training set, the AUC values for the three models were 0.96 (95% CI = 0.90-1.00), 0.86 (95% CI = 0.76-0.95), and 0.82 (95% CI = 0.71-0.93), respectively. In the validation set, the AUC values were 0.94 (95% CI = 0.85-1.00), 0.77 (95% CI = 0.56-0.98), and 0.74 (95% CI = 0.48-1.00), respectively. Conclusion The nodal model based on delta-RFs performed well in distinguishing responders from non-responders and could identify patients more likely to benefit from immunotherapy. Finally, the nodal model exhibited a higher classification performance than the tumor model.
Collapse
Affiliation(s)
- Dong Xie
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Jinna Yu
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Cong He
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Han Jiang
- Department of Medical Oncology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Yonggang Qiu
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Linfeng Fu
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Lingting Kong
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Hongwei Xu
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| |
Collapse
|
16
|
Jin CX, Liu YS, Qin HN, Teng YB, Sun R, Ma ZJ, Wang AM, Liu JW. Peripheral inflammatory factors as prognostic predictors for first-line PD-1/PD-L1 inhibitors in advanced non-small cell lung cancer. Sci Rep 2025; 15:11206. [PMID: 40175366 PMCID: PMC11965408 DOI: 10.1038/s41598-024-84469-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 12/24/2024] [Indexed: 04/04/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have significantly improved the efficacy and prognosis of patients with non-small cell lung cancer (NSCLC). However, there remains a lack of optimal predictive biomarkers for assessing the response of ICIs. This study aimed to evaluate peripheral inflammatory factors as potential predictive biomarkers for NSCLC patients treated with ICIs. We retrospectively analyzed the correlation between peripheral inflammatory factors and the efficacy and prognosis of 124 patients with driver gene-negative advanced NSCLC who received first-line ICIs at our center from September 2018 to June 2022. Progression-free survival (PFS) was estimated using the Kaplan-Meier method. The association between the factors and multiple endpoints were investigated using univariate and multivariate analyses. A total of 124 patients were enrolled in this study. The objective response rate (ORR) was 49.2% and the disease control rate (DCR) was 97.6%, respectively. The median PFS was 12.7 months. The ORR differed statistically between groups based on the NLR, SII, with higher ORR observed in patients with an NLR ratio < 0.68, SII at 6 weeks < 531.26, and SII ratio < 0.74 (p < 0.05). The univariate analysis indicated that ECOG 0-1, smoking, NLR at 6 weeks < 2.72, NLR ratio < 0.68, LMR < 1.34, LMR ratio ≥ 1.38, and SII at 6 weeks < 531.26 were associated with longer PFS (p < 0.05). The multivariate analysis revealed that smoking (p = 0.013), baseline LMR (p = 0.015), and SII at 6 weeks (p = 0.010) were independent predictors of PFS. NLR, LMR, and SII maybe biomarkers for predicting the efficacy and prognosis of first-line ICIs therapy in driver gene-negative advanced NSCLC.
Collapse
Affiliation(s)
- Chen-Xing Jin
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Yan-Song Liu
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
- Department of Anesthesiology, Obstetrics & Gynecology Hospital of Fudan University, Shanghai, 200011, Shanghai, China
| | - He-Nan Qin
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Yi-Bin Teng
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Rui Sun
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Zhong-Jing Ma
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - A-Man Wang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China.
| | - Ji-Wei Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China.
| |
Collapse
|
17
|
Cho BC, Johnson M, Bar J, Schaefer E, Yoh K, Zer A, Moskovitz M, Lee SH, Moreno V, de Miguel M, Okuma Y, Kim JH, Lee CH, Peguero J, Ansell P, Biesdorf C, Saab R, Freise KJ, Ramies D, Jeng EE, Camidge DR. A phase 1b study of cofetuzumab pelidotin monotherapy in patients with PTK7-expressing recurrent non-small cell lung cancer. Lung Cancer 2025; 202:108492. [PMID: 40086026 DOI: 10.1016/j.lungcan.2025.108492] [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: 10/24/2024] [Revised: 02/24/2025] [Accepted: 03/08/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Protein tyrosine kinase 7 (PTK7) overexpression in lung cancer is associated with tumor progression. Cofetuzumab pelidotin (Cofe-P) is an antibody-drug conjugate comprising an anti-PTK7 antibody conjugated to a microtubule inhibitor. Herein, we report the results of a phase 1b study evaluating Cofe-P safety, efficacy, and pharmacokinetics in patients with recurrent non-small cell lung cancer (NSCLC). METHODS This signal-seeking phase 1b, open-label, multicenter, single-arm study enrolled patients with PTK7-expressing recurrent NSCLC. Cofe-P was administered at 2.8 mg/kg intravenously once every 3 weeks. The primary endpoint was objective response rate (ORR) and secondary endpoints were duration of response (DOR), progression-free survival (PFS), and overall survival (OS). RESULTS Overall, 65 patients received Cofe-P; 51 had PTK7 expression of ≥90 % tumor cells with ≥2+ staining intensity by immunohistochemistry. All patients reported adverse events (AEs), most commonly alopecia (52 %) and decreased neutrophil count (43 %); 69 % had grade ≥3 AEs, including grade ≥3 neutropenia in 25 %. Treatment-related AEs were reported in 94 % of patients; none were fatal. Overall, ORR was 18 %; in patients with PTK7 expression of ≥90 % tumor cells with ≥2+ staining and non-squamous epidermal growth factor receptor (EGFR) wild type or mutant, or squamous NSCLC, ORR was 21 %, 15 %, and 13 %, respectively. Overall, median DOR was 7.2 months, median PFS was 5.5 months, and median OS was 12.6 months; longest DOR (median 9.0 months), PFS (median 6.8 months), and OS (median 12.6 months) were in patients with non-squamous EGFR-mutant NSCLC. CONCLUSIONS Cofe-P demonstrated a manageable safety profile and preliminary efficacy across NSCLC histologies and EGFR mutation status. These data support PTK7 as a valid therapeutic target for NSCLC.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/metabolism
- Male
- Female
- Middle Aged
- Aged
- Lung Neoplasms/drug therapy
- Lung Neoplasms/mortality
- Lung Neoplasms/pathology
- Lung Neoplasms/metabolism
- Adult
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/pathology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Cell Adhesion Molecules/metabolism
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptor Protein-Tyrosine Kinases/antagonists & inhibitors
- Aged, 80 and over
- Immunoconjugates/therapeutic use
- Immunoconjugates/pharmacokinetics
- Immunoconjugates/adverse effects
- Treatment Outcome
- Antigens, Neoplasm/metabolism
Collapse
Affiliation(s)
- Byoung Chul Cho
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | | | - Jair Bar
- Jusidman Cancer Center, Institute of Oncology, Sheba Medical Center, Ramat Gan, Israel.
| | - Eric Schaefer
- Section of Medical Oncology, Highlands Oncology Group, Fayetteville, AR, USA.
| | - Kiyotaka Yoh
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| | - Alona Zer
- Oncology Institute, Rambam Health Care Campus, Rappaport Faculty of Medicine, Technion, Haifa, Israel.
| | - Mor Moskovitz
- Thoracic Cancer Service, Rabin Medical Center Davidoff Cancer Center, Beilinson Campus, Petah Tikva, Israel.
| | - Se-Hoon Lee
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Victor Moreno
- START Madrid-FJD, Medical Oncology Department, University Hospital Fundación Jiménez Diaz, Instituto de Investigación Sanitaria FJD, Madrid, Spain.
| | - Maria de Miguel
- START Madrid HM CIOCC, Instituto de Investigación Sanitaria HM Hospitales, Universidad San Pablo CEU, Madrid, Spain.
| | - Yusuke Okuma
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Joo-Hang Kim
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea.
| | - Chun-Hui Lee
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City, Taiwan.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Busakhala N, Atundo L, Kiprono H, Keitany K, Melly E, Ruto R, Wanja M, Chepsiror D, Rangoonwala H, Kipchirchir C, Chesori E, Oguda J, Opakas J, Loehrer PJ, Diero L, Morgan J. Characteristics and Associated Survival of Patients Diagnosed With Non-Small Cell Lung Cancer in a Designated Lung Cancer Program in Western Kenya. JCO Glob Oncol 2025; 11:e2400212. [PMID: 40184569 DOI: 10.1200/go.24.00212] [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: 05/16/2024] [Revised: 10/10/2024] [Accepted: 02/11/2025] [Indexed: 04/06/2025] Open
Abstract
PURPOSE Although lung cancer is a major cause of cancer incidence and mortality worldwide, lung cancer studies in sub-Saharan Africa are scarce. Here, we present outputs from a designated lung cancer program in western Kenya, part of the Multi-National Lung Cancer Control Program, which focused on case finding, diagnosis, and treatment. METHODS We retrospectively reviewed patients with pathologically confirmed non-small cell lung cancer (NSCLC) enrolled in this program at Moi Teaching and Referral Hospital from January 2018 to December 2022. Clinical data were analyzed using descriptive statistics, Kaplan-Meier methods, and proportional hazards regression model. RESULTS Two hundred forty-nine patients diagnosed with NSCLC were included with a median age at diagnosis of 61 (IQR, 52-70) years. Most patients were married (n = 177; 71%) and nonsmokers (n = 177; 71%) with 58 (23%) having received tuberculosis treatment and 93 (37%) having Eastern Cooperative Oncology Group (ECOG) performance status (PS) of ≥ 2. At diagnosis, adenocarcinoma was the prominent histology (n = 187; 75%) along with clinical stage IV (n = 195; 78% stage IV) or unstaged (n = 40; 16%) disease. Most patients received chemotherapy and radiotherapy (n = 176; 71%) with few palliative care referrals (n = 2; 0.8%). The median overall survival (OS) was only 3.7 months (IQR, 2.7-5.4). ECOG PS (3 or 4) and being unstaged were predictors of poor 1-year OS. CONCLUSION Patients with NSCLC enrolled in this program presented with advanced disease and poor survival. Despite a designated case finding effort, late diagnosis remained common and highlights a need for locally relevant interventions targeting community and provider education as well as innovative diagnostics that can improve early recognition of lung cancer. These interventions must also be paired with access to proven treatments including molecular therapies and palliative care which can extend lung cancer survival.
Collapse
Affiliation(s)
- Naftali Busakhala
- Academic Model for Providing Access to Healthcare (AMPATH), Eldoret, Kenya
- Moi University College of Health Sciences, Eldoret, Kenya
- Moi University Teaching and Referral Hospital, Eldoret, Kenya
| | - Lawrence Atundo
- Academic Model for Providing Access to Healthcare (AMPATH), Eldoret, Kenya
| | | | - Kibet Keitany
- Moi University Teaching and Referral Hospital, Eldoret, Kenya
| | - Elias Melly
- Kenya National Cancer Institute, Nairobi, Kenya
| | - Ruth Ruto
- Academic Model for Providing Access to Healthcare (AMPATH), Eldoret, Kenya
| | - Madrine Wanja
- Academic Model for Providing Access to Healthcare (AMPATH), Eldoret, Kenya
| | | | | | | | - Erick Chesori
- Moi University Teaching and Referral Hospital, Eldoret, Kenya
| | - John Oguda
- Indiana University School of Medicine, Indianapolis, IN
| | - Jesse Opakas
- Moi University Teaching and Referral Hospital, Eldoret, Kenya
| | - Patrick J Loehrer
- Academic Model for Providing Access to Healthcare (AMPATH), Eldoret, Kenya
- Indiana University School of Medicine, Indianapolis, IN
| | - Lameck Diero
- Academic Model for Providing Access to Healthcare (AMPATH), Eldoret, Kenya
- Moi University College of Health Sciences, Eldoret, Kenya
- Moi University Teaching and Referral Hospital, Eldoret, Kenya
| | - Jennifer Morgan
- Academic Model for Providing Access to Healthcare (AMPATH), Eldoret, Kenya
- Indiana University School of Medicine, Indianapolis, IN
- University of Minnesota School of Medicine, Minneapolis, MN
| |
Collapse
|
19
|
Park BJ, Warning AW, Akella SS. Checkpoint inhibitor-related myasthenia-myocarditis-myositis overlap syndrome in the orbit. Orbit 2025; 44:200-206. [PMID: 38796779 DOI: 10.1080/01676830.2024.2351519] [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: 03/03/2024] [Accepted: 04/30/2024] [Indexed: 05/29/2024]
Abstract
We report a series of three patients who developed checkpoint inhibitor-related myocarditis with orbital myositis and/or myasthenia gravis overlap syndrome, with varying degrees of severity. In all cases, checkpoint inhibitor therapy was immediately discontinued upon diagnosis and corticosteroids were initiated. While two patients achieved substantial recovery, one patient passed away on hospital day three. These cases underscore the critical need for prompt recognition of adverse events associated with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Benjamin Jinsung Park
- Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center and The Ohio State University Comprehensive Cancer Center - James Cancer Center and Solove Research Institute, Columbus, Ohio, USA
| | - Aaron W Warning
- Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center and The Ohio State University Comprehensive Cancer Center - James Cancer Center and Solove Research Institute, Columbus, Ohio, USA
| | - Sruti S Akella
- Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center and The Ohio State University Comprehensive Cancer Center - James Cancer Center and Solove Research Institute, Columbus, Ohio, USA
| |
Collapse
|
20
|
Ziogas DC, Theocharopoulos C, Aravantinou K, Boukouris AE, Stefanou D, Anastasopoulou A, Lialios PP, Lyrarakis G, Gogas H. Clinical benefit of immune checkpoint inhibitors in elderly cancer patients: Current evidence from immunosenescence pathophysiology to clinical trial results. Crit Rev Oncol Hematol 2025; 208:104635. [PMID: 39889861 DOI: 10.1016/j.critrevonc.2025.104635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025] Open
Abstract
The age-related decline in immunity appears to be associated not only with cancer development but also with differential responses to immune checkpoint inhibitors (ICIs). Despite their increasing utility across various malignancies and therapeutic settings, limited data -derived primarily from subgroup analyses of randomized controlled trials (RCTs), pooled meta-analyses, and retrospective studies- are available on the effects of aging on their efficacy and toxicity. Immunosenescence, characterized by the progressive decline of the function of the immune system, and inflammaging, a state of persistent low-grade sterile inflammation, may influence ICI outcomes. Additionally, the incidence, severity, and subtypes of immune-related adverse events (irAEs) may differ between older and younger individuals due to loss of immunotolerance. In the current review, starting from a a comprehensive discussion of the pathophysiology of immunosenescence, we proceed to critically review age-related retrospective and randomized evidence supporting FDA-approved ICIs. We highlight similarities or differences across age groups and the clinical benefit of ICIs in elderly versus younger cancer patients. The optimal integration of ICIs in geriatric oncology necessitates greater inclusion of this patient demographic in RCTs along with real-world data in order to acquire robust data which will guide evidence-based treatment decisions for this population.
Collapse
Affiliation(s)
- Dimitrios C Ziogas
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Charalampos Theocharopoulos
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Katerina Aravantinou
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Aristeidis E Boukouris
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Dimitra Stefanou
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Amalia Anastasopoulou
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Panagiotis-Petros Lialios
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - George Lyrarakis
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| |
Collapse
|
21
|
Hattori S, Uno H. On Sample Size Determination for Augmented Tests Based on Restricted Mean Survival Time in Randomized Clinical Trials. Biom J 2025; 67:e70046. [PMID: 40123326 PMCID: PMC11931358 DOI: 10.1002/bimj.70046] [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: 09/04/2023] [Revised: 09/30/2024] [Accepted: 10/29/2024] [Indexed: 03/25/2025]
Abstract
Restricted mean survival time (RMST) is gaining attention as a measure to quantify the treatment effect on survival outcomes in randomized clinical trials. Several methods to determine sample size based on the RMST-based tests have been proposed. However, to the best of our knowledge, there is no discussion about the power and sample size regarding the augmented version of RMST-based tests, which utilize baseline covariates for a gain in estimation efficiency and in power for testing no treatment effect. The conventional event-driven study design based on the logrank test allows us to calculate the power for a given hazard ratio without specifying the survival functions. In contrast, the existing sample size determination methods for the RMST-based tests relies on the adequacy of the assumptions of the entire survival curves of two groups. Furthermore, to handle the augmented test, the correlation between the baseline covariates and the martingale residuals must be handled. To address these issues, we propose an approximated sample size formula for the augmented version of the RMST-based test, which does not require specifying the entire survival curve in the treatment group, and also a sample size recalculation approach to update the correlations between the baseline covariates and the martingale residuals with the blinded data. The proposed procedure will enable the studies to have the target power for a given RMST difference even when correct survival functions cannot be specified at the design stage.
Collapse
Affiliation(s)
- Satoshi Hattori
- Department of Biomedical StatisticsGraduate School of Medicine and Integrated Frontier Research for Medical Science DivisionInstitute for Open and Transdisciplinary Research Initiatives (OTRI)Osaka UniversitySuita CityOsakaJapan
| | - Hajime Uno
- Department of Medical OncologyDivision of Population SciencesDana‐Farber Cancer InstituteDepartment of MedicineHarvard Medical SchoolMassachusettsUSA
| |
Collapse
|
22
|
Zhou DDX, Dalrymple J, Klingberg D, Lin FPY, Lord SJ, Cooper WA, Zaheed M, Simes RJ, John T, Lee CK. Clinical Impact of Somatic Genomic Variants of Oncogenes and Tumor Suppressor Genes in Previously Treated Advanced Non-Small Cell Lung Cancer. JCO Precis Oncol 2025; 9:e2400673. [PMID: 40239138 DOI: 10.1200/po-24-00673] [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: 10/09/2024] [Revised: 12/18/2024] [Accepted: 03/12/2025] [Indexed: 04/18/2025] Open
Abstract
PURPOSE Next-generation sequencing in non-small cell lung cancer (NSCLC) identifies somatic genomic variants (SGVs) in cancer susceptibility genes (CSGs). We hypothesized that SGVs would be associated with poorer overall survival (OS) but greater benefit with immune checkpoint inhibitors over chemotherapy. We investigated the prevalence and predictive value of SGVs, using data from OAK and POPLAR trials comparing atezolizumab with docetaxel. METHODS We curated a list of SGVs (excluding TP53, EGFR, ALK, and ROS1) on the basis of CSGs associated with tumorigenesis. We classified participants as SGV mutant or wild-type using baseline plasma analyzed by the FoundationOne Liquid CDx assay. Cox regression analyses and interaction tests between SGV status and treatment were performed. RESULTS Of 762 participants, 29% harbored an SGV. The SGV mutant group had worse OS (hazard ratio [HR], 1.28, 95% CI, 1.06 to 1.54), and within each treatment arm (docetaxel: HR, 1.31; atezolizumab: HR, 1.27). In the atezolizumab arm, the SGV mutant group compared with wild-type had worse OS in the PD-L1 high (HR, 1.31 [95% CI, 0.59 to 2.91]) and low (HR, 1.38 [95% CI, 0.98 to 1.93]) subgroups. SGV with missense, splice, and nonsense mutations had significantly worse OS than wild-type in the docetaxel arm (log-rank P = .01) but not in the atezolizumab arm (log-rank P = .33). SGV status did not predict greater OS benefit with atezolizumab over docetaxel (interaction P = .67). CONCLUSION In advanced NSCLC after chemotherapy progression, plasma-detected SGVs are common, and associated with inferior OS. Plasma SGV status should be considered as a stratification factor in future trials.
Collapse
Affiliation(s)
- Deborah Di-Xin Zhou
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- St George Hospital, Kogarah, NSW, Australia
| | | | | | - Frank Po-Yen Lin
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Sarah J Lord
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Wendy A Cooper
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, NSW Health Pathology, Camperdown, NSW, Australia
- Faculty of Medicine and Health and Western Sydney University School of Medicine, University of Sydney, Camperdown, NSW, Australia
| | - Milita Zaheed
- Prince of Wales Hereditary Cancer Centre, Randwick, NSW, Australia
| | - Robert John Simes
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Thomas John
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Chee Khoon Lee
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- St George Hospital, Kogarah, NSW, Australia
| |
Collapse
|
23
|
Zhang W, Wang J, Liang J, He Z, Wang K, Lin H. RNA methylation of CD47 mediates tumor immunosuppression in EGFR-TKI resistant NSCLC. Br J Cancer 2025; 132:569-579. [PMID: 39900985 PMCID: PMC11920402 DOI: 10.1038/s41416-025-02945-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Although immune checkpoint inhibitors (ICIs) have been successfully utilized in patients with non-small cell lung cancer (NSCLC), EGFR-mutated patients didn't benefit from ICIs. The underlying mechanisms for the poor efficacy of this subgroup remain unclear. METHODS CD8+T cells cytotoxicity, DCs phagocytosis and immunofluorescence assay were applied to examine the immunosuppressive microenvironment of NSCLC. m6A RNA immunoprecipitation, luciferase assay and immunohistochemistry were used to explore the relationship between CD47 and ALKBH5 in EGFR-TKI resistant NSCLC. Autochthonous EGFR-driven lung tumor mouse model and PDXs were performed to explore the therapeutic potential of CD47 antibody and EGFR-TKI combination. RESULTS We found that EGFR-TKI resistance promoted a more immunosuppressive tumor microenvironment and inhibited anti-tumor functions of CD8+ T cells. Mechanistically, the m6A eraser ALKBH5 was inhibited in EGFR-TKI resistant NSCLC, which subsequently upregulates CD47 by catalyzing m6A demethylation and causes immunosuppression. Combined treatment with EGFR-TKI and inhibitors of CD47 enhances antitumor immunity and EGFR-TKI efficacy in vivo. CONCLUSIONS Collectively, our findings reveal the possible underlying mechanism for poor immune response of ICIs in EGFR-TKI resistant NSCLC and provide preclinical evidence that targeted therapy combined with innate immune checkpoint blockade may provide synergistic effects in NSCLC treatment.
Collapse
Affiliation(s)
- Wei Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jiawen Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jialu Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhanghai He
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Kefeng Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Huayue Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
24
|
Chung C, Umoru G. Prognostic and predictive biomarkers with therapeutic targets in nonsmall-cell lung cancer: A 2023 update on current development, evidence, and recommendation. J Oncol Pharm Pract 2025; 31:438-461. [PMID: 38576390 DOI: 10.1177/10781552241242684] [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: 04/06/2024]
Abstract
BackgroundSince the publication of the original work in 2014, significant progress has been made in the characterization of genomic alterations that drive oncogenic addiction of nonsmall cell lung cancer (NSCLC) and how the immune system can leverage non-oncogenic pathways to modulate therapeutic outcomes. This update evaluates and validates the recent and emerging data for prognostic and predictive biomarkers with therapeutic targets in NSCLC.Data sourcesWe performed a literature search from January 2015 to October 2023 using the keywords non-small cell lung cancer, clinical practice guidelines, gene mutations, genomic assay, immune cancer therapy, circulating tumor DNA, predictive and prognostic biomarkers, and targeted therapies.Study selection and data extractionWe identified, reviewed, and evaluated relevant clinical trials, meta-analyses, seminal articles, and published clinical practice guidelines in the English language.Data synthesisRegulatory-approved targeted therapies include those somatic gene alterations of EGFR ("classic" mutations, exon 20 insertion, and rare EGFR mutations), ALK, ROS1, BRAF V600, RET, MET, NTRK, HER2, and KRAS G12C. Data for immunotherapy and circulating tumor DNA in next-generation sequencing are considered emerging, whereas the predictive role for PIK3CA gene mutation is insufficient.ConclusionsAdvances in sequencing and other genomic technologies have led to identifying novel oncogenic drivers, novel resistance mechanisms, and co-occurring mutations that characterize NSCLC, creating further therapeutic opportunities. The benefits associated with immunotherapy in the perioperative setting hold initial promise, with their long-term results awaiting.
Collapse
Affiliation(s)
- Clement Chung
- Department of Pharmacy, Houston Methodist West Hospital, Houston, TX, USA
| | - Godsfavour Umoru
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
25
|
Wang T, Chen L, Bao X, Han Z, Wang Z, Nie S, Gu Y, Gong J. Short-term peri- and intra-tumoral CT radiomics to predict immunotherapy response in advanced non-small cell lung cancer. Transl Lung Cancer Res 2025; 14:785-797. [PMID: 40248738 PMCID: PMC12000948 DOI: 10.21037/tlcr-24-973] [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: 10/21/2024] [Accepted: 02/08/2025] [Indexed: 04/19/2025]
Abstract
Background Predicting response to immunotherapy is crucial for advanced non-small cell lung cancer (NSCLC) treatment planning, but effective predictive markers for immunotherapy efficacy are still lacking. This study aimed to develop an explainable machine learning model for predicting immunotherapy responses in advanced NSCLC patients. Methods A total of 245 advanced NSCLC patients from two centers who received immunotherapy were retrospectively enrolled. For each primary tumor, three regions of interest were analyzed, namely, the intratumoral region (ITR), peritumoral region (PTR), and combined intratumoral and PTR (IPTR). Pre-radiomics features and delta-radiomics features reflecting the rate of change between radiomics features before and after treatment were extracted. Models for predicting immunotherapy responses were established via the extreme gradient boosting (XGBoost) classifier and assessed in terms of discrimination, calibration, and clinical utility. The SHapley Additive exPlanations (SHAP) tool was employed to explore the interpretability of the model. Kaplan-Meier (KM) analysis of progression-free survival (PFS) was conducted to evaluate the prognostic value of the prediction models. Results The delta-radiomics models of ITR and IPTR demonstrated optimal performance in predicting immunotherapy response, significantly improving the area under the curve (AUC) to 0.85 and 0.83 in the internal validation cohort and 0.84 and 0.86 in the external validation cohort. SHAP revealed a strong relationship between the delta-radiomics feature values and the model-predicted probabilities. KM curves indicated that the high-risk groups identified by the delta-radiomics models had significantly worse PFS than did the low-risk groups across all cohorts. Conclusions The results demonstrated that a model based on multiple time points outperformed one based on a single time point. The delta-radiomics model has been proved a noninvasive approach for assessing the response of advanced NSCLC patients to immunotherapy and facilitates individualized treatment decision making.
Collapse
Affiliation(s)
- Ting Wang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei Chen
- Department of Radiology, Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiao Bao
- Department of Radiology, Shanghai Pulmonary Hospital, Shanghai, China
| | - Zijuan Han
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Zezhou Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Cancer Prevention, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shengdong Nie
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing Gong
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Cosci I, De Toni L, Del Fiore P, Di Nisio A, Carraro S, Radu CM, Bertazza L, Mocellin S, Pigozzo J, Crivellaro G, Coppola M, Ferlin A. Anti-CTLA-and anti-PD-1 immune checkpoint inhibitor antibodies impair human sperm motility in-vitro. Front Pharmacol 2025; 16:1534975. [PMID: 40230698 PMCID: PMC11994717 DOI: 10.3389/fphar.2025.1534975] [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: 11/26/2024] [Accepted: 03/21/2025] [Indexed: 04/16/2025] Open
Abstract
Background Immune checkpoint inhibitors (ICIs), namely, anti-cytotoxic T lymphocyte-associated protein 4 (CTLA-4) monoclonal antibody Ipilimumab and anti- and programmed cell death 1 (PD-1) monoclonal antibodies Nivolumab, and Pembrolizumab, have improved the treatment outcomes for many other cancer types. However, their impact on fertility remains under-explored. Methods The possible direct effects of ICIs on human sperm was investigated. Spermatozoa from ten normozoospermic donors were exposed to Ipilimumab, Nivolumab, or Pembrolizumab at concentrations ranging from 1 to 100 ng/mL. Sperm motility was assessed through standard laboratory process. Cell viability and apoptosis markers were evaluated by flow-cytometry using fluorescent Annexin-V probe and Terminal Uridine Nick-End Label (TUNEL) assays. Protein-A-purified therapeutic antibodies (IgG) were also evaluated. Results Spermatozoa had high PD-1 (>99%) and negligible CTLA-4 expression. Exposure to ICIs, was associated with a concentration-dependent impairment of sperm motility, noticeable for Pembrolizumab and Ipilimumab since 10 ng/mL, and for Nivolumab since 100 ng/mL. However, no significant effect on cell apoptosis or viability was shown. Purified IgG from ICIs maintained the adverse effect on cell motility without affecting viability. Conclusion ICIs, specifically Pembrolizumab, Nivolumab, and Ipilimumab, adversely affect human sperm motility in vitro. Further research is required to understand the underlying mechanisms and clinical implications.
Collapse
Affiliation(s)
- Ilaria Cosci
- Department of Medicine, Unit of Andrology and Reproductive Medicine, University of Padova, Padova, Italy
| | - Luca De Toni
- Department of Medicine, Unit of Andrology and Reproductive Medicine, University of Padova, Padova, Italy
| | - Paolo Del Fiore
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Andrea Di Nisio
- Department of Medicine, Unit of Andrology and Reproductive Medicine, University of Padova, Padova, Italy
- Department of Health, Nutrition and Sport, Pegaso Telematic University, Naples, Italy
| | - Samuela Carraro
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
| | - Claudia Maria Radu
- Department of Medicine, Thrombotic and Haemorrhagic Disease Unit and Haemophilia Center, University of Padova, Padova, Italy
| | - Loris Bertazza
- Departiment of Medicine, Unit of Endocrinology, University of Padova, Padova, Italy
| | - Simone Mocellin
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
- Department of Surgical, Oncological and Gastroenterological Sciences (DISCOG), University of Padova, Padova, Italy
| | - Jacopo Pigozzo
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | | | | | - Alberto Ferlin
- Department of Medicine, Unit of Andrology and Reproductive Medicine, University of Padova, Padova, Italy
| |
Collapse
|
27
|
Coniac S, Costache-Outas MC, Antone-Iordache IL, Barbu AM, Bardan VT, Zamfir A, Ionescu AI, Badiu C. Real-World Evaluation of Immune-Related Endocrinopathies in Metastatic NSCLC Patients Treated with ICIs in Romania. Cancers (Basel) 2025; 17:1198. [PMID: 40227797 PMCID: PMC11987770 DOI: 10.3390/cancers17071198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025] Open
Abstract
(1) Background: Exploring real-world data (RWD) regarding immune-related adverse events (irAEs) is crucial to better understand the efficacy and safety of immunotherapy in cancer patient populations excluded from clinical trials. An analysis was conducted to evaluate the presumptive predictive causality between endocrine irAEs and the efficacy of immune check-point inhibitors (ICIs) in metastatic non-small-cell lung cancer (mNSCLC) patients treated in daily practice in Romania. (2) Methods: This was a retrospective cohort study of mNSCLC patients treated with ICIs in a tertiary level hospital in Romania for a period of almost seven years, from November 2017 till July 2024. Endocrine irAEs were well defined as any occurring autoimmune endocrinopathy during ICIs and related to immunotherapy. The hospital endocrinologist (M.C.C.O) diagnosed, treated, and followed these endocrine irAEs in a multidisciplinary approach. We investigated multiple medical variables to assess their impact on ICI effectiveness. Descriptive and statistical analyses were performed. (3) Results: Of 487 cancer patients treated with ICIs, we identified 215 mNSCLC patients who were evaluated for endocrine irAEs and co-medications during ICI therapy. Forty-seven (21.8%) patients experienced endocrine irAEs, thyroiditis being the most frequent and prevalent autoimmune endocrinopathy in 60% of cases. Endocrine irAEs were statistically significant, correlated with ICI efficacy (p = 0.002) for survival analysis. Steroids and proton-pump inhibitors used as co-medication during ICIs had a negative impact on response to therapy. (4) Conclusions: Endocrine irAEs might be considered predictive biomarkers for successful immunotherapy in mNSCLC patients. Co-medication during ICIs had a major influence on the effectiveness of these cutting-edge therapies. RWD plays an important role for oncology daily practice whenever clinical trial evidence is not available to guide decision.
Collapse
Affiliation(s)
- Simona Coniac
- Department of Medical Oncology, Hospice Hope Bucharest, 023642 Bucharest, Romania;
- Department of Endocrinology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-M.B.); (V.T.B.); (A.Z.); (C.B.)
| | | | | | - Ana-Maria Barbu
- Department of Endocrinology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-M.B.); (V.T.B.); (A.Z.); (C.B.)
| | - Victor Teodor Bardan
- Department of Endocrinology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-M.B.); (V.T.B.); (A.Z.); (C.B.)
| | - Andreea Zamfir
- Department of Endocrinology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-M.B.); (V.T.B.); (A.Z.); (C.B.)
| | - Andreea-Iuliana Ionescu
- Department of Endocrinology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-M.B.); (V.T.B.); (A.Z.); (C.B.)
- Department of Radiotherapy, Coltea Clinical Hospital, 030167 Bucharest, Romania;
- Department of Medical Oncology, Colțea Clinical Hospital, 030167 Bucharest, Romania
| | - Corin Badiu
- Department of Endocrinology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-M.B.); (V.T.B.); (A.Z.); (C.B.)
- C.I. Parhon National Institute of Endocrinology, 011863 Bucharest, Romania
| |
Collapse
|
28
|
Tang FH, Wong HYT, Tsang PSW, Yau M, Tam SY, Law L, Yau K, Wong J, Farah FHM, Wong J. Recent advancements in lung cancer research: a narrative review. Transl Lung Cancer Res 2025; 14:975-990. [PMID: 40248731 PMCID: PMC12000946 DOI: 10.21037/tlcr-24-979] [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: 10/21/2024] [Accepted: 01/27/2025] [Indexed: 04/19/2025]
Abstract
Background and Objective Lung cancer remains the leading cause of cancer-related mortality worldwide, with a 5-year survival rate ranging from 10% to 20%. The majority of cases are categorized as non-small cell lung cancer (NSCLC) (80%) and small cell lung cancer (SCLC) (20%), with NSCLC being the more prevalent type. Tobacco use, particularly cigarette smoking, is a significant contributor to over 80% of lung cancer cases. Early diagnosis is challenging due to limitations in screening methods, resulting in many cases being identified only in advanced stages. Moreover, current treatment options often exhibit low efficacy, partly due to an inadequate understanding of the disease's pathogenesis. This narrative review aims to summarize recent discoveries and advancements in lung cancer research, focusing on improvements in diagnosis, treatment, and understanding of the disease. Methods A comprehensive literature review was performed utilizing the PubMed Central database to identify recent studies relevant to lung cancer. This review synthesizes findings from various research articles to provide a cohesive summary of advancements in the field. Key Content and Findings In the past decade, notable progress has been achieved in lung cancer research, particularly concerning diagnostics and treatment strategies. Novel therapeutic approaches, including immunotherapy and genomic-targeted therapies, have demonstrated promising results. Understanding the tumor microenvironment (TME) and the role of T lymphocytes has become crucial for developing effective treatments. Additionally, advancements in immune checkpoint inhibitors (ICIs) have shown potential in enhancing patient outcomes. Improvements in tumor detection technologies are also anticipated to facilitate earlier diagnosis, ultimately contributing to better survival rates. Conclusions Significant strides have been made in lung cancer research over the last ten years, particularly in diagnostics and treatment methodologies. Future research should prioritize exploring the TME, the function of T lymphocytes, and the efficacy of ICIs while continuing to innovate in tumor detection technologies. Such efforts are essential for enhancing treatment outcomes and improving the overall quality of life for lung cancer patients.
Collapse
Affiliation(s)
- Fuk Hay Tang
- School of Medical and Health Sciences, Tung Wah College, Hong Kong, China
| | - Heylie Y. T. Wong
- School of Medical and Health Sciences, Tung Wah College, Hong Kong, China
| | | | - Mabel Yau
- School of Medical and Health Sciences, Tung Wah College, Hong Kong, China
| | - Shing Yau Tam
- School of Medical and Health Sciences, Tung Wah College, Hong Kong, China
| | - Lawla Law
- School of Medical and Health Sciences, Tung Wah College, Hong Kong, China
| | - Katherine Yau
- School of Nursing, Tung Wah College, Hong Kong, China
| | - Jade Wong
- Library, Tung Wah College, Hong Kong, China
| | | | - Jacky Wong
- School of Medical and Health Sciences, Tung Wah College, Hong Kong, China
| |
Collapse
|
29
|
Shalata W, Gothelf I, Dudnik Y, Cohen AY, Abu Jama A, Liba T, Dan O, Tourkey L, Shalata S, Agbarya A, Meirovitz A, Yakobson A. Correlation Between Body Mass Index and Immunotherapy Response in Advanced NSCLC. Cancers (Basel) 2025; 17:1149. [PMID: 40227730 PMCID: PMC11988004 DOI: 10.3390/cancers17071149] [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: 02/27/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have revolutionized the management of advanced non-small cell lung cancer (NSCLC). Emerging evidence suggests a potential association between elevated body mass index (BMI) and enhanced ICI efficacy, yet this relationship remains inconclusive and warrants further investigation. This study aims to evaluate the impact of BMI on treatment efficacy and survival outcomes in advanced NSCLC patients treated with first-line ICI therapy. METHODS A retrospective study was conducted at a multi-center registry to evaluate the impact of baseline BMI on overall survival (OS) and progression-free survival (PFS) in patients with stage IV NSCLC who received first-line ICI therapies. Treatment regimens included pembrolizumab or the combination of ipilimumab and nivolumab, administered either as monotherapy or in combination with chemotherapy, at the oncology department between January 2018 and December 2023. BMI was categorized according to the World Health Organization (WHO) classification, and OS and PFS were evaluated using Kaplan-Meier survival analysis and the Cox proportional hazards regression model. RESULTS Among 346 patients, 12.72% were underweight, 45.38% normal weight, 29.19% overweight, and 12.72% obese. Overweight and obese patients were more likely to receive pembrolizumab (p = 0.039) and less likely to undergo chemotherapy (p = 0.012). No significant differences in median overall survival (OS, log-rank: p = 0.155) or progression-free survival (PFS, log-rank: p = 0.370) were observed across BMI categories. However, differences emerged upon further analysis of PD-L1 levels (OS, log-rank: p = 0.029; PFS, log-rank: p = 0.044), additional chemotherapy (OS, log-rank: p = 0.009; PFS, log-rank: p = 0.021), type of immune checkpoint inhibitor (OS, log-rank: p < 0.001; PFS, log-rank: p < 0.001), and histologic diagnosis (OS, log-rank: p = 0.011; PFS, log-rank: p = 0.003). CONCLUSIONS BMI was not an independent predictor of survival outcomes in advanced NSCLC treated with ICI. Incorporating BMI with other patient-specific factors into personalized immunotherapy strategies highlights the importance of tailored approaches to improve patient care and clinical outcomes.
Collapse
Affiliation(s)
- Walid Shalata
- The Legacy Heritage Cancer Center, Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 8410501, Israel
- Medical School for International Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Itamar Gothelf
- Goldman Medical School, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Yulia Dudnik
- The Legacy Heritage Cancer Center, Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 8410501, Israel
- Medical School for International Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Ahron Yehonatan Cohen
- The Legacy Heritage Cancer Center, Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 8410501, Israel
- Medical School for International Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Ashraf Abu Jama
- The Legacy Heritage Cancer Center, Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 8410501, Israel
- Medical School for International Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Tom Liba
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 5290002, Israel
| | - Ofir Dan
- Goldman Medical School, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Lena Tourkey
- The Legacy Heritage Cancer Center, Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 8410501, Israel
| | - Sondos Shalata
- Nutrition Unit, Galilee Medical Center, Nahariya 2210006, Israel
| | - Abed Agbarya
- Oncology Department, Bnai Zion Medical Center, Haifa 3104701, Israel
| | - Amichay Meirovitz
- The Legacy Heritage Cancer Center, Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 8410501, Israel
- Medical School for International Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Alexander Yakobson
- The Legacy Heritage Cancer Center, Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 8410501, Israel
- Medical School for International Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| |
Collapse
|
30
|
Li Y, Cao L, Liu L, Ding Y, Cao F. Peripheral blood markers predict prognosis and irAEs of stage IV driver gene-negative lung adenocarcinoma treated with ICIs. Front Immunol 2025; 16:1538392. [PMID: 40226623 PMCID: PMC11985769 DOI: 10.3389/fimmu.2025.1538392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
Objective This study aims to evaluate the prognostic significance of peripheral blood biomarkers in relation to outcomes and immune-related adverse events (irAEs) among patients with stage IV driver gene-negative lung adenocarcinoma receiving treatment with immune checkpoint inhibitors (ICIs). Methods We conducted a retrospective analysis of clinicopathological data from 102 patients diagnosed with stage IV driver gene-negative lung adenocarcinoma who were treated with ICIs at the Fourth Hospital of Hebei Medical University between January 1, 2019, and December 31, 2023. We employed the Kaplan-Meier method to perform a univariate analysis of progression-free survival (PFS) and overall survival (OS), generated survival curves, and assessed differences in survival between groups using the log-rank test. The Cox regression model was utilized for multivariate analysis of PFS and OS. Additionally, we assessed the predictive value of peripheral blood markers for irAEs using logistic regression models. Results The 1-, 3-, and 5-year PFS rates for the cohort of 102 patients were recorded at 35.3%, 3.9%, and 0%, respectively; similarly, the OS rates at these time points were observed to be 66.7%, 8.8%, and 2.9%. Multivariate analysis identified that the prognostic nutritional index (PNI) and metastatic status served as independent prognostic factors influencing PFS outcomes in this patient population; furthermore, D-dimer levels, PNI, metastatic status, and the occurrence of irAEs emerged as independent prognostic indicators impacting OS rates among patients. Subsequent logistic regression analysis revealed that both platelet-to-lymphocyte ratio (PLR) and lymphocyte-to-monocyte ratio (LMR) functioned as independent predictors of irAEs with statistical significance (P=0.032; P=0.02). Conclusion For patients with stage IV driver gene-negative lung adenocarcinoma undergoing ICI therapy, PNI and metastatic status can serve as initial predictors of PFS. Additionally, D-dimer levels, PNI, metastatic status, and the presence of irAEs can initially predict OS, aiding in the identification of populations that may benefit from ICI therapy in clinical practice. Furthermore, our findings indicate a need for heightened attention to PLR and LMR concerning the occurrence of irAEs.
Collapse
Affiliation(s)
- Yu Li
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lei Cao
- Department of Geriatric Respiratory, Hebei General Hospital, Shijiazhuang, China
| | - Lei Liu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yawen Ding
- Clinical Laboratory, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Feng Cao
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
31
|
Yang Z, Luo Y, Lu R, Liu X, Liu H, Liu S, Huang C, Tian J, Zhang L. Incidence Rates of Cutaneous Immune-Related Adverse Events in Patients with Lung Cancer: A Systematic Review and Meta-Analysis. Curr Oncol 2025; 32:195. [PMID: 40277752 DOI: 10.3390/curroncol32040195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/26/2025] Open
Abstract
OBJECTIVE Cutaneous immune-related adverse events (cirAEs) represent a prevalent manifestation of adverse reactions linked to immune checkpoint inhibitors (ICIs) therapy, substantially affecting patients' quality of life. This systematic review and meta-analysis aimed to quantify the pooled incidence of cirAEs in this population and strengthen clinical awareness for early recognition and management. METHODS A comprehensive search of PubMed, Embase, CINAHL, Cochrane Library, CBM, CNKI, and Wanfang databases was conducted from inception to December 2022. Literature that reported the incidence of cirAEs in patients with lung cancer receiving ICIs therapy was included. A meta-analysis was conducted using R software, version 4.4.1 to estimate the pooled incidence of cirAEs, and a random-effects model was used for data synthesis. Begg's rank correlation and funnel plots were used to assess publication bias. RESULTS A total of 99 articles involving 23,814 patients with lung cancer receiving ICIs therapy were included, with publication dates ranging from 2012 to 2022. The meta-analysis results reveal that the incidence of cirAEs in patients with lung cancer was 20.26% (95% confidence interval [CI (17.12-23.81)]. Significant differences were observed between all subgroups, including continent, study type, combination therapy, dual ICIs therapy, and diagnostic criteria for cirAEs for Grade 1-2 and Grade 3-4 incidences. CONCLUSIONS The incidence of cirAEs in patients with lung cancer is relatively high, particularly undergoing combined or dual ICIs therapy. To comprehensively characterize cirAEs in patients with lung cancer, large-scale multicenter studies integrating real-world pharmacovigilance data are warranted to establish precise incidence estimates and identify clinically significant risk factors. IMPLICATIONS FOR CLINICAL PRACTICE This review's insights aroused clinical staff's attention and concern about cirAEs, potentially enhancing the quality of life of patients with cancer.
Collapse
Affiliation(s)
- Zhihui Yang
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China
- Evidence Based Nursing and Midwifery Practice PR China: A JBI Centre of Excellence, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China
| | - Yuanyuan Luo
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China
| | - Ruiqi Lu
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China
| | - Xinqi Liu
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China
| | - Hanyu Liu
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China
| | - Suting Liu
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China
| | - Chen Huang
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China
| | - Jinhui Tian
- Evidence Based Medicine Centre, School of Basic Medical Sciences, Lanzhou University, No. 199, Donggang West Road, Lanzhou 730000, China
| | - Lili Zhang
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China
- Evidence Based Nursing and Midwifery Practice PR China: A JBI Centre of Excellence, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China
| |
Collapse
|
32
|
Enoch F, Pons-Tostivint E. [Atezolizumab in first line in patients with metastatic non-small lung cancer who are platinum-doublet ineligible]. Bull Cancer 2025:S0007-4551(25)00120-1. [PMID: 40140317 DOI: 10.1016/j.bulcan.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 12/24/2024] [Accepted: 02/01/2025] [Indexed: 03/28/2025]
Affiliation(s)
- Faustine Enoch
- Département d'oncologie médicale, hôpital de la Timone, Assistance publique-Hôpitaux de Marseille, Marseille, France.
| | - Elvire Pons-Tostivint
- Service d'oncologie médical, centre hospitalier universitaire de Nantes, Nantes University, Nantes, France
| |
Collapse
|
33
|
Koyama J, Morise M, Tanaka I, Hori S, Matsuzawa R, Ozone S, Matsushita A, Matsuo M, Asano S, Tanaka T, Shima K, Kimura T, Sakamoto K, Kondoh Y, Hashimoto N. Clinical benefit of PD-1/PD-L1 inhibitors for poor performance status patients with advanced non-small cell lung cancer. J Chemother 2025:1-10. [PMID: 40129132 DOI: 10.1080/1120009x.2025.2481349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 02/14/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025]
Abstract
The benefit of programmed cell death protein-1 (PD-1)/programmed cell death protein ligand-1 (PD-L1) inhibitors remains unclear in non-small cell lung cancer (NSCLC) patients with poor performance status (PS). In the current multi-centre retrospective cohort study, advanced or recurrent NSCLC patients treated with PD-1/PD-L1 inhibitors were enrolled. Of the 219 patients enrolled, 44 had PS 2-4. The objective response rate (ORR) of patients with PS 2-4 in 1st line was 33%. Among 1st line group, median progression-free survival (PFS) in patients with PS 2 was significantly longer compared to that in patients with PS 3-4 (15.3 months vs. 0.9 months, P = 0.039, Log-rank test). Among previously treated patients, the ORR of patients with PS 2-4 was only 4%, and PFS and overall survival was poor even in patients with PS 2. PD-1/PD-L1 inhibitors can be an option for PS 2 NSCLC patients in 1st line setting.
Collapse
Affiliation(s)
- Junji Koyama
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiro Morise
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ichidai Tanaka
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sho Hori
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Reiko Matsuzawa
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sachiko Ozone
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Masaki Matsuo
- Department of Respiratory Medicine, Chubu Rosai Hospital, Nagoya, Japan
| | - Shuichi Asano
- Department of Respiratory Medicine, Chukyo Hospital, Nagoya, Japan
| | - Taro Tanaka
- Department of Respiratory Medicine, Nagoya Ekisaikai Hospital, Nagoya, Japan
| | - Koichiro Shima
- Department of Respiratory Medicine, Nagoya Ekisaikai Hospital, Nagoya, Japan
| | - Tomoki Kimura
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, Seto, Japan
| | - Koji Sakamoto
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Kondoh
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, Seto, Japan
| | - Naozumi Hashimoto
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Respiratory Medicine, Fujita Health University, Toyoake, Japan
| |
Collapse
|
34
|
Zhang T, Zhou H. Machine Learning and Weighted Gene Coexpression Network-Based Identification of Biomarkers Predicting Immune Profiling and Drug Resistance in Lung Adenocarcinoma. Int J Genomics 2025; 2025:9923294. [PMID: 40161493 PMCID: PMC11955064 DOI: 10.1155/ijog/9923294] [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: 11/28/2024] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Background: The prognosis for lung adenocarcinoma (LUAD) is poor, and the recurrence rate is high. Thus, to evaluate patients' prognoses and direct therapy choices, new prognostic markers are desperately needed. Methods: First, gene modules associated with LUAD were identified by weighted gene coexpression network analysis (WGCNA) analysis. The expression profiles obtained were intersected with the differential expressed genes taken between LUAD samples and paracancerous samples. Afterward, stepwise regression analysis and the LASSO were used to compress the genes further, and a risk model was created. Furthermore, a nomogram based on risk scores and clinical features was created to validate the model. After that, the distinctions between the pertinent biological processes and signaling pathways among the various subgroups were investigated. Additionally, drug sensitivity testing, immunotherapy, immune infiltration analysis, and enrichment analysis were carried out. Finally, the biological role of ANLN in LUAD was explored by qPCR, cell scratch assay, and transwell. Results: A total of 257 intersected genes were obtained by taking the intersection of the differential genes between 2866 LUAD samples and paraneoplastic samples with the module genes after we screened two particular modules that had the strongest link with LUAD by WGCNA. ANLN, CASS4, and NMUR1 were found to be distinctive genes for the development of risk models after the intersecting genes were screened to find 176 genes linked to the prognosis for LUAD. Based on risk assessments, high- and low-risk groups of LUAD patients were divided. Low-risk patients exhibited a significantly higher overall survival (OS) than those in the high-risk group. Expression of model genes correlates with infiltration of the vast majority of immune cells. Significant differences in the biological pathways, immune microenvironment, and abundance of immune cell infiltration were found between the two groups. The drug sensitivity study showed that patients in the high-risk group had higher IC50 values for BMS-754807_2171 and Doramapimod_10424. Finally, in vitro experiments demonstrated that knocking down ANLN noticeably inhibited the viability, migration, and invasion of A549 cells. Conclusion: This study may provide a theoretical reference for future exploration of potential diagnostic and prognostic biomarkers for LUAD.
Collapse
Affiliation(s)
- Tian Zhang
- Pharmacy Department, Xiangxi Autonomous Prefecture People's Hospital, Jishou, China
| | - Han Zhou
- Pharmacy Department, Xiangxi Autonomous Prefecture People's Hospital, Jishou, China
| |
Collapse
|
35
|
Liu Z, Yang Z, Wu J, Zhang W, Sun Y, Zhang C, Bai G, Yang L, Fan H, Chen Y, Zhang L, Jiang B, Liu X, Ma X, Tang W, Liu C, Qu Y, Yan L, Zhao D, Wu Y, He S, Xu L, Peng L, Chen X, Zhou B, Zhao L, Zhao Z, Tan F, Zhang W, Yi D, Li X, Gao Q, Zhang G, Wang Y, Yang M, Fu H, Guo Y, Hu X, Cai Q, Qi L, Bo Y, Peng H, Tian Z, She Y, Zou C, Zhu L, Cheng S, Zhang Y, Zhong W, Chen C, Gao S, Zhang Z. A single-cell atlas reveals immune heterogeneity in anti-PD-1-treated non-small cell lung cancer. Cell 2025:S0092-8674(25)00291-0. [PMID: 40147443 DOI: 10.1016/j.cell.2025.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/20/2024] [Accepted: 03/09/2025] [Indexed: 03/29/2025]
Abstract
Anti-PD-(L)1 treatment is standard for non-small cell lung cancer (NSCLC), but patients show variable responses to the same regimen. The tumor immune microenvironment (TIME) is associated with immunotherapy response, yet the heterogeneous underlying therapeutic outcomes remain underexplored. We applied single-cell RNA and TCR sequencing (scRNA/TCR-seq) to analyze surgical tumor samples from 234 NSCLC patients post-neoadjuvant chemo-immunotherapy. Analyses revealed five distinct TIME subtypes with varying major pathological response (MPR) rates. MPR patients had elevated levels of FGFBP2+ NK/NK-like T cells, memory B cells, or effector T cells, while non-MPR patients showed higher CCR8+ Tregs. T cell clonal expansion analyses unveiled heterogeneity in non-MPR patients, marked by varying expansions of Tex-relevant cells and CCR8+ Tregs. Precursor exhausted T cells (Texp cells) correlated with recurrence-free survival, identifying a patient subgroup with reduced recurrence risk despite lack of MPR. Our study dissects TIME heterogeneity in response to chemoimmunotherapy, offering insights for NSCLC management.
Collapse
Affiliation(s)
- Zedao Liu
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Zhenlin Yang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Junqi Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Wenjie Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Yuxuan Sun
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Guangyu Bai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Peking University School of Oncology, Beijing, China
| | - Li Yang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hongtao Fan
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Yawen Chen
- National Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Lei Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Benyuan Jiang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Xiaoyan Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xiaoshi Ma
- Department of Urology, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University), Shenzhen 518020, China
| | - Wei Tang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Chang Liu
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Yang Qu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lixu Yan
- Department of Pathology, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Deping Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Yilong Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Shun He
- Department of Endoscopy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Long Xu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Lishan Peng
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Xiaowei Chen
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Bolun Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Liang Zhao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhangyi Zhao
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wanting Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Dingcheng Yi
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | | | - Qianqian Gao
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Guangjian Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yongjie Wang
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Minglei Yang
- Department of Thoracic Surgery, Ningbo No.2 Hospital, Ningbo 315010, China
| | - Honghao Fu
- Department of General Thoracic Surgery, Jining First People's Hospital, Affiliated Hospital of Shandong First Medical University, Jining 272000, China
| | - Yongjun Guo
- Department of Molecular Pathology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xueda Hu
- Analytical Biosciences Limited, Beijing, China
| | - Qingyuan Cai
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Lu Qi
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China; Changping Laboratory, Beijing 102206, China
| | - Yufei Bo
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Hui Peng
- National Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Zhigang Tian
- National Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China.
| | - Yunlang She
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China.
| | - Chang Zou
- School of Medicine, Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China; Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University), Shenzhen 518020, China.
| | - Linnan Zhu
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China.
| | - Sijin Cheng
- Changping Laboratory, Beijing 102206, China; Chongqing Medical University, Chongqing, China.
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Zhongyuan Cell Therapy and Immunotherapy Laboratory, Zhengzhou 450000, China.
| | - Wenzhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China.
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China; Chongqing Medical University, Chongqing, China.
| |
Collapse
|
36
|
Hashinokuchi A, Kinoshita F, Iimori M, Kosai K, Ono Y, Tomonaga T, Giacomo B, Matsudo K, Nagano T, Akamine T, Kohno M, Takenaka T, Oda Y, Yoshizumi T. DNA Polymerase Delta 2 Activates Cell Cycle in Lung Adenocarcinoma, Leading to High Malignancy and Poor Prognosis. Ann Surg Oncol 2025:10.1245/s10434-025-17118-x. [PMID: 40095310 DOI: 10.1245/s10434-025-17118-x] [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: 11/05/2024] [Accepted: 02/18/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND DNA polymerase delta 2 (POLD2) plays a crucial role in DNA repair and replication. POLD2 is upregulated and related to poor prognosis in several types of cancer. However, the biological and clinical importance of POLD2 in lung adenocarcinoma remains unclear. PATIENTS AND METHODS We investigated the relationship between POLD2 expression and tumor malignancy in lung adenocarcinoma cell lines. Immunohistochemistry (IHC) was performed on 373 patients with completely resected lung adenocarcinoma, and the association between POLD2 expression, clinicopathological features, and prognosis was examined. RESULTS POLD2 knockdown decreases cell proliferation and migration, resulting in apoptosis and G1 arrest in the cell cycle in lung adenocarcinoma cells. Additionally, POLD2 knockdown attenuates MYC expression, which may decrease the expression of cyclin-dependent kinases 4 and 6, pRb, Rb, and E2F1. Furthermore, among 373 patients with completely resected lung adenocarcinoma, smoking history, advanced pathological stage, and vascular invasion were significantly more prevalent in patients with high POLD2 expression (n = 146, 39.3%) than in those with low POLD2 expression (n = 227, 60.7%). Patients with high POLD2 expression also had a significantly worse recurrence-free survival (RFS) and overall survival. In the multivariable analysis, high POLD2 expression was an independent poor prognostic factor of RFS. CONCLUSIONS We provide the possibility of POLD2 as a potential new therapeutic target because high POLD2 expression is associated with high malignancy and poor prognosis in specimens from patients with lung adenocarcinoma and POLD2 depletion triggers the suppression of cell migration, cell cycle progression, and cell proliferation.
Collapse
Affiliation(s)
- Asato Hashinokuchi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Fumihiko Kinoshita
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Makoto Iimori
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Keisuke Kosai
- Department of Thoracic Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Yuya Ono
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Oita, Japan
| | - Takumi Tomonaga
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Bassi Giacomo
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kyoto Matsudo
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Taichi Nagano
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takaki Akamine
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mikihiro Kohno
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoyoshi Takenaka
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
37
|
Wang M, Yang J, Wang S, Gill H, Cheng H. Immunotherapy and the Tumor Microenvironment in Brain Metastases from Non-Small Cell Lung Cancer: Challenges and Future Directions. Curr Oncol 2025; 32:171. [PMID: 40136375 PMCID: PMC11941645 DOI: 10.3390/curroncol32030171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/09/2025] [Accepted: 03/15/2025] [Indexed: 03/27/2025] Open
Abstract
Brain metastases (BMs) are a relatively common and severe complication in advanced non-small cell lung cancer (NSCLC), significantly affecting patient prognosis. Metastatic tumor cells can alter the brain tumor microenvironment (TME) to promote an immunosuppressive state, characterized by reduced infiltration of tumor-infiltrating lymphocytes (TILs), diminished expression of programmed death-ligand 1 (PD-L1), and changes in other proinflammatory factors and immune cell populations. Microglia, the resident macrophages of the brain, play a pivotal role in modulating the central nervous system (CNS) microenvironment through interactions with metastatic cancer cells, astrocytes, and infiltrating T cells. The M2 phenotype of microglia contributes to immunosuppression in BM via the activation of signaling pathways such as STAT3 and PI3K-AKT-mTOR. Recent advances have enhanced our understanding of the immune landscape of BMs in NSCLC, particularly regarding immune evasion within the CNS. Current immunotherapeutic strategies, including immune checkpoint inhibitors, have shown promise for NSCLC patients with BM, demonstrating intracranial activity and manageable safety profiles. Future research is warranted to further explore the molecular and immune mechanisms underlying BM, aiming to develop more effective treatments.
Collapse
Affiliation(s)
- Meng Wang
- Department of Oncology (Medical Oncology), Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.W.)
| | - Jihua Yang
- Department of Oncology (Medical Oncology), Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.W.)
| | - Shuai Wang
- Department of Oncology (Medical Oncology), Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.W.)
| | - Harjot Gill
- Department of Pathology, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Haiying Cheng
- Department of Oncology (Medical Oncology), Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.W.)
| |
Collapse
|
38
|
Gao W, Wang L, Zhao Y, Zhu L. The role of PD-L1 in EGFR-mutant non-small cell lung cancer. Discov Oncol 2025; 16:307. [PMID: 40072720 PMCID: PMC11904073 DOI: 10.1007/s12672-025-02089-y] [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: 09/20/2024] [Accepted: 03/06/2025] [Indexed: 03/14/2025] Open
Abstract
Lung cancer remains the leading cause of cancer-related deaths globally. In China, nearly half of non-small cell lung cancer (NSCLC) patients carry epidermal growth factor receptor (EGFR) mutations. EGFR tyrosine kinase inhibitors (EGFR-TKIs) have significantly improved the prognosis for patients with EGFR mutations and are considered the preferred treatment for these individuals. Programmed death-ligand 1 (PD-L1) expression levels are now widely used as a biomarker to predict the effectiveness of PD-1/PD-L1 immunotherapy in NSCLC. Additionally, the impact of PD-L1 expression on the efficacy of EGFR-TKIs has garnered considerable attention from researchers. This review explores recent studies on the epidemiology and clinical outcomes associated with PD-L1 in NSCLC patients harboring driver gene mutations.
Collapse
Affiliation(s)
- Wentao Gao
- Hangzhou First People's Hospital, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Lingling Wang
- Hangzhou First People's Hospital, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Yanyan Zhao
- Hangzhou First People's Hospital, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Lucheng Zhu
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Zhejiang Chinese Medical University, No. 34, Yanguan Lane, Hangzhou, 310002, People's Republic of China.
| |
Collapse
|
39
|
Ding J, Jiang Y, Jiang N, Xing S, Ge F, Ma P, Tang Q, Miao H, Zhou J, Fang Y, Cui D, Liu D, Han Y, Yu W, Wang Y, Zhao G, Cai Y, Wang S, Sun N, Li N. Bridging the gap: unlocking the potential of emerging drug therapies for brain metastasis. Brain 2025; 148:702-722. [PMID: 39512184 DOI: 10.1093/brain/awae366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/30/2024] [Accepted: 09/29/2024] [Indexed: 11/15/2024] Open
Abstract
Brain metastasis remains an unmet clinical need in advanced cancers with an increasing incidence and poor prognosis. The limited response to various treatments is mainly derived from the presence of the substantive barrier, blood-brain barrier (BBB) and brain-tumour barrier (BTB), which hinders the access of potentially effective therapeutics to the metastatic tumour of the brain. Recently, the understanding of the structural and molecular features of the BBB/BTB has led to the development of efficient strategies to enhance BBB/BTB permeability and deliver drugs across the BBB/BTB to elicit the anti-tumour response against brain metastasis. Meanwhile, novel agents capable of penetrating the BBB have rapidly developed and been evaluated in preclinical studies and clinical trials, with both targeted therapies and immunotherapies demonstrating impressive intracranial activity against brain metastasis. In this review, we summarize the recent advances in the biological properties of the BBB/BTB and the emerging strategies for BBB/BTB permeabilization and drug delivery across the BBB/BTB. We also discuss the emerging targeted therapies and immunotherapies against brain metastasis tested in clinical trials. Additionally, we provide our viewpoints on accelerating clinical translation of novel drugs into clinic for patients of brain metastasis. Although still challenging, we expect this review to benefit the future development of novel therapeutics, specifically from a clinical perspective.
Collapse
Affiliation(s)
- Jiatong Ding
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yale Jiang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ning Jiang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shujun Xing
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Fan Ge
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- 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
| | - Peiwen Ma
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qiyu Tang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Huilei Miao
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jiawei Zhou
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yuan Fang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Dandan Cui
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Dongyan Liu
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yanjie Han
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Weijie Yu
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yuning Wang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Guo Zhao
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yuanting Cai
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shuhang Wang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- 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
| | - Ning Li
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
40
|
Li X, Xu H, Hong R, Yang H, Xu L, Zheng G, Xie B. Frontline pemetrexed and cisplatin based-chemotherapy combined with NRT promoted the antitumor in a mouse model of lung carcinoma. Int Immunopharmacol 2025; 149:114174. [PMID: 39929101 DOI: 10.1016/j.intimp.2025.114174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/22/2025]
Abstract
The efficacy of neoantigen-reactive T cells (NRT) therapy in solid tumors, encompassing aspects such as infiltration, recognition, cytotoxicity, and enduring persistence, is notably influenced by the immunological microenvironment. This study endeavors to investigate whether the co-administration of pemetrexed and cisplatin augments the therapeutic efficacy of NRT therapy in lung cancer. Neoantigens were predicted using a comprehensive analysis of mutation data from Lewis lung carcinoma cells and mouse tail tissues. The immunogenicity of NRT cells was assessed through flow cytometry and IFN-γ ELISpot assays. A mouse model of NSCLC was used to investigate the anti-tumor effects of NRT combined with chemotherapy. The combination of NRT cells and chemotherapy significantly inhibited tumor growth in a mouse model, increased CD3+/CD137+ T cells to promote IFN-γ secretion from NRT cells, and up-regulated the levels of inflammatory cytokine proteins including IFN-γ, TNF, IL-6 and IL-10. Immunofluorescence analysis confirmed increased T-cell infiltration in tumor tissues without adverse effects on vital organs. In addition, transcriptome analyses indicated that the tumor microenvironment was altered to favor M1-like macrophages with an increased M1/M2 ratio, creating a pro-inflammatory environment. The integration of NRT with frontline chemotherapy for lung cancer could yield profoundly ideal therapeutic outcomes.
Collapse
Affiliation(s)
- Xiaoqin Li
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Hang Xu
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Rujun Hong
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Haitao Yang
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Lihuan Xu
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China
| | - Guanying Zheng
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China.
| | - Baosong Xie
- Department of Respiratory Medicine and Critical Care Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou Fujian China.
| |
Collapse
|
41
|
Li Z, Zhu N, Liu Y, Yu Y, Wang T, Zou C, Wang S, Ou X. A disproportionality analysis of real-world events from the FDA Adverse Event Reporting System (FAERS) for Atezolizumab. BMC Pharmacol Toxicol 2025; 26:51. [PMID: 40038564 DOI: 10.1186/s40360-025-00879-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 02/21/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND An increasing number of clinical studies have highlighted the use of atezolizumab in tumor immunotherapy. However, There is still a lack of comprehensive research on its associated adverse events (AEs). To improve our understanding of its toxicological profile and to provide valuable clinical insights regarding into the effectiveness of immunotherapy, this study utilized data from the US Food and Drug Administration Adverse Event Reporting System (FAERS) to conduct a retrospective analysis of AEs linked to atezolizumab. METHODS We extracted the reports of AEs related to atezolizumab from the FAERS database from the first quarter of 2004 to the first quarter of 2024. We quantified them using the reporting odds ratio (ROR) and proportional reporting ratio (PRR), along with chi-square value (χ²), and conducted systematic classification of the AE signal mining results through SAS 9.4 software. RESULTS A total of 19,563 valid reports were incorporated, involving 20 distinct system organ class categories. The AEs related to atezolizumab, reported at the preferred term level, mainly encompassed anemia [ROR 2.33, 95% confidence interval (CI) lower limit 2.09, PRR 2.31, χ² 255.977], febrile neutropenia (ROR 2.81, 95% CI lower limit 2.50, PRR 2.79, χ² 333.586), neutrophil count decreased (ROR 2.14, 95% CI lower limit 1.89, PRR 2.13, χ² 150.688), white blood cell count decreased (ROR 2.35, 95% CI lower limit 2.03, PRR 2.34, χ² 136.673), sepsis (ROR 2.21, 95% CI lower limit 1.91, PRR 2.20, χ² 117.741), alanine aminotransferase increased (ALT) (ROR 2.86, 95% CI lower limit 2.44, PRR 2.85, χ² 180.031), and aspartate aminotransferase increased (AST) (ROR 2.79, 95% CI lower limit 2.38, PRR 2.78, χ² 170.955). CONCLUSIONS Apart from various degrees of hepatotoxicity, such as increased ALT and AST, the immune-related hematological toxicity of atezolizumab should also be noted. In clinical practice, healthcare providers should always be vigilant for the occurrence of such medication-related AEs and take measures to enhance the safety of clinical medication use.
Collapse
Affiliation(s)
- Zhuoyang Li
- School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Ning Zhu
- Division of Head & Neck Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuwei Liu
- School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Yan Yu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Tianhong Wang
- The Department of Clinical Research, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Congcong Zou
- Anesthesia and Surgery Center of West China Xiamen Hospital, Sichuan University, 699 Jinyuan West Road, Xingbin Street, Jimei District, Xiamen, Fujian Province, China
| | - Siman Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaofeng Ou
- Department of Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
42
|
Xiao F, Liu Y, Wang X. The Efficacy of Immune Checkpoint Inhibitors in the EGFR Mutant and Wild-Type Non-Small Cell Lung Cancer Is Positively Associated With the Maturation and Abundance of Dendritic Cells. Thorac Cancer 2025; 16:e70049. [PMID: 40130724 PMCID: PMC11934210 DOI: 10.1111/1759-7714.70049] [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: 01/17/2025] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Dendritic cells (DCs) are known to be crucial in initiating immune responses, but their role in regulating immune checkpoint inhibitor (ICI) efficacy in EGFR mutant NSCLC remains unclear. METHODS Peripheral blood mononuclear cells (PBMCs) were co-cultured with EGFR mutant cells to evaluate immune scores and DC maturation via high-throughput sequencing. TIDE scores were used to predict the efficacy of ICI treatment. Gene set enrichment analysis (GSEA) was carried out on DCs to explore the signaling pathway changes underlying the diverse responses to ICIs. RESULTS A significant decrease in CD8+ T lymphocytes and cytotoxicity scores was found in EGFR mutant LUAD compared to wild-type (p < 0.001). Three datasets (GSE135222, GSE126044, and GSE136961) showed that higher DC gene expression was associated with a more favorable response to ICIs (p = 0.028). The CSE241934 dataset showed that the number of conventional DC 1 (cDC1) was higher in the ICI-sensitive group. The TIDE model suggested that cDC1 was associated with ICIs efficacy. However, GSE32863, GSE75037, and GSE72094 showed no differences in cDC subpopulations between EGFR mutant and wild-type LUAD. EGFR mutant cells exhibited more suppression in the expression of HLA-DR, CD40, CD83, and CD86 than the control group. The TIDE model suggested DC maturity was associated with ICI efficacy. GSE241934-IIT showed that DC maturity was more abundant in the ICI-sensitive group than that in the resistant group. CONCLUSIONS Both the number and maturation capacity of DCs are positively correlated with ICI efficacy. The cause of poor ICI efficacy in EGFR mutant LUAD is more likely to be low DC maturity, not number, compared to EGFR wild-type LUAD.
Collapse
Affiliation(s)
- Fengqi Xiao
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanShandongChina
| | - Yanguo Liu
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanShandongChina
| | - Xiuwen Wang
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanShandongChina
| |
Collapse
|
43
|
Vinjamuri S, Pant V. Demystifying the Role of Immuno PET-CT in Non-Small Cell Lung Cancer: Clinical Value and Research Trends. Semin Nucl Med 2025; 55:212-220. [PMID: 40016063 DOI: 10.1053/j.semnuclmed.2025.02.004] [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: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 03/01/2025]
Abstract
The management of Lung cancer, especially non-small cell lung cancer has undergone a paradigm shift recently with the advent of new treatment approaches including focused radiotherapy as well as evolution of a newer class of immunotherapy agents. Treatment efficacy and survival rates have improved and it is now even more important that patients are selected for appropriate interventions on the basis of a comprehensive assessment including a range of imaging as well as in-vitro tests such as immunohistochemistry. A new class of tracers targeting programmed cell death such as PD1 and PDL1 (broadly classed as Immuno PET) are being increasingly used in the molecular characterisation of patients deemed resistant to standard treatment approaches and being considered for additional interventions such as immunotherapy. In this review, we review the latest evidence in the field and propose a summary of clinical usefulness and provide a review of the research trends in this exciting and evolving field.
Collapse
Affiliation(s)
- Sobhan Vinjamuri
- Department of Nuclear Medicine, Royal Liverpool University Hospital, Liverpool, UK.
| | - Vineet Pant
- Department of Nuclear Medicine, Royal Liverpool University Hospital, Liverpool, UK
| |
Collapse
|
44
|
Bréart B, Williams K, Krimm S, Wong T, Kayser BD, Wang L, Cheng E, Cruz Tleugabulova M, Bouziat R, Lu T, Yuen K, Firmino NS, Bravo DD, Roels J, Bhakta A, Bevers J, Lehoux I, Gutierrez A, Chestnut Y, Klementowicz JE, Arenzana TL, Akhmetzyanova I, Dixon E, Chen M, Tasneem K, Yadav R, Koeppen H, Oh SA, Delamarre L, Huang H, Lim SA, Nakamura G, Wang J, Gao C, Corpuz R, Müller S, West NR. IL-27 elicits a cytotoxic CD8 + T cell program to enforce tumour control. Nature 2025; 639:746-753. [PMID: 39910298 DOI: 10.1038/s41586-024-08510-w] [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: 10/04/2023] [Accepted: 12/10/2024] [Indexed: 02/07/2025]
Abstract
Although cytotoxic CD8+ T lymphocytes (CTLs) are essential for anti-tumour immunity, they are frequently dysfunctional in tumours1. Cytokines that sustain CTL activity are attractive for cancer immunotherapy, but avoiding inflammatory toxicity remains a challenge for their clinical use2. Here we show that expression of a CTL signature is strongly associated with IL27 expression in human and mouse tumours. In mice, IL-27 acts directly on tumour-specific CTLs to promote their persistence and effector function in the tumour microenvironment. Moreover, treatment with inducible IL-27 overexpression or a half-life-extended IL-27 protein in vivo is well tolerated, induces regression of established tumours, drives an enhanced cytotoxic program in anti-tumour CTLs and synergizes with PD-L1 blockade. In patients with cancer who were treated with anti-PD-1/PD-L1 therapy, high expression of IL-27 correlates with a favourable clinical response, and IL-27 supports human CTL function during chronic antigen stimulation ex vivo. Our data demonstrate that endogenous IL-27 is essential for anti-tumour immunity and that IL-27 receptor agonism can safely improve anti-tumour T cell responses alone or in combination with PD-L1 blockade.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Kobe Yuen
- Genentech, South San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | - Min Chen
- Genentech, South San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | | - Chan Gao
- Genentech, South San Francisco, CA, USA
| | | | | | | |
Collapse
|
45
|
Gille R, Pérol M. [First line treatment of non-oncogene-addicted metastatic non-small cell lung cancer]. Bull Cancer 2025; 112:3S64-3S74. [PMID: 40155079 DOI: 10.1016/s0007-4551(25)00159-6] [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] [Indexed: 04/01/2025]
Abstract
Since 2017, anti-PD-(L)1 immunotherapy has been the cornerstone of first-line treatment for stage IV non-oncogene-addicted non-small cell lung cancer. Its phase I development has established that the level of PD-L1 expression by tumor cells is predictive of response rate and progression-free survival. Above 50%, it makes chemotherapy not mandatory, with a median survival for pembrolizumab monotherapy of around 26 months and five-year survival of 32%. Large phase III studies have also validated the combination of anti-PD-(L)1 immunotherapy and platinum-based chemotherapy regardless of PD-L1 level of expression, increasing five-year survival from 10% to 18%. Dual immunotherapy combining anti-CTLA-4 and anti-PD-(L)1 might be interesting, especially in PD-L1 negative tumors, but is not available in France. Treatment personalization, particularly in the case of PD-L 1 expression >50%, should be based on response and non-response factors to immunotherapy, including patient-related factors such as performans status, age, smoking status, as well as tumor-related factors such as disease aggressiveness, tumor volume, mutational profile, along with concomitant medications. The optimal duration of immunotherapy is uncertain and arbitrarily set at two years. Many options are currently being explored to improve first-line treatment outcomes, as the majority of patients experience resistance to immunotherapy.
Collapse
Affiliation(s)
- Romane Gille
- Centre Léon-Bérard, 28 rue Laennec, 69008 Lyon, France.
| | - Maurice Pérol
- Centre Léon-Bérard, 28 rue Laennec, 69008 Lyon, France
| |
Collapse
|
46
|
Wang Y, Safi M, Hirsch FR, Lu S, Peters S, Govindan R, Rosell R, Park K, Zhang JJ. Immunotherapy for advanced-stage squamous cell lung cancer: the state of the art and outstanding questions. Nat Rev Clin Oncol 2025; 22:200-214. [PMID: 39762577 DOI: 10.1038/s41571-024-00979-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2024] [Indexed: 02/26/2025]
Abstract
Immune-checkpoint inhibitors (ICIs) have transformed the treatment paradigm for advanced-stage squamous non-small-cell lung cancer (LUSC), a histological subtype associated with inferior outcomes compared with lung adenocarcinoma. However, only a subset of patients derive durable clinical benefit. In the first-line setting, multiple ICI regimens are available, including anti-PD-(L)1 antibodies as monotherapy, in combination with chemotherapy, or with an anti-CTLA4 antibody with or without chemotherapy. Several important questions persist regarding the optimal regimen for individual patients, particularly how to identify patients who might benefit from adding chemotherapy and/or anti-CTLA4 antibodies to anti-PD-(L)1 antibodies. An urgent need exists for predictive biomarkers beyond PD-L1 to better guide precision oncology approaches. Deeper knowledge of the underlying molecular biology of LUSC and its implications for response to ICIs will be important in this regard. Integration of this knowledge into multi-omics methods coupled with artificial intelligence might enable the development of more robust biomarkers. Finally, several novel therapeutic strategies, including novel ICIs, bispecific antibodies and personalized cancer vaccines, are emerging. Addressing these unresolved questions through innovative clinical trials and translational research will be crucial to further improving the outcomes of patients with LUSC. In this Review, we provide a comprehensive overview of current immunotherapeutic approaches, unresolved challenges and emerging strategies for patients with LUSC.
Collapse
Affiliation(s)
- Yibei Wang
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mohammed Safi
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Fred R Hirsch
- Center for Thoracic Oncology, Tisch Cancer Institute and Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Shun Lu
- Department of Medical Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Solange Peters
- Oncology Department, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | | | - Rafael Rosell
- Dr. Rosell Oncology Institute, Dexeus University Hospital, Barcelona, Spain
| | - Keunchil Park
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
- Division of Hematology/Oncology, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Jianjun J Zhang
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genomic Medicine, the University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
47
|
Chen J, Ma N, Chen B, Huang Y, Li J, Li J, Chen Z, Wang P, Ran B, Yang J, Bai J, Ning S, Ai J, Wei Q, Liu L, Cao D. Synergistic effects of immunotherapy and adjunctive therapies in prostate cancer management. Crit Rev Oncol Hematol 2025; 207:104604. [PMID: 39732304 DOI: 10.1016/j.critrevonc.2024.104604] [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: 11/15/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 12/30/2024] Open
Abstract
In recent years, cancer immunotherapy has received widespread attention due to significant tumor clearance in some malignancies. Various immunotherapy approaches, including vaccines, immune checkpoint inhibitors, oncolytic virotherapy, bispecific T cell engagers, and adoptive T cell transfer, have completed or are undergoing clinical trials for prostate cancer. Despite immune checkpoint blockade's extraordinary effectiveness in treating a variety of cancers, targeted prostate cancer treatment using the immune system is still in its infancy. Multiple factors including the heterogeneity of prostate cancer, the cold tumor microenvironment, and a low level of neoantigens, contribute to the poor immunotherapy response. Significant effort is being devoted to improving immune-based prostate cancer therapy. Recently, several key discoveries demonstrate that prostate cancer immunotherapy agents may be used to promise better prognosis for patients as part of combination strategies with other agents targeting tumor-associated immune mechanism of resistance. Here, this review comprehensively examines the recent advancements in immunotherapy for prostate cancer, exploring its potential synergistic effects when combined with other treatment modalities to enhance clinical efficacy.
Collapse
Affiliation(s)
- Jie Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Ma
- Department of Pediatrics, West China Second University Hospital, Sichuan University, No. 20, 3rd section, South Renmin Road, Chengdu 610041, China
| | - Bo Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yin Huang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jinze Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zeyu Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Puze Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Biao Ran
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiahao Yang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingxing Bai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shu Ning
- Department of Urologic Surgery, University of California Davis, Davis, CA, USA
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liangren Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Dehong Cao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
48
|
Mitrakas AG, Kakouratos C, Lamprou I, Xanthopoulou E, Koukourakis MI. Oncogenic Mutations and the Tumor Microenvironment: Drivers of Non-Small Cell Lung Cancer Progression. Cancers (Basel) 2025; 17:853. [PMID: 40075700 PMCID: PMC11899603 DOI: 10.3390/cancers17050853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Non-small cell lung cancer (NSCLC) is a major cause of cancer-related deaths globally. The study focuses on understanding the interplay between genetic mutations, cancer stem cells (CSCs), and the tumor microenvironment (TME) in driving NSCLC progression, resistance to therapies, and relapse. METHODS A systematic search was conducted in PubMed and Scopus databases to identify significant and valuable studies relevant to NSCLC, focusing on genetic mutations, CSCs, and the TME. Articles were selected based on their relevance, methodological severity, date of publication, and scientific soundness related to NSCLC biology and therapeutic strategies. This review synthesized findings from these sources to highlight key mechanisms and potential therapeutic interventions. RESULTS Mutations in critical genes in KRAS, EGFR, TP53, and other key genes interfere with stem cell regulation, promoting CSC-like behavior, resistance to therapy, and immune evasion. The tumor microenvironment (TME), including immune cells, fibroblasts, and extracellular matrix components, further supports tumor growth and reduction in treatment efficacy. Promising strategies, including CSC targeting, TME modulation, and the development of novel biomarkers, have shown potential in preclinical and clinical studies. CONCLUSIONS The association between genetic alterations, CSCs, the TME, and other cellular pathways-including cell metabolism and immune evasion-plays a crucial role in therapy resistance, highlighting the need for comprehensive treatment strategies. The combination of genomic profiling with TME-targeting therapies could lead to personalized treatment approaches, offering hope for better clinical outcomes and reduced mortality in NSCLC patients.
Collapse
Affiliation(s)
- Achilleas G. Mitrakas
- Department of Radiotherapy/Oncology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (C.K.); (I.L.); (E.X.)
| | | | | | | | - Michael I. Koukourakis
- Department of Radiotherapy/Oncology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (C.K.); (I.L.); (E.X.)
| |
Collapse
|
49
|
Yao J, Li S, Bai L, Chen J, Ren C, Liu T, Qiu J, Dang J. Efficacy and safety of immune checkpoint inhibitors in elderly patients with advanced non-small cell lung cancer: a systematic review and meta-analysis. EClinicalMedicine 2025; 81:103081. [PMID: 39975700 PMCID: PMC11836518 DOI: 10.1016/j.eclinm.2025.103081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/06/2025] [Accepted: 01/14/2025] [Indexed: 02/21/2025] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) are the preferred treatments for advanced non-small cell lung cancer (NSCLC) without targetable oncogene alterations. However, evidence in the elderly population (aged ≥ 65 years) remains limited. Methods We searched PubMed, Embase, Cochrane Library, Web of Science, and Scopus databases for eligible publications until September 30, 2024. The primary outcome of interest was overall survival (OS). A random-effects model was used for the statistical analysis. Findings A total of 35 phase 3 randomized controlled trials (RCTs) involving 9788 patients and 64 real-world studies involving 37,111 patients were included. Results from phase 3 RCTs revealed that ICIs significantly improved OS (hazard ratio [HR] = 0.78, 95% confidence interval [CI]: 0.74-0.82) and progression-free survival (PFS) (HR = 0.67, 95% CI: 0.60-0.75) compared to chemotherapy. The association between ICIs and improved OS was independent of patient characteristics (race and histological type) or treatment-related factors (ICI drug type, treatment mode, and treatment line). However, significantly prolonged OS was not observed in subgroups of aged ≥ 75 years and PD-L1 < 1%. In real-world studies, the pooled median OS of ICIs were 11.8 months (95% CI: 11.2-12.4); Eastern Cooperative Oncology Group (EOCG) score, histological type, PD-L1 status, with immune-related adverse events (irAEs), and treatment mode were predictive for OS; rates of irAEs and discontinuation were numerically higher for combination therapy vs. monotherapy. Interpretation ICIs are associated with a significant improvement in OS and PFS compared to chemotherapy in elderly patients with advanced NSCLC. Nevertheless, some patient characteristics such as aged ≥ 75 years, ECOG score ≥ 2, and PD-L1 < 1% seem to have a negative impact on the efficacy of ICIs, while these findings require further validation in large RCTs. Funding None.
Collapse
Affiliation(s)
- Jiacheng Yao
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Sihan Li
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Lu Bai
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Jun Chen
- Department of Radiation Oncology, Shenyang Tenth People’s Hospital, Shenyang, China
| | - Chengbo Ren
- Department of Radiation Oncology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Tingting Liu
- Department of Radiation Oncology, Anshan Cancer Hospital, Anshan, China
| | - Jingping Qiu
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Jun Dang
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
50
|
Zhu P, Li Z, Sun Y, Liu T, Yin R. Persist or resist: Immune checkpoint inhibitors in EGFR-mutated NSCLC. Cancer Sci 2025; 116:581-591. [PMID: 39673162 PMCID: PMC11875763 DOI: 10.1111/cas.16428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/16/2024] Open
Abstract
Although epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs), especially third-generation TKIs, have significantly improved the progression-free survival and overall survival of non-small cell lung cancer (NSCLC) patients with EGFR mutation, TKI resistance is inevitable for most patients. Over the past few years, immune checkpoint inhibitors (ICIs) have significantly improved the survival for EGFR-wild type NSCLC patients. However, no significantly improved benefits were observed with ICI monotherapy in EGFR-mutated patients. EGFR-mutated NSCLC shows more heterogeneity in tumor mutational burden (TMB), programmed cell death-ligand 1 (PD-L1), and immune microenvironment characteristics. Whether ICIs are suitable for EGFR-mutated NSCLC patients remains to be elucidated. In this review, we summarized clinical trials of ICIs or combined therapy in EGFR-mutated NSCLC patients. We further discussed the factors determining the efficacy of ICIs in EGFR-mutated NSCLC patients, the mutation subtypes and microenvironment characteristics of potential responders. More importantly, we provided insights into areas worth further investigation in the future.
Collapse
Affiliation(s)
- Pengcheng Zhu
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchJiangsu Cancer Hospital & Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjingChina
- The Fourth Clinical College of Nanjing Medical UniversityNanjingChina
| | - Zhitong Li
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchJiangsu Cancer Hospital & Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjingChina
- The Fourth Clinical College of Nanjing Medical UniversityNanjingChina
| | - Yuxiang Sun
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchJiangsu Cancer Hospital & Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjingChina
- The Fourth Clinical College of Nanjing Medical UniversityNanjingChina
| | - Tongyan Liu
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchJiangsu Cancer Hospital & Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjingChina
- Department of Scientific Research, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical UniversityJiangsu Key Laboratory of Molecular and Translational Cancer ResearchNanjingChina
| | - Rong Yin
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchJiangsu Cancer Hospital & Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjingChina
- Jiangsu Biobank of Clinical ResourcesNanjingChina
- Collaborative Innovation Center for Cancer Personalized MedicineNanjing Medical UniversityNanjingChina
| |
Collapse
|