1
|
Rocha P, Bach R, Masfarré L, Hernandez S, Navarro-Gorro N, Rossell A, Villanueva X, Giner M, Sanchéz I, Galindo M, Del Rey-Vergara R, Iñañez A, Sanchéz-Espiridion B, Lu W, Acedo-Terrades A, Berenguer-Molins P, Sánchez-Font A, Chalela R, Curull V, Taus Á, Hardy-Werbin M, Sausen M, Georgiadis A, White J, Jackson JB, Moliner L, Clavé S, Bellosillo B, Rovira A, Wistuba I, Soto LMS, Perera-Bel J, Arriola E. Molecular and immunological features associated with long-term benefits in metastatic NSCLC patients undergoing immune checkpoint blockade. Oncoimmunology 2025; 14:2469377. [PMID: 39991958 PMCID: PMC11853546 DOI: 10.1080/2162402x.2025.2469377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 01/22/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025] Open
Abstract
INTRODUCTION Immunotherapy is firmly established as a treatment regimen in various solid tumors, driven by its exceptional benefits in a selected group of patients. Despite widespread adoption of immune checkpoint blockade (ICB) across diverse solid tumors, the quest for a clinically informative biomarker for long-term benefit remains unmet. METHODS A total of 49 patients with metastatic NSCLC treated with ICB were included. Long-term (LTR) and short-term responders (STR) were defined as those with a response to ICB lasting more than 24 months or less than 6 months, respectively. Longitudinal blood specimens were collected before ICB treatment initiation and early-on treatment. Plasma ctDNA next-generation sequencing panel (NGS) and serum proteomics were performed. GeoMx DSP on baseline tumor tissue was performed in a subset of patients. RESULTS Our analysis revealed specific characteristics of LTR compared with STR, namely higher PD-L1 in tumor cells (p = 0.005) and higher incidence of irAEs (p = 0.001). Genomic features associated with lack of benefit from ICB included co-occurring mutations in KRAS/STK11 and TP53/KMT2D (p < 0.05). At a baseline, LTR patients exhibited higher serum levels of proteins related with apoptosis (CASP8, PRKRA), chemotaxis, immune proteasome, processing of MHC class I (S100A4, PSMD9, RNF41) and immune homeostasis (HAVCR1, ARG1) (p < 0.05). Protein spatial profiling of tumor samples showed higher levels of proteins linked with the presence of immune cells (CD45), T cells (CD8), antigen presentation (HLA-DR) and immune regulation proteins (PD-L1, IDO1) within the tumor and tumor stroma component (p < 0.05) in LTR patients. Serum longitudinal analysis identified a set of proteins that presented distinct dynamics in LTR compared to STR, making them interesting candidates to evaluate as early predictors of treatment efficacy. CONCLUSIONS Our multimodal analysis of patients with metastatic NSCLC treated with ICB identified clinicopathological and immunological features associated with long-term benefits. The presence of preexisting antitumor immunity emerged as a strong predictor of long-term benefits, providing insights for potential biomarkers and therapeutic strategies for enhancing ICB outcomes in metastatic NSCLC.
Collapse
Affiliation(s)
- Pedro Rocha
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Rafael Bach
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Laura Masfarré
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Sharia Hernandez
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Adrià Rossell
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | | | - Mario Giner
- Pathology Department, Hospital del Mar, Barcelona, Spain
| | | | - Miguel Galindo
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | | | - Albert Iñañez
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Beatriz Sanchéz-Espiridion
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Wei Lu
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | - Victor Curull
- Pulmonology Department, Hospital del Mar, Barcelona, Spain
| | - Álvaro Taus
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | | | | | | | | | | | | | - Sergi Clavé
- Pathology Department, Hospital del Mar, Barcelona, Spain
| | - Beatriz Bellosillo
- Pathology Department, Hospital del Mar, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ana Rovira
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Luisa M Solis Soto
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Edurne Arriola
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|
2
|
Huang Q, Li Y, Huang Y, Wu J, Bao W, Xue C, Li X, Dong S, Dong Z, Hu S. Advances in molecular pathology and therapy of non-small cell lung cancer. Signal Transduct Target Ther 2025; 10:186. [PMID: 40517166 DOI: 10.1038/s41392-025-02243-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/02/2025] [Accepted: 03/31/2025] [Indexed: 06/16/2025] Open
Abstract
Over the past two decades, non-small cell lung cancer (NSCLC) has witnessed encouraging advancements in basic and clinical research. However, substantial unmet needs remain for patients worldwide, as drug resistance persists as an inevitable reality. Meanwhile, the journey towards amplifying the breadth and depth of the therapeutic effect requires comprehending and integrating diverse and profound progress. In this review, therefore, we aim to comprehensively present such progress that spans the various aspects of molecular pathology, encompassing elucidations of metastatic mechanisms, identification of therapeutic targets, and dissection of spatial omics. Additionally, we also highlight the numerous small molecule and antibody drugs, encompassing their application alone or in combination, across later-line, frontline, neoadjuvant or adjuvant settings. Then, we elaborate on drug resistance mechanisms, mainly involving targeted therapies and immunotherapies, revealed by our proposed theoretical models to clarify interactions between cancer cells and a variety of non-malignant cells, as well as almost all the biological regulatory pathways. Finally, we outline mechanistic perspectives to pursue innovative treatments of NSCLC, through leveraging artificial intelligence to incorporate the latest insights into the design of finely-tuned, biomarker-driven combination strategies. This review not only provides an overview of the various strategies of how to reshape available armamentarium, but also illustrates an example of clinical translation of how to develop novel targeted drugs, to revolutionize therapeutic landscape for NSCLC.
Collapse
Affiliation(s)
- Qing Huang
- Department of Medical Oncology, Huazhong University of Science and Technology, Tongji Medical College, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China
| | - Yuanxiang Li
- Department of Medical Oncology, Huazhong University of Science and Technology, Tongji Medical College, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China
| | - Yingdan Huang
- Department of Medical Oncology, Huazhong University of Science and Technology, Tongji Medical College, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China
| | - Jingyi Wu
- Department of Medical Oncology, Huazhong University of Science and Technology, Tongji Medical College, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China
| | - Wendai Bao
- Center for Neurological Disease Research, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Chang Xue
- Department of Medical Oncology, Huazhong University of Science and Technology, Tongji Medical College, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China
| | - Xiaoyu Li
- Department of Medical Oncology, Huazhong University of Science and Technology, Tongji Medical College, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China
| | - Shuang Dong
- Department of Medical Oncology, Huazhong University of Science and Technology, Tongji Medical College, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China
| | - Zhiqiang Dong
- Department of Medical Oncology, Huazhong University of Science and Technology, Tongji Medical College, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China.
- Center for Neurological Disease Research, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
| | - Sheng Hu
- Department of Medical Oncology, Huazhong University of Science and Technology, Tongji Medical College, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China.
| |
Collapse
|
3
|
Zhou X, Yan S, Ma X, Zhu H, Liu B, Yang X, Jia B, Yang Z, Wu N, Li N. Efficacy of radiolabelled PD-L1-targeted nanobody in predicting and evaluating the combined immunotherapy and chemotherapy for resectable non-small cell lung cancer. Eur J Nucl Med Mol Imaging 2025; 52:2343-2354. [PMID: 39912938 DOI: 10.1007/s00259-025-07115-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/24/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND This study aimed to assess the predictive and evaluative value of PD-L1 targeted 68Ga-THP-APN09 PET/CT in the neoadjuvant immunotherapy combined with chemotherapy for resectable non-small cell lung cancer (NSCLC), and to explore its potential in indicating immunotherapy-related adverse effects (irAEs). METHODS Fifty patients with resectable NSCLC enrolled in this prospective study underwent baseline 68Ga-THP-APN09 PET/CT and 18F-FDG PET/CT, with follow-up 18F-FDG PET/CT conducted, additionally, 36 patients received follow-up 68Ga-THP-APN09 PET/CT. Surgery was performed following 2-4 cycles of toripalimab combined with chemotherapy if R0 resection was feasible. The major pathologic response (MPR) state of the post-operative specimen and the adverse effects following combined therapy were documented. The correlation between PD-L1 expression and baseline 68Ga-THP-APN09 PET/CT uptake was determined. The predictive and evaluative efficacies of baseline and follow-up 68Ga-THP-APN09 PET/CT, along with 18F-FDG PET/CT, in determining MPR, were compared. RESULTS The SUVmax values of baseline 68Ga-THP-APN09 PET/CT were significantly higher in patients exhibiting high-positive PD-L1 expression compared to those with low-positive and negative expression (P = 0.001). And the SUVmax values of baseline 68Ga-THP-APN09 PET/CT in the response group, as determined by 18F-FDG PET/CT evaluation, were significantly higher than those in the non-response group (3.4 vs. 2.4, P < 0.001). Totally, 41 patients underwent surgery, of which 27 achieved MPR, while 14 did not. The SUVmax in baseline 68Ga-THP-APN09 PET/CT demonstrated statistical significance between the MPR and non-MPR groups, with area under the ROC curve (AUC) of 0.88 (95%CI: 0.77-0.99) in identifying MPR. However, the SUVmax in baseline 18F-FDG PET/CT failed to demonstrated significant predictive power, with AUC values of 0.68 (95%CI: 0.50-0.86, P = 0.076). While the SUVmax in follow-up 68Ga-THP-APN09 and 18F-FDG PET/CT, along with their change rate (ΔSUVmax%), demonstrated good predictive efficacy in identifying MPR, with AUC values of 0.81 (0.64-0.98), 0.91 (0.82-1.00), 0.93 (0.84-1.00), and 0.96 (0.89-1.00), respectively. Furthermore, the follow-up 68Ga-THP-APN09 PET/CT could remarkedly indicate the potential for thyroiditis. CONCLUSION Baseline 68Ga-THP-APN09 PET/CT alone could predict efficacy and assist in patient screening for immunotherapy combined chemotherapy in resectable NSCLC, and the follow-up 68Ga-THP-APN09 PET/CT and their change rates could aid in therapy evaluation. Additionally, follow-up 68Ga-THP-APN09 PET/CT could be utilized to monitor the immunotherapy-related thyroiditis during the therapy. TRIAL REGISTRATION NCT05156515, registered 8 December 2021, https://register. CLINICALTRIALS gov/prs/app/action/SelectProtocol?sid=S000BMSI%26;selectaction=Edit%26;uid=U000503E%26;ts=2%26;cx=zeghuw .
Collapse
Affiliation(s)
- Xin Zhou
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing, 100142, China
| | - Shi Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Xiaopan Ma
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing, 100142, China
- Department of Nuclear Medicine, Affiliated Hospital of Hubei, Xiangyang Central Hospital, University of Arts and Science, Xiangyang, 441138, China
| | - Hua Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing, 100142, China
| | - Bing Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Xin Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Bing Jia
- Medical Isotopes Research Center, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, No.38 Xueyuan Rd, Beijing, 100191, China.
| | - Zhi Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing, 100142, China.
| | - Nan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China.
| | - Nan Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing, 100142, China.
| |
Collapse
|
4
|
Du W, Zhan J, Wu K, Wang Y, Zhang L, Hong S. Early kinetics of serum amyloid A predict clinical benefit to first-line chemoimmunotherapy and immunotherapy in advanced non-small cell lung cancer: a retrospective analysis. Biomark Res 2025; 13:76. [PMID: 40410893 PMCID: PMC12102904 DOI: 10.1186/s40364-025-00791-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have transformed the treatment landscape for advanced non-small cell lung cancer (NSCLC), yet durable responses remain limited in a subset of patients. Serum amyloid A (SAA), an acute-phase protein linked to systemic inflammation, may reflect dynamic immune responses. This retrospective study analyzed 242 advanced NSCLC patients treated with first-line chemoimmunotherapy or immunotherapy between August 2016 and December 2024. Patients were stratified by early SAA kinetics into flare-responders (initial rise followed by decline), responders (sustained decline), and non-responders. Clinical outcomes, including progression-free survival (PFS) and overall survival (OS), were evaluated using Kaplan-Meier and Cox regression analyses. In the chemoimmunotherapy cohort, SAA flare-responders demonstrated significantly prolonged median PFS (29.8 months, 95% CI: 9.95-49.65; HR: 0.31, 95% CI: 0.15-0.64; p < 0.01) compared to non-responders (7.4 months, 95% CI: 4.67-10.13). Similarly, in the immunotherapy cohort, SAA flare-responders showed superior PFS. (19.9 vs. 2.1 months, HR 0.31, p < 0.01). Multivariate analysis confirmed early SAA kinetics as an independent prognostic factor for both PFS and OS in both treatment groups. Early SAA kinetics serve as a promising non-invasive biomarker for predicting clinical outcomes in advanced NSCLC treated with first-line chemoimmunotherapy or immunotherapy. These findings highlight SAA kinetics as a potential non-invasive biomarker and monitoring SAA dynamics may aid in identifying patients with higher likelihood of clinical benefit; however, prospective studies are required to determine its utility in guiding therapeutic decisions.
Collapse
Affiliation(s)
- Wei Du
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Jianhua Zhan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Kai Wu
- Shantou University Medical College, Shantou University, Shantou, China
| | - Yanming Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
- Department of VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
| | - Shaodong Hong
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
| |
Collapse
|
5
|
Zheng Y, Sadée C, Ozawa M, Howitt BE, Gevaert O. Single-cell multimodal analysis reveals tumor microenvironment predictive of treatment response in non-small cell lung cancer. SCIENCE ADVANCES 2025; 11:eadu2151. [PMID: 40408481 PMCID: PMC12101509 DOI: 10.1126/sciadv.adu2151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 04/22/2025] [Indexed: 05/25/2025]
Abstract
Non-small cell lung cancer (NSCLC) constitutes over 80% of lung cancer cases and remains a leading cause of cancer-related mortality worldwide. Despite the advent of immune checkpoint inhibitors, their efficacy is limited to 27 to 45% of patients. Identifying likely treatment responders is essential for optimizing healthcare and improving quality of life. We generated multiplex immunofluorescence (mIF) images, histopathology, and RNA sequencing data from human NSCLC tissues. Through the analysis of mIF images, we characterized the spatial organization of 1.5 million cells based on the expression levels for 33 biomarkers. To enable large-scale characterization of tumor microenvironments, we developed NucSegAI, a deep learning model for automated nuclear segmentation and cellular classification in histology images. With this model, we analyzed the morphological, textural, and topological phenotypes of 45.6 million cells across 119 whole-slide images. Through unsupervised phenotype discovery, we identified specific lymphocyte phenotypes predictive of immunotherapy response. Our findings can improve patient stratification and guide selection of effective therapeutic regimens.
Collapse
Affiliation(s)
- Yuanning Zheng
- Stanford Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Christoph Sadée
- Stanford Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Michael Ozawa
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Brooke E. Howitt
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA 94305, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
6
|
Pu X, Shou J, Xiao Z, Chen J, Xiao M, Guo Q, Ma Z, Hong W, Wang Q, Wang Y, Li J, Rao C, Weng J, Lu L, Wu L, Fang Y. Anlotinib Plus Docetaxel is Promising in Advanced NSCLC Progressing on First-Line Immunotherapy: A Pooled Analysis of Two Randomized Trials. Adv Ther 2025:10.1007/s12325-025-03170-2. [PMID: 40354010 DOI: 10.1007/s12325-025-03170-2] [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: 10/31/2024] [Accepted: 03/05/2025] [Indexed: 05/14/2025]
Abstract
INTRODUCTION Therapeutic options for patients with advanced non-small cell lung cancer (NSCLC) after progression on immune checkpoint inhibitors (ICIs) remain a significant challenge. This analysis compared outcomes of anlotinib plus docetaxel versus docetaxel alone in this population using the pooled data from two prospective randomized trials (ALTER-L016; ALTER-L018). METHODS Adult patients with EGFR/ALK/ROS1 wild-type advanced NSCLC progressing on first-line ICIs were eligible. Patients were randomly assigned to anlotinib plus docetaxel or docetaxel alone. The primary endpoint was progression-free survival (PFS). The secondary endpoints were overall survival (OS), objective response rate (ORR), disease control rate (DCR), and safety. RESULTS Seventy-one patients were included in this pooled analysis (L016, n = 39; L018, n = 32), of whom, 40 received anlotinib plus docetaxel and 31 received docetaxel. The median follow-up of all patients was 27.0 months. The median PFS was longer with anlotinib plus docetaxel than with docetaxel alone (5.4 months [95% CI 5.0-9.3] vs. 2.3 months [95% CI 1.4-2.9]; hazard ratio [HR], 0.34; 95% CI 0.18-0.63; P < 0.001). Both ORR (25.0% vs. 12.9%) and DCR (82.5% vs. 45.2%) were higher in the anlotinib plus docetaxel group than in the docetaxel group. Median OS was 16.2 months (95% CI 8.3-21.3) with anlotinib plus docetaxel versus 13.7 months (95% CI 4.6-22.3) with docetaxel (HR = 0.82; 95% CI 0.47-1.44; P = 0.488). Subsequent ICI therapy was associated with a longer OS. Grade 3 treatment-related adverse events occurred in 32.5% of patients receiving anlotinib plus docetaxel and 6.5% of patients receiving docetaxel. CONCLUSION Anlotinib plus docetaxel improved PFS but not OS versus docetaxel in patients with advanced NSCLC progressing on ICIs. Larger standardized phase 3 trials are needed to verify these findings. TRIAL REGISTRATION ALTER-L016 (ClinicalTrials.gov identifier, NCT03726736). ALTER-L018 (ClinicalTrials.gov identifier, NCT03624309).
Collapse
Affiliation(s)
- Xingxiang Pu
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jiawei Shou
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zemin Xiao
- Department of Oncology, The First People's Hospital of Changde City, Changde, China
| | - Jun Chen
- Tumor Radiochemotherapy Department, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Maoliang Xiao
- Department of Oncology, Hunan Province Directly Affiliated TCM Hospital, Zhuzhou, China
| | - Qunyi Guo
- Hematology Oncology, Taizhou Hospital of Zhejiang Province, Taizhou, China
| | - Zhongxia Ma
- Department of Thoracic Oncology Surgery, The First People's Hospital of Chenzhou City, Chenzhou, China
| | - Wei Hong
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Qianzhi Wang
- Department of Thoracic Medical Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yonghui Wang
- Department of Medical Oncology, Thoracic Cancer, Lishui Municipal Central Hospital, Lishui, China
| | - Jia Li
- Department of Thoracic Medical Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Chuangzhou Rao
- Department of Radiotherapy and Chemotherapy, Ningbo No.2 Hospital, Ningbo, China
| | - Jie Weng
- Department of Oncology, Yueyang Central Hospital, Yueyang, China
| | - Liqin Lu
- Medical Oncology, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Lin Wu
- Department of Thoracic Medical Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.
| | - Yong Fang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
7
|
Hata T, Yamada T, Goto Y, Amano A, Negi Y, Watanabe S, Furuya N, Oba T, Ikoma T, Nakao A, Tanimura K, Taniguchi H, Yoshimura A, Fukui T, Murata D, Kaira K, Shiotsu S, Hibino M, Okada A, Chihara Y, Kawachi H, Kijima T, Takayama K. Regimen Selection for Chemoimmunotherapy in Nonsquamous Non-Small Cell Lung Cancer with Low PD-L1 Expression: A Multicenter Retrospective Cohort Study. Clin Lung Cancer 2025; 26:e190-e198.e4. [PMID: 39864962 DOI: 10.1016/j.cllc.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/16/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND Although chemoimmunotherapy is recommended for advanced nonsquamous non-small cell lung cancer (NSCLC) with low programmed cell death ligand 1 (PD-L1) expression, no head-to-head comparisons of immune checkpoint inhibitors (ICIs) have been performed. Therefore, we compared the effect and safety of regimens in these patients to guide evidence-based treatment. METHODS This retrospective study included patients with advanced nonsquamous NSCLC with a PD-L1 tumor proportion score of 1% to 49% administered ICI combination platinum-based chemotherapy between May 2018 and May 2023 at 19 institutions in Japan. The main analysis compared survival outcomes and the incidence of grade ≥3 adverse events among regimens. RESULTS Among 316 included patients (median [range] age, 69 [36-89] years; 242 males; 41 never smokers), 200 (63%), 68 (22%), and 48 (15%) received chemotherapy combined with anti-programmed cell death protein 1 (PD-1), anti-PD-L1, and anti-PD-1/cytotoxic T-lymphocyte associated protein 4 (CTLA-4) antibodies, respectively. The median overall survival times were 28.6, 23.1, and 24.4 months (P = .41), and the median progression-free survival times were 9.4, 7.2, and 8.7 months (P = .28) in the anti-PD-1/Chemo, anti-PD-L1/Chemo and anti-PD-1/CTLA-4/Chemo groups, respectively. The anti-PD-1/CTLA-4/Chemo group had the lowest incidence of hematologic toxicity (P = .13) and the highest incidence of nonhematologic toxicity (P = .07). The incidence of grade ≥3 pneumonitis was significantly lower in the anti-PD-L1/Chemo group (P = .049). CONCLUSIONS Despite comparable survival benefits, adverse events differed among three regimens in patients with low PD-L1 expression. Notably, anti-PD-L1 antibody combination chemotherapy may reduce the risk of severe pneumonitis.
Collapse
Affiliation(s)
- Tae Hata
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tadaaki Yamada
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Yasuhiro Goto
- Department of Respiratory Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| | - Akihiko Amano
- Department of Respiratory Medicine, Kurashiki Central Hospital, Kurashiki, Japan
| | - Yoshiki Negi
- Department of Respiratory Medicine and Hematology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Satoshi Watanabe
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Naoki Furuya
- Division of Respiratory Medicine, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Tomohiro Oba
- Department of Respiratory Medicine, Saitama Red Cross Hospital, Saitama, Japan
| | - Tatsuki Ikoma
- Department of Thoracic Oncology, Kansai Medical University, Hirakata, Japan
| | - Akira Nakao
- Department of Respiratory Medicine, Fukuoka University Hospital, Fukuoka, Japan
| | - Keiko Tanimura
- Department of Medical Oncology, Fukuchiyama City Hospital, Fukuchiyama, Japan
| | - Hirokazu Taniguchi
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Akihiro Yoshimura
- Department of Respiratory Medicine, Japanese Red Cross Kyoto Daini Hospital, Kyoto, Japan
| | - Tomoya Fukui
- Department of Respiratory Medicine, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Daiki Murata
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Kyoichi Kaira
- Department of Respiratory Medicine, International Medical Center, Saitama Medical University, Hidaka, Japan
| | - Shinsuke Shiotsu
- Department of Respiratory Medicine, Japanese Red Cross Kyoto Daiichi Hospital, Kyoto, Japan
| | - Makoto Hibino
- Department of Respiratory Medicine, Shonan Fujisawa Tokushukai Hospital, Fujisawa, Japan
| | - Asuka Okada
- Department of Respiratory Medicine, Saiseikai Suita Hospital, Suita, Japan
| | - Yusuke Chihara
- Department of Respiratory Medicine, Uji-Tokushukai Medical Center, Uji, Japan
| | - Hayato Kawachi
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takashi Kijima
- Department of Respiratory Medicine and Hematology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Koichi Takayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
8
|
Hu Z, Hu Z, Chen K, Huang H, Zhong X, Wang Y, Chen J, He X, Shi D, Zeng Y, Li J, Zhou X, Wei P. The Spatial Proximity of CD8 + FoxP3 +PD-1 + Cells to Tumor Cells: A More Accurate Predictor of Immunotherapy Outcomes in Advanced Non-Small-Cell Lung Cancer. Curr Oncol 2025; 32:262. [PMID: 40422521 DOI: 10.3390/curroncol32050262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND To optimize precision immunotherapy for advanced NSCLC, comprehensive tumor immune microenvironment (TIME) characterization is crucial for efficacy prediction. METHODS Pretreatment tumor samples from 46 advanced NSCLC patients treated with PD-1/PD-L1 inhibitors were analyzed. The subregional abundance and spatial proximity scores of TIME cell subpopulations in 27 samples were assessed via multiplex immunohistochemistry (mIHC) targeting pan-CK, CD163, CD8, FoxP3, PD-1, and PD-L1. Correlations between the TIME features, clinicopathologic factors, treatment response, and prognosis were evaluated. RESULTS CD8+FoxP3+ cells were identified in NSCLC tissues, predominantly expressing PD-1/PD-L1. The PD-L1 TPS subgroups showed significant immune cell density/proximity differences, but CD8+FoxP3+PD-1+ infiltration was PD-L1 TPS-independent. Responders had higher CD8+FoxP3+PD-1high density (p = 0.0497) and proximity scores (p = 0.0099) than non-responders. The CD8+FoxP3+PD-1+ presence and tumor proximity were essential for favorable outcomes. In low-PD-L1 TPS patients, the CD8+FoxP3+PD-1+ abundance and proximity scores strongly predicted the response (AUC: 0.79 and 0.75 vs. PD-L1 TPS AUC = 0.58). A survival analysis linked the presence and proximity score of CD8+FoxP3+PD-1+ cells to prolonged overall survival (OS) and progression-free survival (PFS). Notably, a low proximity score of CD8+FoxP3+PD-1+ cells emerged as an independent risk factor for a shorter PFS (HR = 6.16, 95% CI: 2.12-17.93, p = 0.001). CONCLUSION The CD8+FoxP3+PD-1+ spatial proximity to tumor cells robustly predicts improved immunotherapy outcomes in advanced NSCLC.
Collapse
Affiliation(s)
- Zijuan Hu
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Zhihuang Hu
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Keji Chen
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Huixia Huang
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Xinyang Zhong
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Yaxian Wang
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Jiayu Chen
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Xuefeng He
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Di Shi
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
- Institute of Pathology, Fudan University, 270 Dong'an Road, Shanghai 200032, China
| | - Yupeng Zeng
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
- Institute of Pathology, Fudan University, 270 Dong'an Road, Shanghai 200032, China
| | - Jiwei Li
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xiaoyan Zhou
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
- Institute of Pathology, Fudan University, 270 Dong'an Road, Shanghai 200032, China
| | - Ping Wei
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| |
Collapse
|
9
|
Hektoen HH, Tsuruda KM, Brustugun OT, Neumann K, Andreassen BK. Real-world comparison of pembrolizumab alone and combined with chemotherapy in metastatic lung adenocarcinoma patients with PD-L1 expression ≥50. ESMO Open 2025; 10:105073. [PMID: 40305908 PMCID: PMC12088754 DOI: 10.1016/j.esmoop.2025.105073] [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: 07/28/2024] [Revised: 03/10/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
OBJECTIVES The frontline treatment of metastatic lung adenocarcinoma with high Programmed death-ligand 1 (PD-L1) expression (≥50%) includes immune checkpoint inhibitors (ICIs) either as monotherapy or combined with chemotherapy. The added benefit of chemotherapy in this context lacks direct comparison in head-to-head trials. We aimed to compare these two ICI treatment modalities both overall and within relevant patient subgroups in a real-world setting. MATERIALS AND METHODS This retrospective, nationwide study included 410 individuals diagnosed in Norway during 2017 to 2021 with stage IV non-small-cell lung adenocarcinoma, PD-L1 expression ≥50%, and treated first line with the ICI pembrolizumab, either as monotherapy (n = 317) or in combination with platinum-based chemotherapy (n = 93). We analyzed early (6-month) and overall (3-year) risk of death after treatment initiation using Cox regression, adjusted for and stratified by sex, age, stage, PD-L1 expression, performance status, and education. RESULTS Patients treated with combination therapy had a higher median overall survival compared with monotherapy (22.6 months versus 14.2 months), and reduced risk of overall death, although not statistically significant after adjustment [hazard ratio (HR) 0.74, 95% confidence interval (CI) 0.54-1.00]. However, the risk of early death was significantly lower in patients receiving combination therapy, even after adjustment (HR 0.41, 95% CI 0.23-0.76). Across most subgroups, patients receiving combination therapy had comparable or superior survival outcomes relative to those receiving monotherapy. Particularly noteworthy were the observed benefits from combination therapy over monotherapy among females, individuals with stage IVB disease, and those with PD-L1 expression exceeding 75%. CONCLUSION Our real-world study demonstrates that combination therapy with ICI and chemotherapy provides superior early survival benefits over monotherapy in PD-L1-high patients. Additionally, certain subgroups showed enhanced overall survival. These findings challenge current treatment practices and underscore the need for further validation to optimize patient selection for monotherapy versus combination therapy, in particular to reassess the role of PD-L1 in treatment decisions.
Collapse
Affiliation(s)
- H H Hektoen
- Department of Research, Cancer Registry of Norway, Norwegian Institute of Public Health Oslo, Norway; Department of Cancer Genetics, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway. https://twitter.com/HektoenHH
| | - K M Tsuruda
- Department of Research, Cancer Registry of Norway, Norwegian Institute of Public Health Oslo, Norway
| | - O T Brustugun
- Section of Oncology, Drammen Hospital, Vestre Viken Health Trust, Drammen, Norway; Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - K Neumann
- Department of Pulmonology, Akershus University Hospital, Lørenskog, Norway
| | - B K Andreassen
- Department of Research, Cancer Registry of Norway, Norwegian Institute of Public Health Oslo, Norway.
| |
Collapse
|
10
|
Orooji N, Babaei S, Fadaee M, Abbasi-Kenarsari H, Eslami M, Kazemi T, Yousefi B. Novel therapeutic approaches for non-small cell lung cancer: an updated view. J Drug Target 2025:1-16. [PMID: 40186594 DOI: 10.1080/1061186x.2025.2489986] [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: 03/05/2025] [Revised: 03/24/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Non-small cell lung cancer (NSCLC) continues to be one of the leading causes of cancer-related mortality globally. Most patients who undergo surgical procedures may encounter distant metastasis or local recurrence, necessitating supplementary treatments such as radiation therapy, chemotherapy, or targeted therapy as adjuvant alternatives. Recent advancements in molecular biology and immunotherapy have paved the way for innovative therapeutic approaches that target specific genetic mutations and promote the immune response against tumour cells. This review explores emerging therapies, including targeted therapies such as tyrosine kinase inhibitors (TKIs) for actionable mutations (e.g., EGFR, ALK, ROS1), as well as the role of immune checkpoint inhibitors (ICIs) that employ the body's immune system to combat cancer. Additionally, we discuss the potential of exosome therapies, as well as promising nanotherapeutic options for the treatment of NSCLC. This study attempts to provide a thorough overview of the changing landscape of NSCLC treatment and its implications for enhancing patient outcomes by presenting these innovative techniques.
Collapse
Affiliation(s)
- Niloufar Orooji
- Department of Immunology, School of Medicine, Semnan University of Medical Science, Semnan, Iran
| | - Shabnam Babaei
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Manouchehr Fadaee
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajar Abbasi-Kenarsari
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Majid Eslami
- Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Department of Immunology, School of Medicine, Semnan University of Medical Science, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
11
|
Kumar RI, Jain K, Rai KR, Arora P, Gururajan H, Sarkar K. Function of antigen-presenting cells in non-small-cell lung cancer (NSCLC). Med Oncol 2025; 42:162. [PMID: 40221637 DOI: 10.1007/s12032-025-02703-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 03/29/2025] [Indexed: 04/14/2025]
Abstract
The most common type of lung cancer called NSCLC avoids immune monitoring by blocking antigen display and T cell response activation. Anti-tumor immunity requires the essential function of antigen-presenting cells (APCs) which include dendritic cells and macrophages and B cells. NSCLC causes APCs to stop their normal function because they fail to properly display tumor antigens and activate adaptive immune responses. APC dysfunction in NSCLC is mainly caused by the tumor microenvironment (TME) which actively reprograms these cells through inhibitory cytokines and metabolic constraints and immune checkpoints. As a result, NSCLC exhibits poor responses to immunotherapies, such as checkpoint inhibitors. The analysis of APC-TME interactions enables researchers to develop strategies that will enhance APC function along with antigen presentation while improving immunotherapy effectiveness. The research examines APC dysfunction in NSCLC together with its TME mechanisms and develops therapeutic strategies to combat immune suppression for better clinical results.
Collapse
Affiliation(s)
- R Ilaya Kumar
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Kavya Jain
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Karan Raj Rai
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Prashasti Arora
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Harshnna Gururajan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Koustav Sarkar
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India.
| |
Collapse
|
12
|
Zhou X, Yan S, Li D, Zhu H, Liu B, Liu S, Zhao W, Yang Z, Wu N, Li N. Radiolabelled anti-PD-L1 peptide PET/CT in predicting the efficacy of neoadjuvant immunotherapy combined with chemotherapy in resectable non-small cell lung cancer. Ann Nucl Med 2025; 39:364-372. [PMID: 39673015 DOI: 10.1007/s12149-024-02009-0] [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: 06/26/2024] [Accepted: 12/02/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND This study aimed to evaluate the predictive value of baseline PD-L1 targeted peptide 68Ga-NOTA-WL12 PET/CT in neoadjuvant immunotherapy combined with chemotherapy of resectable NSCLC. METHODS Patients with resectable NSCLC (n = 20) enrolled in this prospective study received baseline paired 68Ga-NOTA-WL12 PET/CT and 18F-FDG PET/CT. After 2-4 cycles of toripalimab plus nab-paclitaxel and cisplatin, surgery was performed if R0 resection was available. The major pathologic response (MPR) state of the post-operative specimen was recorded. The imaging parameters of the 68Ga-NOTA-WL12 PET/CT, 18F-FDG PET/CT and CT between the MPR and non-MPR groups and their predictive efficacy of MPR were compared. RESULTS Among 20 patients, 17 patients underwent surgery, 10 achieved an MPR and 7 did not. The SUVmax and tumour-to-blood pool (TBR) of baseline 68Ga-NOTA-WL12 in the MPR group were higher than those in the non-MPR group, and the difference in TBR was statistically significant. The ΔSULpeak% of 18F-FDG exhibited differences between the MPR and non-MPR groups with no significance. Baseline 18F-FDG PET/CT parameters and ΔD% failed to differentiate the two groups. The areas under the ROC curves of SUVmax, TBR in 68Ga-NOTA-WL12 PET/CT, ΔD% and ΔSULpeak% in 18F-FDG PET/CT were 0.76, 0.79, 0.71 and 0.80, respectively, in predicting MPR. CONCLUSION Baseline 68Ga-NOTA-WL12 PET/CT has a potential to predict the pathological response of neoadjuvant immunotherapy combined with chemotherapy in patients with resectable NSCLC, whose efficacy is comparable to that of therapy evaluations employing baseline and follow-up CT and 18F-FDG PET/CT examinations. TRIAL REGISTRATION NCT04304066, registered 13 November 2020, https://register. CLINICALTRIALS gov/prs/app/action/SelectProtocol?sid=S000AEI9&selectaction=Edit&uid=U000503E&ts=2&cx=-awajet .
Collapse
Affiliation(s)
- Xin Zhou
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Shi Yan
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Dan Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Hua Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Bing Liu
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Shiwei Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Wei Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Zhi Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China.
| | - Nan Wu
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China.
| | - Nan Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China.
| |
Collapse
|
13
|
Tu DH, Qu R, Wen F, Zhou Q, Liu Q, Huang L, Chen T. Successful conversion surgery following tislelizumab with chemotherapy in a patient with stage IIIC lung adenocarcinoma harboring RET fusions: A case report and review of the literature. Exp Ther Med 2025; 29:70. [PMID: 39991722 PMCID: PMC11843209 DOI: 10.3892/etm.2025.12820] [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: 08/15/2024] [Accepted: 01/28/2025] [Indexed: 02/25/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have emerged as a beacon of hope for most patients with stage III non-small cell lung cancer (NSCLC) who are no longer surgical candidates. However, the literature on the use of immunotherapy in patients with NSCLC with rearranged during transfection (RET) gene fusions is scant. The present study reports the case of a 61-year-old female patient, diagnosed with stage IIIC lung adenocarcinoma, exhibiting two RET gene fusions and high programmed death-ligand 1 expression. Following four treatment cycles of tislelizumab in combination with pemetrexed and cisplatin, the patient was successfully downstaged, enabling radical surgery. The post-operative pathology analysis indicated a major pathologic response. This case study contributes to the growing body of evidence supporting the use of ICIs in treating locally advanced NSCLC with RET gene fusions.
Collapse
Affiliation(s)
- De-Hao Tu
- Department of Thoracic Surgery, Yueyang Central Hospital, Yueyang, Hunan 414000, P.R. China
| | - Rirong Qu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Fang Wen
- Department of Oncology, Yueyang Central Hospital, Yueyang, Hunan 414000, P.R. China
| | - Qiang Zhou
- Department of Oncology, Yueyang Central Hospital, Yueyang, Hunan 414000, P.R. China
| | - Qianyun Liu
- Department of Medical Imaging, Yueyang Central Hospital, Yueyang, Hunan 414000, P.R. China
| | - Lingmei Huang
- Department of Pulmonary and Critical Care Medicine, Yueyang Central Hospital, Yueyang, Hunan 414000, P.R. China
| | - Tao Chen
- Department of Thoracic Surgery, Yueyang Central Hospital, Yueyang, Hunan 414000, P.R. China
| |
Collapse
|
14
|
Ji X, Liu M, Zhang T, Zhang W, Xue F, Wan Q, Liu Y. KRAS/PI3K axis driven GTF3C6 expression and promotes LUAD via FAK pathway. J Adv Res 2025; 70:243-254. [PMID: 38685529 PMCID: PMC11976405 DOI: 10.1016/j.jare.2024.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024] Open
Abstract
INTRODUCTION Effective targeting drugs for KRAS mutation-mediated Lung Adenocarcinoma (LUAD) are currently are limited. OBJECTIVES Investigating and intervening in the downstream key target genes of KRAS is crucial for clinically managing KRAS mutant-driven LUAD. GTF3C6, a newly identified member of the general transcription factor III (GTF3) family, plays a role in the transcription of RNA polymerase III (pol III)-dependent genes. However, its involvement in cancer remains unexplored. METHODS This study examined the expression, roles, and potential molecular mechanisms of GTF3C6 in LUAD tissues, LSL-KrasG12D/+;LSL-p53-/- LUAD mouse models, and LUAD patients-derived organoid using Western blot, qRT-PCR, immunofluorescence, immunohistochemistry, and gene manipulation assays. RESULTS We present the first evidence that GTF3C6 is highly expressed in LUAD tissues, LSL-KrasG12D/+;LSL-p53-/- LUAD mouse models, and LUAD organoids, correlating with poor clinical prognosis. Furthermore, GTF3C6 was found to promote anchorage-independent proliferation, migration, and invasion of LUAD cells. Mechanistically, KRAS mutation drives GTF3C6 expression through the PI3K pathway, and GTF3C6 knockdown reverses the malignant phenotype of KRAS mutation-driven LUAD cells. Additionally, the FAK pathway emerged as a crucial downstream signaling pathway through which GTF3C6 mediates the malignant phenotype of LUAD. Finally, GTF3C6 knockdown suppresses LUAD organoid formation and inhibits tumor growth in vivo. CONCLUSION Our findings demonstrate that GTF3C6, driven by KRAS mutation, promotes LUAD development by regulating FAK phosphorylation, suggesting its potential as a biomarker and therapeutic target in KRAS mutant-driven LUAD.
Collapse
Affiliation(s)
- Xingzhao Ji
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Infections Respiratory Disease, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mingqiang Liu
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Department of Pharmacy, Pingdu People's Hospital, Qingdao, Shandong 266799, China
| | - Tianyi Zhang
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Weiying Zhang
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Fuyuan Xue
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Qiang Wan
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| | - Yi Liu
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Infections Respiratory Disease, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
15
|
Liu T, Ji W, Cheng X, Lv L, Yu X, Wang N, Li M, Hu T, Shi Z. Revealing a Novel Methylated Integrin Alpha-8 Related to Extracellular Matrix and Anoikis Resistance Using Proteomic Analysis in the Immune Microenvironment of Lung Adenocarcinoma. Mol Biotechnol 2025; 67:1137-1155. [PMID: 38514598 DOI: 10.1007/s12033-024-01114-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 02/07/2024] [Indexed: 03/23/2024]
Abstract
Genomic epigenetics of extracellular matrix (ECM) play an important role in lung adenocarcinoma (LUAD). Our study identified a signature of potential prognostic genes associated with ECM and constructed immune risk-related prognosis model in LUAD. We downloaded mRNAs transcriptome data, miRNAs expression data, and clinical patient information for LUAD based on The Cancer Genome Atlas. "Limma, clusterProfiler, ggplot2" R packages and GSEA were used to analyze meaningful genes and explore potential biological function. A competing endogenous RNA network was constructed to reveal the mechanism of ECM-related genes. Combined with clinical LUAD patients' characteristics, univariate and multivariate Cox regression analyses were used to build prognostic immune risk model. Next, we calculated AUC value of ROC curve, and explored survival probability of different risk groups. A total of 2966 mRNAs were differently expressed in LUAD samples and normal samples. Function enrichment analyses proved mRNAs were associated with many tumor pathways, such as cell adhesion, vascular smooth muscle contraction, and cell cycle. There were 18 mRNAs related to ECM receptor signaling pathway, and 7 mRNAs expressions were correlated with EGFR expression, but only 5mRNAs were associated with the long-term prognosis. Based on Integrin alpha-8 (ITGA8) molecule, we identified potential 3 miRNAs from several databases. The promoter of ITGA8 was higher-methylated and lower-expressed in LUAD. And lower-expressed group has poor prognosis for patients. 66 immunomodulators related to ITGA8 were performed to construct immune correlation prediction model (p < 0.05). Comprehensive analyses of ITGA8 revealed it combined focal adhesion kinase to activate PI3K/AKT signaling pathway to influence the occurrence and development of LUAD. A novel immune prognostic model about ITGA8 was constructed and verified in LUAD patients. Combined with non-coding genes and genomic epigenetics, identification of potential biomarkers provided new light on therapeutic strategy for clinical patients.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xian Jiaotong University, Xian, Shanxi, China
| | - Wen Ji
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xian Jiaotong University, Xian, Shanxi, China
| | - Xue Cheng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xian Jiaotong University, Xian, Shanxi, China
| | - Lin Lv
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xian Jiaotong University, Xian, Shanxi, China
| | - Xiaohui Yu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xian Jiaotong University, Xian, Shanxi, China
| | - Na Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xian Jiaotong University, Xian, Shanxi, China
| | - Mengcong Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xian Jiaotong University, Xian, Shanxi, China
| | - Tinghua Hu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xian Jiaotong University, Xian, Shanxi, China
| | - Zhihong Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xian Jiaotong University, Xian, Shanxi, China.
| |
Collapse
|
16
|
Dong S, Li X, Huang Q, Li Y, Li J, Zhu X, Xue C, Chen R, Zeng Y, Wu J, Zhong Y, Hu S. Resistance to immunotherapy in non-small cell lung cancer: Unraveling causes, developing effective strategies, and exploring potential breakthroughs. Drug Resist Updat 2025; 81:101215. [PMID: 40081220 DOI: 10.1016/j.drup.2025.101215] [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: 08/21/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 03/15/2025]
Abstract
Over the last two decades, advancements in deciphering the intricate interactions between oncology and immunity have fueled a meteoric rise in immunotherapy for non-small cell lung cancer, typified by an explosive growth of immune checkpoint inhibitors. However, resistance to immunotherapy remains inevitable. Herein we unravel the labyrinthine mechanisms of resistance to immunotherapy, characterized by their involvement of nearly all types of cells within the body, beyond the extrinsic cancer cells, and importantly, such cells are not only (inhibitory or excitatory, or both) signal recipients but also producers, acting in a context-dependent manner. At the molecular level, these mechanisms underlie genetic and epigenetic aberrations, which are regulated by or regulate various protein kinases, growth factors, and cytokines with inherently dynamic and spatially heterogeneous properties. Additionally, macroscopic factors such as nutrition, comorbidities, and the microbiome within and around organs or tumor cells are involved. Therefore, developing therapeutic strategies combined with distinct action informed by preclinical, clinical, and real-world evidence, such as radiotherapy, chemotherapy, targeted therapy, antibody-drug conjugates, oncolytic viruses, and cell-based therapies, may stand as a judicious reality, although the ideality is to overcome resistance point-by-point through a novel drug. Notably, we highlight a realignment of treatment aims, moving the primary focus from eliminating cancer cells -- such as through chemotherapy and radiotherapy -- to promoting immune modulation and underscore the value of regulating various components within the host macro- or micro-environment, as their effects, even if seemingly minimal, can cumulatively contribute to visible clinical benefit when applied in combination with ICIs. Lastly, this review also emphasizes the current hurdles scattered throughout preclinical and clinical studies, and explores evolving directions in the landscape of immunotherapy for NSCLC.
Collapse
Affiliation(s)
- Shuang Dong
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Xiaoyu Li
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Qing Huang
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Yuanxiang Li
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | | | - Xianmin Zhu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Chang Xue
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Runzhi Chen
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Yuan Zeng
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Jingyi Wu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Yi Zhong
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China.
| | - Sheng Hu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China.
| |
Collapse
|
17
|
Wang H, Cheng L, Chen J, Chen P, Tang Z, Wang Q, Ma Y, Zhao C, Li X, Jiang T, Zhou F, Chen X, Zhou C. Efficacy of PD-1 blockade plus chemotherapy in patients with oncogenic-driven non-small-cell lung cancer. Cancer Immunol Immunother 2025; 74:89. [PMID: 39891730 PMCID: PMC11787076 DOI: 10.1007/s00262-024-03937-6] [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: 09/24/2024] [Accepted: 12/30/2024] [Indexed: 02/03/2025]
Abstract
BACKGROUND PD-1 blockade plus chemotherapy has become the first-line standard of care for patients with advanced non-small-cell lung cancer (NSCLC) without oncogenic drivers. Oncogenic-driven advanced NSCLC showed limited response to PD-1 blockade monotherapy or chemotherapy alone. Whether NSCLC patients with oncogenic drivers could benefit from PD-1 blockade plus chemotherapy remains undetermined. METHODS Three hundred twelve NSCLC patients with at least one oncogenic driver alteration received PD-1 plus chemotherapy or each monotherapy were retrospectively identified. Objective response rate (ORR), progression-free survival (PFS), and overall survival (OS) were compared to evaluate the therapeutic outcomes differences among patients with different oncogenic drivers. RESULTS One hundred sixty-two patients received PD-1 blockade plus chemotherapy, 57 received PD-1 blockade monotherapy and 93 received chemotherapy alone were included. Oncogenic driver mutations including KRAS (31.4%), EGFR (28.8%), HER2 (14.7%), BRAF (10.6%), RET (7.4%), and other mutations (7.1%) were identified. Patients with oncogenic drivers who received PD-1 blockade plus chemotherapy had significantly better outcomes compared to those received PD-1 blockade or chemotherapy alone (ORR: 51% vs. 18% vs. 25%, P < 0.001; median PFS: 10.0 [95% CI: 8.9-12.6] vs. 3.7 [95% CI: 2.9-5.1] vs. 5.3 [95% CI: 4.5-6.2] months, P < 0.001; median OS: 26.0 [95% CI: 23.0-30.0] vs. 14.3 [95% CI: 9.6-19.8] vs. 16.1 [95% CI: 11.6-21.9] months, P < 0.001). The superior efficacy was consistently found in separate analyses for patients received first-line and second/third line treatments. Among individual gene alterations, patients with KRAS, EGFR, or BRAF mutations treated with PD-1 blockade plus chemotherapy achieved markedly improved PFS and OS than those received PD-1 blockade or chemotherapy alone. Multivariate Cox regression analysis revealed that PD-1 blockade plus chemotherapy was independently associated with better PFS and OS. CONCLUSION PD-1 blockade plus chemotherapy demonstrated superior efficacy than PD-1 blockade monotherapy or chemotherapy alone in patients with oncogenic-driven advanced NSCLC, particularly in KRAS, EGFR and BRAF subgroups. These findings suggest that PD-1 blockade plus chemotherapy may be considered as an optional treatment option for patients without available targeted therapies.
Collapse
Affiliation(s)
- Haowei Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Lei Cheng
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jian Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Peixin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Zhuoran Tang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Qianyi Wang
- Department of Medical Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China
| | - Ying Ma
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chao Zhao
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xuefei Li
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Fei Zhou
- Department of Medical Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China.
| | - Xiaoxia Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China.
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China.
- Department of Medical Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
18
|
Angelicola S, Giunchi F, Ruzzi F, Frascino M, Pitzalis M, Scalambra L, Semprini MS, Pittino OM, Cappello C, Siracusa I, Chillico IC, Di Noia M, Turato C, De Siervi S, Lescai F, Ciavattini T, Lopatriello G, Bertoli L, De Jonge H, Iamele L, Altimari A, Gruppioni E, Ardizzoni A, Rossato M, Gelsomino F, Lollini PL, Palladini A. PD-L1 and IFN-γ modulate Non-Small Cell Lung Cancer (NSCLC) cell plasticity associated to immune checkpoint inhibitor (ICI)-mediated hyperprogressive disease (HPD). J Transl Med 2025; 23:2. [PMID: 39748404 PMCID: PMC11697469 DOI: 10.1186/s12967-024-06023-8] [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: 08/03/2024] [Accepted: 12/22/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Non-Small Cell Lung Cancer (NSCLC) is the leading cause of cancer death worldwide. Although immune checkpoint inhibitors (ICIs) have shown remarkable clinical efficacy, they can also induce a paradoxical cancer acceleration, known as hyperprogressive disease (HPD), whose causative mechanisms are still unclear. METHODS This study investigated the mechanisms of ICI resistance in an HPD-NSCLC model. Two primary cell cultures were established from samples of a NSCLC patient, before ICI initiation ("baseline", NSCLC-B) and during HPD ("hyperprogression", NSCLC-H). The cell lines were phenotypically and molecularly characterized through immunofluorescence, Western Blotting and RNA-Seq analysis. To assess cell plasticity and aggressiveness, cellular growth patterns were evaluated both in vitro and in vivo through 2D and 3D cell growth assays and patient-derived xenografts establishment. In vitro investigations, including the evaluation of cell sensitivity to interferon-gamma (IFN-γ) and cell response to PD-L1 modulation, were conducted to explore the influence of these factors on cell plasticity regulation. RESULTS NSCLC-H exhibited increased expression of specific CD44 isoforms and a more aggressive phenotype, including organoid formation ability, compared to NSCLC-B. Plastic changes in NSCLC-H were well described by a deep transcriptome shift, that also affected IFN-γ-related genes, including PD-L1. IFN-γ-mediated cell growth inhibition was compromised in both 2D-cultured NSCLC-B and NSCLC-H cells. Further, the cytokine induced a partial activation of both type I and type II IFN-pathway mediators, together with a striking increase in NSCLC-B growth in 3D cell culture systems. Finally, low IFN-γ doses and PD-L1 modulation both promoted plastic changes in NSCLC-B, increasing CD44 expression and its ability to produce spheres. CONCLUSIONS Our findings identified plasticity as a relevant hallmark of ICI-mediated HPD by demonstrating that ICIs can modulate the IFN-γ and PD-L1 pathways, driving tumor cell plasticity and fueling HPD development.
Collapse
Affiliation(s)
- Stefania Angelicola
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Francesca Giunchi
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesca Ruzzi
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | | | - Mary Pitzalis
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Laura Scalambra
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Maria Sofia Semprini
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Olga Maria Pittino
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Chiara Cappello
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Irene Siracusa
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | | | - Martina Di Noia
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Cristian Turato
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Silvia De Siervi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Francesco Lescai
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | | | | | - Luca Bertoli
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Hugo De Jonge
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Luisa Iamele
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Annalisa Altimari
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Elisa Gruppioni
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Andrea Ardizzoni
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Marzia Rossato
- Department of Biotechnology, University of Verona, Verona, Italy
- Genartis S.R.L., Verona, Italy
| | - Francesco Gelsomino
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | - Pier-Luigi Lollini
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Arianna Palladini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.
- Unità Operativa di Oncologia, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| |
Collapse
|
19
|
Ren XH, Guo T, Xu MF, Huang Y, Liao XR, Qi LJ, Cheng SX. A Multiple Targeting Genome Editing System for Remodulation of Circulating Malignant Cells to Eliminate Cancer Immunosuppression and Restore Immune Responses. Adv Healthc Mater 2025; 14:e2401223. [PMID: 39440615 DOI: 10.1002/adhm.202401223] [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: 05/01/2024] [Revised: 09/29/2024] [Indexed: 10/25/2024]
Abstract
Cancer immunotherapy, which aims to eliminate cancer immunosuppression and reactivate anticancer immunity, holds great promise in oncology treatments. However, it is challenging to accurately study the efficacy of immunotherapy based on human-derived cells through animal experiments due to xenogeneic immune rejection. Herein, a personalized and precise strategy to evaluate the effectiveness of immunotherapy using the blood samples of cancer patients is presented. Through the utilization of multiple cancer-targeting delivery system decorated with the epidermal growth factor receptor (EGFR)-specific aptamer CL4 and the AXL-specific aptamer GL21.T to achieve superior efficiency in delivering the genome editing plasmid for MUC1 knockout, effective modulation on the behavior of circulating malignant cells (CMCs) is realized. After genome editing, both mucin 1 (MUC1) and programmed death-ligand 1 (PD-L1) are significantly downregulated in CMCs. The elimination of immunosuppression results in markedly enhanced secretion of pro-inflammatory anticancer cytokines encompassing interleukins 2, 12, and 15 and interferon-γ by immune cells. The study not only provides a strategy to overcome immunosuppression but also yields critical insights for personalized immunotherapy approaches.
Collapse
Affiliation(s)
- Xiao-He Ren
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230011, China
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Tao Guo
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei, Anhui, 230011, China
| | - Ma-Fei Xu
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230011, China
| | - Yun Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei, Anhui, 230011, China
| | - Xin-Ru Liao
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Li-Jin Qi
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Si-Xue Cheng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| |
Collapse
|
20
|
Balbi M, Mazzaschi G, Leo L, Moron Dalla Tor L, Milanese G, Marrocchio C, Silva M, Mura R, Favia P, Bocchialini G, Trentini F, Minari R, Ampollini L, Quaini F, Roti G, Tiseo M, Sverzellati N. Longitudinal Changes of CT-radiomic and Systemic Inflammatory Features Predict Survival in Advanced Non-Small Cell Lung Cancer Patients Treated With Immune Checkpoint Inhibitors. J Thorac Imaging 2025; 40:00005382-990000000-00147. [PMID: 39188157 PMCID: PMC11654449 DOI: 10.1097/rti.0000000000000801] [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: 08/28/2024]
Abstract
PURPOSE This study aims to determine whether longitudinal changes in CT radiomic features (RFs) and systemic inflammatory indices outperform single-time-point assessment in predicting survival in advanced non-small cell lung cancer (NSCLC) treated with immune checkpoint inhibitors (ICIs). MATERIALS AND METHODS We retrospectively acquired pretreatment (T0) and first disease assessment (T1) RFs and systemic inflammatory indices from a single-center cohort of stage IV NSCLC patients and computed their delta (Δ) variation as [(T1-T0)/T0]. RFs from the primary tumor were selected for building baseline-radiomic (RAD) and Δ-RAD scores using the linear combination of standardized predictors detected by LASSO Cox regression models. Cox models were generated using clinical features alone or combined with baseline and Δ blood parameters and integrated with baseline-RAD and Δ-RAD. All models were 3-fold cross-validated. A prognostic index (PI) of each model was tested to stratify overall survival (OS) through Kaplan-Meier analysis. RESULTS We included 90 ICI-treated NSCLC patients (median age 70 y [IQR=42 to 85], 63 males). Δ-RAD outperformed baseline-RAD for predicting OS [c-index: 0.632 (95%CI: 0.628 to 0.636) vs. 0.605 (95%CI: 0.601 to 0.608) in the test splits]. Integrating longitudinal changes of systemic inflammatory indices and Δ-RAD with clinical data led to the best model performance [Integrated-Δ model, c-index: 0.750 (95% CI: 0.749 to 0.751) in training and 0.718 (95% CI: 0.715 to 0.721) in testing splits]. PI enabled significant OS stratification within all the models ( P -value <0.01), reaching the greatest discriminative ability in Δ models (high-risk group HR up to 7.37, 95% CI: 3.9 to 13.94, P <0.01). CONCLUSION Δ-RAD improved OS prediction compared with single-time-point radiomic in advanced ICI-treated NSCLC. Integrating Δ-RAD with a longitudinal assessment of clinical and laboratory data further improved the prognostic performance.
Collapse
Affiliation(s)
- Maurizio Balbi
- Unit of Scienze Radiologiche
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Giulia Mazzaschi
- Medical Oncology Unit
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Ludovica Leo
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Gianluca Milanese
- Unit of Scienze Radiologiche
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Cristina Marrocchio
- Unit of Scienze Radiologiche
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Mario Silva
- Unit of Scienze Radiologiche
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Rebecca Mura
- Unit of Scienze Radiologiche
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Pasquale Favia
- Unit of Scienze Radiologiche
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Francesca Trentini
- Medical Oncology Unit
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Luca Ampollini
- Thoracic Surgery Unit, University Hospital of Parma
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Federico Quaini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Giovanni Roti
- Translational Hematology Unit
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Marcello Tiseo
- Medical Oncology Unit
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Nicola Sverzellati
- Unit of Scienze Radiologiche
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
21
|
Su Z, Guan M, Zhang L, Lian X. Factors associated with immune‑related severe adverse events (Review). Mol Clin Oncol 2025; 22:3. [PMID: 39563998 PMCID: PMC11574706 DOI: 10.3892/mco.2024.2798] [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: 07/31/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) are frequently used in cancer treatment. Despite their clinical benefits, they can also cause a wide range of immune-related adverse events (ir-AEs). The overall incidence of irAEs in cancer patients treated with immunotherapy ranges from 70-90%, while that of immune-related severe adverse events (ir-SAEs) is 10-43%. ir-SAEs pose a significant risk to patient safety as they are extremely frequent and lethal. Due to non-specific manifestations, rapid progression and significant morbidity, it is essential to identify factors associated with ir-SAEs early to predict high-risk groups for treatment safety. However, less information is available on the factors causing ir-SAEs, and further research is needed. The present study reviews the factors associated with ir-SAEs in terms of demographic characteristics, disease-related information and laboratory examinations to provide a clinical reference. In terms of demographic characteristics, age, body mass index, smoking, ethnicity and cancer family history may influence the incidence of ir-SAEs. Regarding disease-related information, the risks factors associated with ir-SAEs may include disease history, treatment regimen and cancer type. For laboratory examinations, risk factors associated with ir-SAEs include the laboratory examination parameters of peripheral blood cells, immunocytes, cytokines/chemokines, genetics, gut microbia, proteins and brain injury markers. All of these risk factors can stimulate the body's inflammatory response, leading to over proliferation of T cells and other inflammatory factors. In addition, the use of ICIs may disrupt gut microbial homeostasis and dysregulate the pre-existing intestinal ecology, which may therefore trigger inflammatory signaling pathways, affect overall immune function and increase the occurrence of ir-SAEs. In response to the aforementioned risk factors, it is recommended that medical professionals incorporate their analysis into routine patient testing for early identification of patient ir-SAEs and to create early individualized interventions to improve the safety for immunotherapy patients.
Collapse
Affiliation(s)
- Zhenzhen Su
- School of Nursing, Peking University, Beijing 100191, P.R. China
| | - Miaomiao Guan
- School of Nursing, Peking University, Beijing 100191, P.R. China
| | - Liyan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Xuemin Lian
- School of Nursing, Peking University, Beijing 100191, P.R. China
- Department of Health and Medical, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
22
|
Huo Y, Wang D, Yang S, Xu Y, Qin G, Zhao C, Lei Q, Zhao Q, Liu Y, Guo K, Ouyang S, Sun T, Wang H, Fan F, Han N, Liu H, Chen H, Miao L, Liu L, Duan Y, Lv W, Liu L, Zhang Z, Cang S, Wang L, Zhang Y. Optimal timing of anti-PD-1 antibody combined with chemotherapy administration in patients with NSCLC. J Immunother Cancer 2024; 12:e009627. [PMID: 39706602 PMCID: PMC11667274 DOI: 10.1136/jitc-2024-009627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 11/17/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Anti-programmed cell death 1 (PD-1) antibody combined with chemotherapy simultaneously is regarded as the standard treatment for patients with advanced non-small cell lung cancer (NSCLC) by current clinical guidelines. Different immune statuses induced by chemotherapy considerably affect the synergistic effects of the chemo-anti-PD-1 combination. Therefore, it is necessary to determine the optimal timing of combination treatment administration. METHODS The dynamic immune status induced by chemotherapy was observed in paired peripheral blood samples of patients with NSCLC using flow cytometry and RNA sequencing. Ex vivo studies and metastatic lung carcinoma mouse models were used to evaluate immune activity and explore the optimal combination timing. A multicenter prospective clinical study of 170 patients with advanced NSCLC was performed to assess clinical responses, and systemic immunity was assessed using omics approaches. RESULTS PD-1 expression on CD8+ T cells was downregulated on day 1 (D1) and D2, but recovered on D3 after chemotherapy administration, which is regulated by the calcium influx-P65 signaling pathway. Programmed cell death 1 ligand 1 expression in myeloid-derived suppressor cells was markedly reduced on D3. RNA sequencing analysis showed that T-cell function began to gradually recover on D3 rather than on D1. In addition, ex vivo and in vivo studies have shown that anti-PD-1 treatment on D3 after chemotherapy may enhance the antitumor response and considerably inhibit tumor growth. Finally, in clinical practice, a 3-day-delay sequential combination enhanced the objective response rate (ORR, 68%) and disease control rate (DCR, 98%) compared with the simultaneous combination (ORR=37%; DCR=81%), and prolonged progression-free survival to a greater extent than the simultaneous combination. The new T-cell receptor clones were effectively expanded, and CD8+ T-cell activity was similarly recovered. CONCLUSIONS A 3-day-delay sequential combination might increase antitumor responses and clinical benefits compared with the simultaneous combination.
Collapse
Affiliation(s)
- Yachang Huo
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dan Wang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuangning Yang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yujie Xu
- Department of Oncology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Guohui Qin
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chenhui Zhao
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qingyang Lei
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qitai Zhao
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yaqing Liu
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Kaiyuan Guo
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Songyun Ouyang
- Department of Respiratory and Critical Care Sleep Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ting Sun
- Department of Respiratory and Critical Care Sleep Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongmin Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feifei Fan
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Na Han
- Department of Oncology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hong Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongjie Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lijun Miao
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuqing Duan
- Department of Tumor Immunotherapy, the Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, Hebei, China
| | - Wei Lv
- Department of Tumor Immunotherapy, the Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, Hebei, China
| | - Lihua Liu
- Department of Tumor Immunotherapy, the Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, Hebei, China
| | - Zhixin Zhang
- Department of Technology, Chengdu ExAb Biotechnology Ltd, Chengdu, Sichuan, China
| | - Shundong Cang
- Department of Oncology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Liping Wang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- School of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
23
|
Feng D, Jiang H, Chen G, Guan W, Yi L, Zhu Y, Li Y, Huang G, He B, Tang J, Tang Y, Zeng J, Zhou W, Shi J, Xie Z, Liu M, Xie X, Lin X, Zhou C. Case report: Therapeutic response of front-line cadonilimab plus chemotherapy on patient with advanced lung adenocarcinoma harboring STK11 genetic aberration. Front Immunol 2024; 15:1485358. [PMID: 39717770 PMCID: PMC11663897 DOI: 10.3389/fimmu.2024.1485358] [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: 08/23/2024] [Accepted: 11/18/2024] [Indexed: 12/25/2024] Open
Abstract
The STK11 gene mutation is a common genetic alteration in non-small cell lung cancer (NSCLC) and is significantly associated with poor responses to current immunotherapy regimens. Despite its prevalence, there is currently no established standard for front-line treatment in this subtype of NSCLC, underscoring the increasing need for personalized therapeutic strategies. In this report, we present a case of a patient with STK11-mutant NSCLC who was treated with first-line cadonilimab (10mg/kg) in combination with pemetrexed (500mg/m^2) plus carboplatin (AUC=5), resulting in a notable extension of progression-free survival (PFS). This case highlights the potential efficacy and feasibility of combining immunotherapy with chemotherapy in patients with STK11-mutant NSCLC. Additionally, we provide a review of recent advancements in research related to STK11 mutations in lung cancer as reported in the literature.
Collapse
Affiliation(s)
- Du Feng
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Nanshan School, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Huixin Jiang
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Gengjia Chen
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Nanshan School, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wenhui Guan
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lin Yi
- Nanshan School, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yue Zhu
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yijia Li
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Gengda Huang
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Bin He
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Junlong Tang
- The First School of Clinical Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yujie Tang
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiyuan Zeng
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wensheng Zhou
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiayu Shi
- Department of Stomatology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhanhong Xie
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ming Liu
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaohong Xie
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xinqing Lin
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chengzhi Zhou
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
24
|
Liu M, Li Q, Meng X, Cui Y, Sun W, Wang H, Gao Q. Identification of gene signatures relevant to the efficacy of immune checkpoint inhibitors in non-small cell lung cancer. Medicine (Baltimore) 2024; 103:e40569. [PMID: 39654181 PMCID: PMC11630944 DOI: 10.1097/md.0000000000040569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/27/2024] [Accepted: 10/30/2024] [Indexed: 12/12/2024] Open
Abstract
Despite significant advancements in the treatment of non-small cell lung cancer (NSCLC) through immunotherapy, many patients still exhibit resistance to this approach. This study aims to identify the characteristics of individuals who can benefit from immunotherapy, especially immune checkpoint inhibitors (ICIs), and to investigate optimal strategies for patients who experience resistance to it. Data on gene expression patterns and clinical information from NSCLC patients who underwent immunotherapy were obtained from the Gene Expression Omnibus databases. A predictive signature for immunotherapy prognosis was developed using a training dataset and validated with validation datasets. Immune landscape and immunotherapy responsiveness analyses were conducted to assess the risk signature. Additionally, data from a study on immunotherapy were used to evaluate the correlation between MNX1 mutation and the effectiveness of ICIs, including clinical data and whole exome sequencing data. We identified 7 genes in NSCLC using RNA-seq data that were significantly associated with the efficacy of immunotherapy. Based on these genes, a risk signature was created to predict the efficacy of ICIs. Patients in the low-risk group had better outcomes compared to those in the high-risk group after receiving ICIs. Additionally, our analysis of the immune landscape revealed a significant association between the high-risk signature and an immunosuppressive state. We also discovered an unexpected role of tumor-specific MNX1 and HOXD1 in suppressing the immune response against cancer. Notably, NSCLC patients with MNX1 mutations experienced prolonged progression-free survival. Furthermore, we identified several medications that exhibited increased sensitivity in patients with high MNX1 expression, with topoisomerase inhibitors showing the highest level of sensitivity. This could be a potential strategy to improve the efficacy of ICIs. The risk signature has demonstrated its effectiveness in forecasting the prognosis of NSCLC treated with ICIs, enabling better patient stratification and more accurate prediction of immunotherapy response. Moreover, MNX1 and HOXD1 have been identified as key molecules related to immunotherapy resistance. Inhibition of these molecules, combined with current ICIs, offers novel strategies for the management of NSCLC patients.
Collapse
Affiliation(s)
- Min Liu
- Department of General Medicine, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Qiao Li
- The Eighth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaohong Meng
- The Eighth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yanan Cui
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Weirong Sun
- Radiological Department, The Eighth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hongmei Wang
- Department of General Medicine, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Qingjun Gao
- Department of Thoracic Surgery, The People’s Hospital of Rushan City, Weihai, PR China
| |
Collapse
|
25
|
Xia Z, Hong J, Yu X, Ran Y, Xie H, Zhou Z, Zuo J, Chen T, Meng J, Yang J. Pembrolizumab versus sintilimab in patients with advanced NSCLC: a retrospective multicenter study with propensity-score matching analysis. Front Oncol 2024; 14:1422039. [PMID: 39703850 PMCID: PMC11655332 DOI: 10.3389/fonc.2024.1422039] [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: 04/23/2024] [Accepted: 10/30/2024] [Indexed: 12/21/2024] Open
Abstract
Background Programmed cell death protein 1 (PD-1) inhibitors are commonly used worldwide for the management of non-small cell lung cancer (NSCLC). However, it remains unclear whether pembrolizumab and sintilimab, two of the most widely used PD-1 inhibitors in China, have significantly different effects on patients with NSCLC. A multicenter retrospective cohort study was designed and implemented using propensity-score matching (PSM) analysis to compare the effectiveness and safety profiles of pembrolizumab and sintilimab in patients with advanced NSCLC undergoing comprehensive therapy. Methods A total of 225 patients who received comprehensive therapy including pembrolizumab (n = 127) or sintilimab (n = 98), from 1 January to 31 December 2020 and met the eligibility criteria were included. PSM analysis (1:1) was performed to balance potential baseline confounding factors. For both treatments, Kaplan-Meier analysis and Cox regression were used to compare 1-year progression-free survival (PFS), disease control rate (DCR), objective response rate (ORR), and rates of all adverse events (AEs). Results PSM analysis resulted in 63 matched pairs of patients. After PSM, the median PFS was 8.68 months in the sintilimab group and 9.46 months in the pembrolizumab group. The 1-year PFS showed no significant difference between the pembrolizumab and sintilimab groups before and after PSM (P = 0.873 and P = 0.574, respectively). Moreover, within the matched cohort, the pembrolizumab group had an ORR of 30.2% and a DCR of 84.1%, whereas the sintilimab group exhibited an ORR of 41.3% and a DCR of 88.9%. There were no significant differences in the ORR and DCR between the two groups (P = 0.248 and P = 0.629, respectively). The incidence of grade 3 or 4 treatment-related AEs was significantly higher in the pembrolizumab group than that in the sintilimab group (42.9% vs. 33.3%, P = 0.043). Multivariable Cox proportional hazards regression analysis indicated that the lines of treatment and regimens significantly influenced the PFS of patients (P <0.05). Conclusions This study demonstrated the similar effectiveness of sintilimab and pembrolizumab in the treatment of patients with advanced NSCLC, with sintilimab potentially displaying a superior clinical safety profile. Clinical trial registration https://www.medicalresearch.org.cn/, identifier MR4423000113.
Collapse
Affiliation(s)
- Zhengzheng Xia
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Juan Hong
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Xiangyang Yu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Yuhua Ran
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Huali Xie
- Department of Pharmacy, Hongkong University Shenzhen Hospital, Shenzhen, China
| | - Ziyuan Zhou
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Jing Zuo
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Tujia Chen
- Department of Pharmacy, Boai Hospital of Zhongshan, Zhongshan, China
| | - Jun Meng
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Jun Yang
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
26
|
Jiang H, Ye Y, Wang M, Sun X, Sun T, Chen Y, Li P, Zhang M, Wang T. The progress on the relationship between gut microbiota and immune checkpoint blockade in tumors. Biotechnol Genet Eng Rev 2024; 40:4446-4465. [PMID: 37191003 DOI: 10.1080/02648725.2023.2212526] [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: 03/19/2023] [Accepted: 05/05/2023] [Indexed: 05/17/2023]
Abstract
Immune checkpoint blockade (ICB) has emerged as a promising immunotherapeutic approach for the treatment of various tumors. However, the efficacy of this therapy is limited in a subset of patients, and it is important to develop strategies to enhance immune responses. Studies have demonstrated a critical role of gut microbiota in regulating the therapeutic response to ICB. Gut microbiota composition, diversity, and function are mediated by metabolites, such as short-chain fatty acids and secondary bile acids, that interact with host immune cells through specific receptors. In addition, gut bacteria may translocate to the tumor site and stimulate antitumor immune responses. Therefore, maintaining a healthy gut microbiota composition, for instance through avoiding the use of antibiotics or probiotic interventions, can be an effective approach to optimize ICB therapy. This review summarizes the current understanding of the microbiota-immunity interactions in the context of ICB therapy, and discusses potential clinical implications of these findings.
Collapse
Affiliation(s)
- Haili Jiang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yingquan Ye
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mingqi Wang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xin Sun
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ting Sun
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yang Chen
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ping Li
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mei Zhang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ting Wang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
27
|
Chen S, He Z, Li M, Weng L, Lin J. Efficacy and safety of metronomic oral vinorelbine and its combination therapy as second- and later-line regimens for advanced non-small-cell lung cancer: a retrospective analysis. Clin Transl Oncol 2024; 26:3202-3210. [PMID: 38851648 DOI: 10.1007/s12094-024-03543-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
OBJECTIVE This retrospective analysis aimed to evaluate the efficacy and adverse reactions of metronomic oral vinorelbine and its combination therapy as second- and later-line regimens for advanced non-small-cell lung cancer (NSCLC). METHODS NSCLC patients undergoing metronomic oral vinorelbine as second- and later-line regimens in Fujian Cancer Hospital from October 2018 to October 2022 were enrolled, and patients' demographic and clinical characteristics were collected. The efficacy and safety of metronomic oral vinorelbine monotherapy and its combination therapy regimens were compared. RESULTS Of 57 study subjects, 63.2% received third- and later-line therapy, with median progression-free survival (mPFS) of 4 months, overall response rate (ORR) of 10.5%, and disease control rate (DCR) of 80.7%. The incidence of therapy-related adverse events was 42.1%, and there was only one case presenting grades 3 and 4 adverse events (1.8%). Among driver gene-negative participants, vinorelbine combination therapy regimens achieved longer mPFS (4.6 vs. 1.2 months, hazards ratio = 0.11, P < 0.0001) and comparable toxicity in relative to metronomic oral vinorelbine, and metronomic oral vinorelbine combined with immune checkpoint inhibitors showed the highest response, with mPFS of 5.6 months (95% CI 4.8 to 6.4 months), ORR of 25%, and DCR of 81.3%. Among participants with gradual resistance to osimertinib, continuing osimertinib in combination with metronomic oral vinorelbine achieved mPFS of 6.3 months (95% CI 0.1 to 12.5 months) and DCR of 86.7%. CONCLUSION Metronomic oral vinorelbine and its combination therapy regimens are favorable options as second- and later-line therapy for advanced NSCLC patients, with acceptable efficacy and tolerable toxicity. Vinorelbine combination therapy regimens show higher efficacy and comparable toxicity in relative to metronomic oral vinorelbine, and metronomic oral vinorelbine may have a synergistic effect with immunotherapy and EGFR-TKI targeted therapy.
Collapse
Affiliation(s)
- ShiJie Chen
- Fujian Cancer Hospital, Clinical Oncology School of Fujian Medical University, Fuma Street, Jinan District, Fuzhou, 350014, China
| | - ZhiYong He
- Fujian Cancer Hospital, Clinical Oncology School of Fujian Medical University, Fuma Street, Jinan District, Fuzhou, 350014, China
| | - MeiFang Li
- Fujian Cancer Hospital, Clinical Oncology School of Fujian Medical University, Fuma Street, Jinan District, Fuzhou, 350014, China
| | - LiHong Weng
- Fujian Cancer Hospital, Clinical Oncology School of Fujian Medical University, Fuma Street, Jinan District, Fuzhou, 350014, China
| | - JingHui Lin
- Fujian Cancer Hospital, Clinical Oncology School of Fujian Medical University, Fuma Street, Jinan District, Fuzhou, 350014, China.
| |
Collapse
|
28
|
Xiao G, Tanzhu G, Gao X, Li L, Liu Z, Xia X, Zhou R. An immune scoring system predicts prognosis and immune characteristics in lung adenocarcinoma brain metastases by RNA sequencing. Acta Neuropathol Commun 2024; 12:181. [PMID: 39593098 PMCID: PMC11590409 DOI: 10.1186/s40478-024-01895-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Previous studies have reported that the tumor immune microenvironment (TIME) was associated with the prognosis of lung cancer patients and the efficacy of immunotherapy. However, given the significant challenges in obtaining specimens of brain metastases (BrMs), few studies explored the correlation between the TIME and the prognosis in patients with BrMs from lung adenocarcinoma (LUAD). METHODS Transcript profiling of archival formalin-fixed and paraffin-embedded specimens of BrMs from 70 LUAD patients with surgically resected BrMs was carried out using RNA sequencing. An immune scoring system, the green-yellow module score (GYMS), was developed to predict prognosis and immune characteristics in both BrMs and primary LUAD using Weighted Correlation Network analysis (WGCNA) and GSVA analysis. We comprehensively evaluated the immunological role of GYMS based on gene expression profile of LUAD BrMs by systematically correlating GYMS with immunological characteristics and immunotherapy responsiveness in the BrMs. Immunohistochemistry was applied for validation. RESULTS We found that the high-GYMS group had better clinical prognosis and inflamed immune landscape including high infiltrations of various immune cells, increased immunomodulatory expression, and enriched immune-related pathways by using RNA-seq and immunohistochemical analysis. Low-GYMS group presented a lacked immune infiltration characteristic. Besides, the high-GYMS group had lower TIDE score and higher T-cell inflamed score than low-GYMS group. The GYMS has been validated in independent BrMs cohorts and primary NSCLC cohort treated with anti-PD-1/PD-L1, showing strong reproducibility and stability in both primary LUAD and BrMs. In addition, we construct a GYMS-related risk signature for patients with LUAD BrMs to predict prognosis. CONCLUSIONS We identified two immune-related subtypes which used to estimate prognosis and immune characteristics and developed a reliable GYMS-related risk signature in LUAD BrMs. These results will enhance the understanding of the immune microenvironment in LUAD BrMs and lay the theoretical foundation for the development of personalized therapies for LUAD patients with BrMs.
Collapse
Affiliation(s)
- Gang Xiao
- Department of Radiation Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Guilong Tanzhu
- Department of Radiation Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xuan Gao
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- GenePlus-Shenzhen Clinical Laboratory, Shenzhen, 518122, China
| | - Lifeng Li
- Geneplus-Beijing, Beijing, 102205, China
| | - Zhiyuan Liu
- Department of Radiation Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | | | - Rongrong Zhou
- Department of Radiation Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, People's Republic of China.
| |
Collapse
|
29
|
Mazzaschi G, Perrone F, Maglietta G, Favari E, Verzè M, Pluchino M, Minari R, Pecci F, Gnetti L, Campanini N, Silini EM, De Filippo M, Maffezzoli M, Giudice GC, Testi I, Tiseo M, Quaini F, Buti S. Intersecting Blood Cytokines With Cholesterol Parameters to Profile Patients With Advanced Solid Tumors Receiving Immune Checkpoint Inhibitors. J Immunother 2024; 47:388-394. [PMID: 38989743 PMCID: PMC11441731 DOI: 10.1097/cji.0000000000000534] [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: 01/23/2024] [Accepted: 06/03/2024] [Indexed: 07/12/2024]
Abstract
The study investigated the relationship between serum proinflammatory cytokine levels, cholesterol metabolism, and clinical outcome in cancer patients undergoing immune checkpoint inhibitors (ICIs). Peripheral blood was collected before therapy from ICI-treated advanced cancer patients. We retrospectively assessed plasma total cholesterol (TC), ABCA1- and ABCG1-mediated cholesterol efflux (CE), passive diffusion (PD), cholesterol loading capacity (CLC), and serum IL-6, IL-10, and TNF-α. The association between blood cholesterol parameters and inflammatory cytokines and their effect on overall survival (OS), progression-free survival (PFS), and clinical benefit (CB) from ICIs were statistically assessed. Among 70 consecutively enrolled patients (nonsmall cell lung cancer: 94%; renal cell carcinoma: 6%), TC, CLC, and cholesterol PD resulted significantly higher in IL-6 low and IL-10 low cases ( P <0.05), whereas ABCA1-mediated CE was increased in IL-10 high patients ( P =0.018). Uni- and multivariable analysis revealed meaningfully longer OS and PFS in IL-6 low (HR 2.13 and 2.97, respectively) and IL-10 low (HR 3.17 and 2.62) groups. At univariate analysis all cholesterol-related indices significantly correlated with OS and PFS, whereas at multivariate only high PD was validated as a protection factor (OS, HR 0.75; PFS, HR 0.84). Finally, uni- and multivariable showed a statistically significant inverse association of CB with ABCG1-CE (OR 0.62), as with IL-6 (OR 0.13) and IL-10 (OR 0.10). In-depth characterization of the interplay between blood cholesterol metabolism and immune-inflammatory cytokines might provide novel insights into the complex relationship among cancer, inflammation, lipids profile, and response to immunotherapy.
Collapse
Affiliation(s)
- Giulia Mazzaschi
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Fabiana Perrone
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Giuseppe Maglietta
- Clinical and Epidemiological Research Unit, University Hospital of Parma, Parma, Italy
| | - Elda Favari
- Food and Drug Department, University of Parma, Parma, Italy
| | - Michela Verzè
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Monica Pluchino
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Roberta Minari
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Federica Pecci
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Letizia Gnetti
- Pathology Unit, University Hospital of Parma, Parma, Italy
| | | | - Enrico Maria Silini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Pathology Unit, University Hospital of Parma, Parma, Italy
| | - Massimo De Filippo
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Radiology Unit, University Hospital of Parma, Parma, Italy
| | - Michele Maffezzoli
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Giulia Claire Giudice
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Irene Testi
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Marcello Tiseo
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Federico Quaini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Sebastiano Buti
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
30
|
Yang Y, Li J, Wang Y, Luo L, Yao Y, Xie X. Prognostic value of the systemic immune-inflammation index in lung cancer patients receiving immune checkpoint inhibitors: A meta-analysis. PLoS One 2024; 19:e0312605. [PMID: 39485761 PMCID: PMC11530041 DOI: 10.1371/journal.pone.0312605] [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: 08/05/2024] [Accepted: 10/09/2024] [Indexed: 11/03/2024] Open
Abstract
PURPOSE To explore the association between the systemic immune-inflammation index (SII) score and prognosis in immune checkpoint inhibitor (ICI)-treated patients with lung cancer. METHODS PubMed, EMBASE, Web of Science, and CNKI databases were searched up to August 1, 2024. Progression-free survival (PFS) and overall survival (OS) were the primary outcomes queried. Hazard ratios (HRs) and 95% confidence intervals (CIs) were combined, and subgroup analysis was based on pathological type [non-small cell lung cancer (NSCLC) vs. small-cell lung cancer (SCLC)], lines of ICIs (first-line vs. second- or further-line), and combinations of other therapies (yes vs. no). RESULTS Twenty retrospective studies with 2424 participants were included. The pooled results demonstrated that an elevated SII was associated with poorer PFS (HR = 1.82, 95% CI: 1.49-2.21; P < 0.001) and OS (HR = 2.31, 95% CI: 1.73-3.09; P < 0.001) in lung cancer patients receiving ICIs. Subgroup analysis stratified by pathological type, lines of ICIs and combinations of other therapies for PFS and OS further revealed the predictive role of the SII in ICI-treated lung cancer patients. CONCLUSION Based on current evidence the SII is significantly related to prognosis and could serve as a reliable prognostic indicator in lung cancer patients receiving ICIs.
Collapse
Affiliation(s)
- Yanhui Yang
- Department of Thoracic Surgery, The First People’s Hospital of Neijiang, Neijiang Affiliated Hospital of Chongqing Medical University, Neijiang, Sichuan, P.R. China
| | - Ji Li
- Department of Thoracic Surgery, The First People’s Hospital of Neijiang, Neijiang Affiliated Hospital of Chongqing Medical University, Neijiang, Sichuan, P.R. China
| | - Yi Wang
- Department of Thoracic Surgery, The First People’s Hospital of Neijiang, Neijiang Affiliated Hospital of Chongqing Medical University, Neijiang, Sichuan, P.R. China
| | - Lei Luo
- Department of Thoracic Surgery, The First People’s Hospital of Neijiang, Neijiang Affiliated Hospital of Chongqing Medical University, Neijiang, Sichuan, P.R. China
| | - Yi Yao
- Department of Thoracic Surgery, The First People’s Hospital of Neijiang, Neijiang Affiliated Hospital of Chongqing Medical University, Neijiang, Sichuan, P.R. China
| | - Xiaoyang Xie
- Department of Thoracic Surgery, The First People’s Hospital of Neijiang, Neijiang Affiliated Hospital of Chongqing Medical University, Neijiang, Sichuan, P.R. China
| |
Collapse
|
31
|
Cai F, Li Y, Liu H, Luo J. Single-cell and Spatial Transcriptomic Analyses Implicate Formation of the Immunosuppressive Microenvironment during Breast Tumor Progression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1392-1401. [PMID: 39283254 DOI: 10.4049/jimmunol.2400025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 08/22/2024] [Indexed: 10/23/2024]
Abstract
Ductal carcinoma in situ and invasive ductal carcinoma represent two stages of breast cancer progression. A multitude of studies have shown that genomic instability increases during tumor development, as manifested by higher mutation and copy number variation rates. The advent of single-cell and spatial transcriptomics has enabled the investigation of the subtle differences in cellular states during the tumor progression at single-cell level, thereby providing more nuanced understanding of the intercellular interactions within the solid tumor. However, the evolutionary trajectory of tumor cells and the establishment of the immunosuppressive microenvironment during breast cancer progression remain unclear. In this study, we performed an exploratory analysis of the single-cell sequencing dataset of 13 ductal carcinoma in situ and invasive ductal carcinoma samples. We revealed that tumor cells became more malignant and aggressive during their progression, and T cells transited to an exhausted state. The tumor cells expressed various coinhibitory ligands that interacted with the receptors of immune cells to create an immunosuppressive tumor microenvironment. Furthermore, spatial transcriptomics data confirmed the spatial colocalization of tumor and immune cells, as well as the expression of the coinhibitory ligand-receptor pairs. Our analysis provides insights into the cellular and molecular mechanism underlying the formation of the immunosuppressive landscape during two typical stages of breast cancer progression.
Collapse
MESH Headings
- Humans
- Tumor Microenvironment/immunology
- Tumor Microenvironment/genetics
- Breast Neoplasms/immunology
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Female
- Single-Cell Analysis
- Disease Progression
- Transcriptome
- Gene Expression Profiling
- Carcinoma, Intraductal, Noninfiltrating/immunology
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Carcinoma, Ductal, Breast/immunology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/pathology
- Gene Expression Regulation, Neoplastic/immunology
Collapse
Affiliation(s)
- Fengfeng Cai
- Department of Breast Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai China
| | - YuanYuan Li
- College of Computer and Information Engineering, Nanjing Tech University, Nanjing, Jiangsu, China
| | - Hui Liu
- College of Computer and Information Engineering, Nanjing Tech University, Nanjing, Jiangsu, China
| | - Judong Luo
- Department of Radiotherapy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
32
|
Teng F, Ju X, Gao Z, Xu J, Li Y, Wang Y, Zou B, Yu J. Perioperative immunotherapy for patients with EGFR mutant non-small cell lung cancer: Unexpected potential benefits. Biochim Biophys Acta Rev Cancer 2024; 1879:189194. [PMID: 39413856 DOI: 10.1016/j.bbcan.2024.189194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/01/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024]
Abstract
Given that immunotherapy has resulted in a significant overall survival (OS) benefit in advanced-stage disease, it is of notable interest to determine the effectiveness of these agents in early-stage non-small cell lung cancer (NSCLC). The potential exists for the immunotherapeutic approach in early-stage NSCLC to mirror the paradigm seen in advanced NSCLC, wherein survival enhancements have notably benefited the majority of patients. However, their performance in early-stage epidermal growth factor receptor (EGFR) mutant NSCLC is controversial. In the limited studies that included patients with EGFR mutation status, we found unexpected, good survival benefits of perioperative immune checkpoint inhibitors (ICIs) in resectable EGFR-positive NSCLC, which is controversial with those in advanced EGFR-mutant NSCLC. It is possible because of the shift toward immunosuppression that the immune environment undergoes during tumor progression. In the early disease stages, the anti-tumor immune response can be activated with fewer hindrances. In the context of EGFR mutant tumors, intratumor genetic heterogeneity can generate treatment-sensitive and -resistant subclones. The subclonality of the resistant subclone is pivotal in therapy response, with tyrosine kinase inhibitors (TKIs) selectively controlling EGFR-mutant cell proliferation and "competitive release" potentially explaining lower pathological responses in adjuvant TKIs trials. This review delves into emerging data on perioperative treatment modalities for early-stage EGFR mutant NSCLC, exploring unique mechanisms and predictive biomarkers to guide perioperative management strategies.
Collapse
Affiliation(s)
- Feifei Teng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Xiao Ju
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zhenhua Gao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Junhao Xu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yikun Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yungang Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Bingwen Zou
- Department of Radiation Oncology, West China Hospital of Sichuan University, Sichuan, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
33
|
Liu SY, Erazo T, Jee J, Arfe A, Gupta A, Pike LRG, Santini FC, Daly B, Schoenfeld A, Eichholz J, Johnson K, Martinez A, Sui J, Riaz N, Chang J, Yang SR, Travis W, Arcila ME, Guo J, Gagne E, Garg K, Baehner F, Lee NY, Drilon A, Kris MG, Scher HI, Razavi P, Gomez DR, Jones DR, Rudin CM, Chandarlapaty S, Isbell JM, Li BT. Optimal systemic treatment and real-world clinical application of ctDNA in patients with metastatic HER2-mutant lung cancer. Eur J Cancer 2024; 210:114257. [PMID: 39151324 DOI: 10.1016/j.ejca.2024.114257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 08/19/2024]
Abstract
INTRODUCTION No definitive answers currently exist regarding optimal first-line therapy for HER2-mutant NSCLC. Access to rapid tissue sequencing is a major barrier to precision drug development in the first-line setting. ctDNA analysis has the potential to overcome these obstacles and guide treatment. METHODS We retrospectively analyzed patients with metastatic HER2-mutant NSCLC who underwent prospective clinical ctDNA sequencing and received systemic therapy at Memorial Sloan Kettering Cancer Center (MSK) from January 2016 to September 2022. HER2 mutations were identified by next-generation sequencing through MSK-IMPACT, MSK-ACCESS or Resolution ctDx LungTM assay. Primary endpoints were time to the next treatment (TTNT) and overall survival (OS). RESULTS Sixty-three patients were included in the primary analysis. Chemoimmunotherapy (33/63, 52.4 %) was the predominant first-line treatment with a median TTNT of 5.1 months (95 %CI 4.1 - 6.1) whereas 55.0 % (22/40) of patients who received second-line T-DXd obtained a median TTNT of 9.2 m (95 % CI, 0-22.2). Plasma ctDNA was tested before first-line therapy in 40 patients with a median OS of 28.0 months (95 % CI 21-34), in whom 31 patients (78.0 %) had detectable ctDNA. HER2 mutations were detected on ctDNA with a median turnaround time of 13 days, occasionally co-occurred with EGFR and MET alterations and were tracked longitudinally correlating with treatment response. Patients with detectable baseline ctDNA had significantly shorter OS (hazard ratio (HR), 5.25; 95 % CI, 1.2-23.9; p = 0.019). CONCLUSION Chemoimmunotherapy remains a major treatment option for metastatic HER2-mutant NSCLC. ctDNA can rapidly detect HER2 and co-mutations, and it has the potential to guide and monitor optimal first-line therapy. As a negative prognostic biomarker, detectable ctDNA at baseline would need to be taken into account for patient selection in future studies.
Collapse
Affiliation(s)
- Si-Yang Liu
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Chinese Thoracic Oncology Group, Guangzhou, China
| | - Tatiana Erazo
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Justin Jee
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrea Arfe
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Avantika Gupta
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Luke R G Pike
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fernando C Santini
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Bobby Daly
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Kaylie Johnson
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Jane Sui
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nadeem Riaz
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jason Chang
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Soo-Ryum Yang
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - William Travis
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria E Arcila
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jiannan Guo
- Resolution Bioscience, Exact Sciences, Kirkland, WA, USA
| | - Eric Gagne
- Resolution Bioscience, Exact Sciences, Kirkland, WA, USA
| | - Kavita Garg
- Resolution Bioscience, Exact Sciences, Kirkland, WA, USA
| | | | - Nancy Y Lee
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Mark G Kris
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Howard I Scher
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pedram Razavi
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel R Gomez
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David R Jones
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Charles M Rudin
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Sarat Chandarlapaty
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - James M Isbell
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Bob T Li
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
34
|
Efil SC, Bilgin B, Ceylan F, Karakaş H, Karahan İ, Özsan SN, Kosku H, Yaman Ş, Bülent Akıncı M, Dede DŞ, Yalçın B, Nahit Şendur MA. A current comprehensive role of immune-checkpoint inhibitors in resectable non-small cell lung cancer: A narrative review. J Oncol Pharm Pract 2024; 30:1214-1239. [PMID: 38860323 DOI: 10.1177/10781552241260864] [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: 06/12/2024]
Abstract
OBJECTIVE The objective of this article is to review the efficacy, safety, and evidence for current use and potential future uses of immune-checkpoint inhibitors (ICIs) in the management of resectable non-small cell lung cancer (NSCLC). DATA SOURCES A literature review was carried out through PubMed to identify completed and ongoing clinical trials evaluating the use, efficacy, and safety of ICIs in the management of resectable NSCLC. DATA SUMMARY To date, four phase 3 trials have emerged that have changed our treatment practice concerning the utilization of ICIs during the adjuvant and neoadjuvant settings. The IMpower010 and KEYNOTE-091 trials examined the application of adjuvant atezolizumab and pembrolizumab, respectively, following surgical resection and adjuvant chemotherapy. In the CheckMate 816 trial, the combination of nivolumab and chemotherapy as a neoadjuvant therapy received approval for patients with resectable NSCLC. Also, for patients with resectable NSCLC, the use of a pembrolizumab and chemotherapy combination as a perioperative therapy received approval based on the results of the KEYNOTE-671 trial. Apart from these trials, there are numerous phase 2 and phase 3 trials, some of which have been published while others are still in progress. CONCLUSION Despite the promising outcomes from these trials there remain several unanswered questions. In this review, we will assess clinical trials involving adjuvant, neoadjuvant, and perioperative ICIs, aiming to address the unresolved questions related to these therapeutic approaches.
Collapse
Affiliation(s)
- Safa Can Efil
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, TR 06800, Turkey
| | - Burak Bilgin
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, TR 06800, Turkey
- Department of Medical Oncology, Ankara Yildirim Beyazit University Faculty of Medicine, Ankara, TR 06031, Turkey
| | - Furkan Ceylan
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, TR 06800, Turkey
| | - Hilal Karakaş
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, TR 06800, Turkey
| | - İrfan Karahan
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, TR 06800, Turkey
| | - Sema Nur Özsan
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, TR 06800, Turkey
| | - Hakan Kosku
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, TR 06800, Turkey
| | - Şebnem Yaman
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, TR 06800, Turkey
- Department of Medical Oncology, Ankara Yildirim Beyazit University Faculty of Medicine, Ankara, TR 06031, Turkey
| | - Muhammed Bülent Akıncı
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, TR 06800, Turkey
- Department of Medical Oncology, Ankara Yildirim Beyazit University Faculty of Medicine, Ankara, TR 06031, Turkey
| | - Didem Şener Dede
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, TR 06800, Turkey
- Department of Medical Oncology, Ankara Yildirim Beyazit University Faculty of Medicine, Ankara, TR 06031, Turkey
| | - Bülent Yalçın
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, TR 06800, Turkey
- Department of Medical Oncology, Ankara Yildirim Beyazit University Faculty of Medicine, Ankara, TR 06031, Turkey
| | - Mehmet Ali Nahit Şendur
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, TR 06800, Turkey
- Department of Medical Oncology, Ankara Yildirim Beyazit University Faculty of Medicine, Ankara, TR 06031, Turkey
| |
Collapse
|
35
|
Fang L, Meng Q, Wang J, Tu Y, Qu H, Diao Y, Li W, Wen H, Fang J, Hang L, Ma P, Jiang G. Multifunctional single-component photosensitizers as metal-free ferroptosis inducers for enhanced photodynamic immunotherapy. Acta Biomater 2024; 186:383-395. [PMID: 39069112 DOI: 10.1016/j.actbio.2024.07.034] [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: 01/19/2024] [Revised: 06/07/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
Immunotherapy can enhance primary tumor efficacy, restrict distant growth, and combat lung metastasis. Unfortunately, it remains challenging to effectively activate the immune response. Here, tertiary butyl, methoxy, and triphenylamine (TPA) were utilized as electron donors to develop multifunctional photosensitizers (PSs). CNTPA-TPA, featuring TPA as the donor (D) and cyano as the acceptor (A), excelled in reactive oxygen species (ROS) generation due to its smaller singlet-triplet energy gap (ΔES-T) and larger spin-orbit coupling constant (SOC). Additionally, cyano groups reacted with glutamate (Glu) and glutathione (GSH), reducing intracellular GSH levels. This not only enhanced PDT efficacy but also triggered redox dyshomeostasis-mediated ferroptosis. The positive effects of photodynamic therapy (PDT) and ferroptosis promoted immunogenic cell death (ICD) and immune activation. By further combining anti-programmed cell death protein ligand-1 (anti-PD-L1) antibody, the powerful treatments of ferroptosis-assisted photodynamic immunotherapy significantly eradicated the primary tumors, inhibited the growth of distant tumors, and suppressed lung metastasis. In this study, a three-pronged approach was realized by single-component CNTPA-TPA, which simultaneously served as metal-free ferroptosis inducers, type-I photosensitizers, and immunologic adjuvants for near-infrared fluorescence imaging (NIR FLI)-guided multimodal phototheranostics of tumor. STATEMENT OF SIGNIFICANCE: (1) CNTPA-TPA shared the smallest singlet-triplet energy gap and the largest spin-orbit coupling constant, which boosted intersystem crossing for efficient type-I photodynamic therapy (PDT); (2) Special reactions between cyano groups with glutamate and glutathione in mild conditions restricted the biosynthesis of intracellular GSH. GSH-depletion efficiently induced glutathione peroxidase 4 inactivation and lipid peroxide, resulting in ferroptosis of tumor cells; (3) The combination treatments of ferroptosis-assisted photodynamic immunotherapy induced by single-component CNTPA-TPA with the participation of anti-PD-L1 antibody resulted in increased T-cell infiltration and profound suppression of both primary and distant tumor growth, as well as lung metastasis.
Collapse
Affiliation(s)
- Laiping Fang
- Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Basic Medicine, School of Medicine, Jinan University, Xingangzhong Road 466, Guangzhou 518037, PR China
| | - Qi Meng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Renmin Street 5625, Changchun 130012, PR China
| | - Jizhuang Wang
- College of Chemistry and Materials Science, Jinan University, Huangpu Avenue West 601, Guangzhou 510632, PR China
| | - Yike Tu
- Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Department of Medical Imaging, Guangdong Second Provincial General Hospital, Xingangzhong Road 466, Guangzhou 518037, PR China
| | - Hong Qu
- Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Department of Medical Imaging, Guangdong Second Provincial General Hospital, Xingangzhong Road 466, Guangzhou 518037, PR China
| | - Yanzhao Diao
- Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Department of Medical Imaging, Guangdong Second Provincial General Hospital, Xingangzhong Road 466, Guangzhou 518037, PR China
| | - Wuming Li
- Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Department of Medical Imaging, Guangdong Second Provincial General Hospital, Xingangzhong Road 466, Guangzhou 518037, PR China
| | - Hua Wen
- Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Department of Medical Imaging, Guangdong Second Provincial General Hospital, Xingangzhong Road 466, Guangzhou 518037, PR China
| | - Jin Fang
- Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Department of Medical Imaging, Guangdong Second Provincial General Hospital, Xingangzhong Road 466, Guangzhou 518037, PR China
| | - Lifeng Hang
- Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Department of Medical Imaging, Guangdong Second Provincial General Hospital, Xingangzhong Road 466, Guangzhou 518037, PR China.
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Renmin Street 5625, Changchun 130012, PR China.
| | - Guihua Jiang
- Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Department of Medical Imaging, Guangdong Second Provincial General Hospital, Xingangzhong Road 466, Guangzhou 518037, PR China.
| |
Collapse
|
36
|
Zheng L, Hu F, Nie W, Lu J, Zhang B, Xu J, Wang S, Li Y, Zheng X, Zhang W, Shen Y, Zhong R, Chu T, Han B, Zhong H, Zhang X. The prognosis and metabolite changes of NSCLC patients receiving first-line immunotherapy combined chemotherapy in different M1c categories according to 9th edition of TNM classification. Cancer Med 2024; 13:e70223. [PMID: 39258530 PMCID: PMC11388058 DOI: 10.1002/cam4.70223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/12/2024] [Accepted: 09/01/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND The 9th edition of the TNM Classification for lung cancer delineates M1c into two subcategories: M1c1 (Multiple extrathoracic lesions within a single organ system) and M1c2 (Multiple extrathoracic lesions involving multiple organ systems). Existing research indicates that patients with lung cancer in stage M1c1 exhibit superior overall survival compared to those in stage M1c2. The primary frontline therapy for patients with advanced non-small cell lung cancer (NSCLC), lacking driver gene mutations, involves the use of immune checkpoint inhibitors (ICIs) combined with chemotherapy. Nevertheless, a dearth of evidence exists regarding potential survival disparities between NSCLC patients with M1c1 and M1c2 undergoing first-line immune-chemotherapy, and reliable biomarkers for predicting treatment outcomes are elusive. Serum metabolic profiles may elucidate distinct prognostic mechanisms, necessitating the identification of divergent metabolites in M1c1 and M1c2 undergoing combination therapy. This study seeks to scrutinize survival discrepancies between various metastatic patterns (M1c1 and M1c2) and pinpoint metabolites associated with treatment outcomes in NSCLC patients undergoing first-line ICIs combined with chemotherapy. METHOD In this study, 33 NSCLC patients lacking driver gene mutations diagnosed with M1c1, and 22 similarly diagnosed with M1c2 according to the 9th edition of TNM Classification, were enrolled. These patients received first-line PD-1 inhibitor plus chemotherapy. The relationship between metastatic patterns and progression-free survival (PFS) in patients undergoing combination therapy was analyzed using univariate and multivariate Cox regression models. Serum samples were obtained from all patients before treatment initiation for untargeted metabolomics analysis, aiming to identify differential metabolites. RESULTS In the univariate analysis of PFS, NSCLC patients in M1c1 receiving first-line PD-1 inhibitor plus chemotherapy exhibited an extended PFS (HR = 0.49, 95% CI, 0.27-0.88, p = 0.017). In multivariate PFS analyses, these M1c1 patients receiving first-line PD-1 inhibitor plus chemotherapy also demonstrated prolonged PFS (HR = 0.45, 95% CI, 0.22-0.92, p = 0.028). The serum metabolic profiles of M1c1 and M1c2 undergoing first-line PD-1 inhibitors plus chemotherapy displayed notable distinctions. In comparison to M1c1 patients, M1c2 patients exhibited alterations in various pathways pretreatment, including platelet activation, linoleic acid metabolism, and the VEGF signaling pathway. Diminished levels of lipid-associated metabolites (diacylglycerol, sphingomyelin) were correlated with adverse outcomes. CONCLUSION NSCLC patients in M1c1, devoid of driver gene mutations, receiving first-line PD-1 inhibitors combined with chemotherapy, experienced superior outcomes compared to M1c2 patients. Moreover, metabolomic profiles strongly correlated with the prognosis of these patients, and M1c2 patients with unfavorable outcomes manifested distinct changes in metabolic pathways before treatment. These changes predominantly involved alterations in lipid metabolism, such as decreased diacylglycerol and sphingomyelin, which may impact tumor migration and invasion.
Collapse
Affiliation(s)
- Liang Zheng
- Department of Respiratory and Critical Care Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fang Hu
- Department of Thoracic Medical OncologyThe Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital)ZhejiangChina
- Hangzhou Institute of Medicine (HlM)Chinese Academy of SciencesZhejiangChina
| | - Wei Nie
- Department of Respiratory and Critical Care Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jun Lu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bo Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jianlin Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shuyuan Wang
- Department of Respiratory and Critical Care Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ying Li
- Department of Respiratory and Critical Care Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiaoxuan Zheng
- Department of Respiratory and Critical Care Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wei Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yinchen Shen
- Department of Respiratory and Critical Care Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Runbo Zhong
- Department of Respiratory and Critical Care Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Tianqing Chu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Baohui Han
- Department of Respiratory and Critical Care Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hua Zhong
- Department of Respiratory and Critical Care Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xueyan Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
37
|
Liu J, Man Y, Gao J, Wang X, Zhang L, Li M, Yu J. Correlation between PD-L1 expression status and efficacy of immunotherapy as second-line or later-line therapy in advanced non-small cell lung cancer patients. Eur J Cancer Prev 2024; 33:448-460. [PMID: 38386588 DOI: 10.1097/cej.0000000000000880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
OBJECTIVE The objective of this study is to evaluate the correlation between tumor proportionality scores (TPS) and the effectiveness of immune checkpoint inhibitors (ICIs) as the second or subsequent line therapies for individuals who received diagnoses of advanced non-small cell lung cancer (NSCLC). METHODS The retrospective analysis was conducted on the medical records of a total of 143 patients who received diagnoses of stage IIIB/IV NSCLC and were admitted to our hospital from the beginning of 2019 to the end of September 2022. The follow-up period ended on 01 January 2023. The study used Kaplan-Meier survival curves to assess the progression-free survival (PFS) and overall survival (OS) of patients. Univariate and multivariate Cox proportional risk models were used to analyze the factors associated with the PFS and OS of advanced-stage NSCLC patients who received ICIs as the second or subsequent lines. RESULTS Patients diagnosed with NSCLC who had a TPS ≥1% and got treatment with ICIs exhibit notably elevated rates of partial response, objective response rate, disease control rate and extended PFS in comparison to NSCLC patients with a TPS of <1% ( P < 0.05). NSCLC patients with TPS within 1-49% [hazard ratio (HR) = 0.372; 95% confidence interval (CI), 0.140-0.993; P = 0.048] or ≥50% (HR = 0.276; 95% CI, 0.095-0.796; P = 0.017) were significantly associated with prolonged PFS, which were conducted by multivariate Cox regression analysis. CONCLUSION Programmed death protein-1 expression status may be predictive markers of the effectiveness of ICIs as the second or subsequent lines of therapies in advanced NSCLC are influenced by TPS.
Collapse
Affiliation(s)
- Jingya Liu
- Department of Medical Oncology, Beidahuang Industry Group General Hospital, Harbin
| | - Yingchun Man
- Department of Medical Oncology, Beidahuang Industry Group General Hospital, Harbin
| | - Jianing Gao
- Department of Urology, Daqing Oilfield General Hospital, Dqing, China
| | - Xinxin Wang
- Department of Medical Oncology, Beidahuang Industry Group General Hospital, Harbin
| | - Lijie Zhang
- Department of Medical Oncology, Beidahuang Industry Group General Hospital, Harbin
| | - Mingheng Li
- Department of Medical Oncology, Beidahuang Industry Group General Hospital, Harbin
| | - Jiahan Yu
- Department of Medical Oncology, Beidahuang Industry Group General Hospital, Harbin
| |
Collapse
|
38
|
Jiang T, Chen J, Wang H, Wu F, Chen X, Su C, Zhang H, Zhou F, Yang Y, Zhang J, Sun H, Zhang H, Zhou C, Ren S. Genomic correlates of the response to first-line PD-1 blockade plus chemotherapy in patients with advanced non-small-cell lung cancer. Chin Med J (Engl) 2024:00029330-990000000-01186. [PMID: 39164816 PMCID: PMC11407809 DOI: 10.1097/cm9.0000000000003094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND Programmed death 1 (PD-1) blockade plus chemotherapy has become the new first-line standard of care for patients with advanced non-small-cell lung cancer (NSCLC). Yet not all NSCLC patients benefit from this regimen. This study aimed to investigate the predictors of PD-1 blockade plus chemotherapy in untreated advanced NSCLC. METHODS We integrated clinical, genomic, and survival data from 287 patients with untreated advanced NSCLC who were enrolled in one of five registered phase 3 trials and received PD-1 blockade plus chemotherapy or chemotherapy alone. We randomly assigned these patients into a discovery cohort (n = 125), a validation cohort (n = 82), and a control cohort (n = 80). The candidate genes that could predict the response to PD-1 blockade plus chemotherapy were identified using data from the discovery cohort and their predictive values were then evaluated in the three cohorts. Immune deconvolution was conducted using transcriptome data of 1014 NSCLC patients from The Cancer Genome Atlas dataset. RESULTS A genomic variation signature, in which one or more of the 15 candidate genes were altered, was correlated with significantly inferior response rates and survival outcomes in patients treated with first-line PD-1 blockade plus chemotherapy in both discovery and validation cohorts. Its predictive value held in multivariate analyses when adjusted for baseline parameters, programmed cell death ligand 1 (PD-L1) expression level, and tumor mutation burden. Moreover, applying both the 15-gene panel and PD-L1 expression level produced better performance than either alone in predicting benefit from this treatment combination. Immune landscape analyses revealed that tumors with one or more variation in the 15-gene panel were associated with few immune infiltrates, indicating an immune-desert tumor microenvironment. CONCLUSION These findings indicate that a 15-gene panel can serve as a negative prediction biomarker for first-line PD-1 blockade plus chemotherapy in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Jian Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Haowei Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Fengying Wu
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Xiaoxia Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Chunxia Su
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Haiping Zhang
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Fei Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Ying Yang
- Genecast Biotechnology Co., Ltd, Wuxi, Jiangsu 214104, China
| | - Jiao Zhang
- Genecast Biotechnology Co., Ltd, Wuxi, Jiangsu 214104, China
| | - Huaibo Sun
- Genecast Biotechnology Co., Ltd, Wuxi, Jiangsu 214104, China
| | - Henghui Zhang
- Genecast Biotechnology Co., Ltd, Wuxi, Jiangsu 214104, China
- Biomedical Innovation Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; School of Oncology, Capital Medical University, Beijing 100038, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| |
Collapse
|
39
|
Horaguchi S, Nakahara Y, Igarashi Y, Kouro T, Wei F, Murotani K, Udagawa S, Higashijima N, Matsuo N, Murakami S, Kato T, Kondo T, Xiang H, Kasajima R, Himuro H, Tsuji K, Mano Y, Komahashi M, Miyagi Y, Saito H, Azuma K, Uehara S, Sasada T. Prognostic Significance of Plasma Neutrophil Extracellular Trap Levels in Patients with Non-Small Cell Lung Cancer Treated with Immune Checkpoint Inhibitors. Biomedicines 2024; 12:1831. [PMID: 39200295 PMCID: PMC11351864 DOI: 10.3390/biomedicines12081831] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 09/02/2024] Open
Abstract
Neutrophil extracellular traps (NETs) released from neutrophils are related to cancer progression. However, the relationship between the therapeutic effects of immune checkpoint inhibitors (ICIs) such as anti-PD-1 and anti-PD-L1 antibodies and plasma NET concentration in patients with non-small cell lung cancer (NSCLC) is poorly understood. In this study, concentrations of citrullinated histone H3 (CitH3), a surrogate marker of NETs, in plasma before/after treatment were examined in patients with advanced or recurrent NSCLC undergoing ICI treatment (n = 185). The clinical significances of NET levels before/after treatment and posttreatment changes were statistically evaluated. As a result, multivariate Cox analysis showed that high NET levels before treatment were statistically significant predictors of unfavorable overall survival (OS; p < 0.001, HR 1.702, 95% CI 1.356-2.137) and progression-free survival (PFS; p < 0.001, HR 1.566, 95% CI 1.323-1.855). The Kaplan-Meier curves showed significant separation between the high- and low-NET groups in OS (p = 0.002) and PFS (p < 0.001). Additionally, high NET levels after treatment were also significantly associated with worse OS (p < 0.001) and PFS (p < 0.001) by multivariate Cox analysis. Notably, the pretreatment NET levels were significantly correlated with the plasma levels of NET-related inflammatory cytokines, such as IL-6 and IL-8, and with NET-related gene expression and immune-suppressive profile in peripheral blood mononuclear cells. Our findings suggest that NETs released from activated neutrophils might reduce the clinical efficacy of ICIs in patients with NSCLC.
Collapse
Affiliation(s)
- Shun Horaguchi
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (S.H.); (T.K.); (F.W.); (H.H.); (K.T.); (Y.M.); (M.K.)
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Yoshiro Nakahara
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama 241-8515, Japan; (Y.N.); (S.M.); (T.K.); (T.K.); (H.S.)
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara 252-0375, Japan
| | - Yuka Igarashi
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
| | - Taku Kouro
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (S.H.); (T.K.); (F.W.); (H.H.); (K.T.); (Y.M.); (M.K.)
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
| | - Feifei Wei
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (S.H.); (T.K.); (F.W.); (H.H.); (K.T.); (Y.M.); (M.K.)
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
| | - Kenta Murotani
- Biostatistics Center, Kurume University School of Medicine, Kurume 830-0011, Japan;
| | - Seiichi Udagawa
- Mathematics Section, Division of Natural Sciences, Nihon University School of Medicine, Tokyo 173-0032, Japan;
| | - Naoko Higashijima
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
| | - Norikazu Matsuo
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Kurume 830-0011, Japan; (N.M.); (K.A.)
| | - Shuji Murakami
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama 241-8515, Japan; (Y.N.); (S.M.); (T.K.); (T.K.); (H.S.)
| | - Terufumi Kato
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama 241-8515, Japan; (Y.N.); (S.M.); (T.K.); (T.K.); (H.S.)
| | - Tetsuro Kondo
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama 241-8515, Japan; (Y.N.); (S.M.); (T.K.); (T.K.); (H.S.)
| | - Huihui Xiang
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (H.X.); (R.K.); (Y.M.)
| | - Rika Kasajima
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (H.X.); (R.K.); (Y.M.)
| | - Hidetomo Himuro
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (S.H.); (T.K.); (F.W.); (H.H.); (K.T.); (Y.M.); (M.K.)
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
| | - Kayoko Tsuji
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (S.H.); (T.K.); (F.W.); (H.H.); (K.T.); (Y.M.); (M.K.)
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
| | - Yasunobu Mano
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (S.H.); (T.K.); (F.W.); (H.H.); (K.T.); (Y.M.); (M.K.)
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
| | - Mitsuru Komahashi
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (S.H.); (T.K.); (F.W.); (H.H.); (K.T.); (Y.M.); (M.K.)
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (H.X.); (R.K.); (Y.M.)
| | - Haruhiro Saito
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama 241-8515, Japan; (Y.N.); (S.M.); (T.K.); (T.K.); (H.S.)
| | - Koichi Azuma
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Kurume 830-0011, Japan; (N.M.); (K.A.)
| | - Shuichiro Uehara
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Tetsuro Sasada
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (S.H.); (T.K.); (F.W.); (H.H.); (K.T.); (Y.M.); (M.K.)
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
| |
Collapse
|
40
|
Li X, Wu D, Tang J, Wu Y. The Efficiency and Safety of Triple-Drug Combination of Albumin-Bound Paclitaxel, Anlotinib and PD-1/L1 Inhibitors in the 2 nd or Above Line of Advanced NSCLC: A Retrospective Cohort Study. Cancer Manag Res 2024; 16:1003-1012. [PMID: 39135711 PMCID: PMC11318595 DOI: 10.2147/cmar.s472196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/02/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Existing research data indicates that albumin-bound paclitaxel (nab-ptx), anlotinib, and PD-1/L1 inhibitors have individually shown efficacy in second-line and subsequent treatments for advanced non-small cell lung cancer (NSCLC). This study seeks to investigate the potential of an optimized treatment regimen in this context by combining these three drugs and evaluating both efficacy and safety outcomes. PATIENTS AND METHODS Between January 2020 and January 2022, we collected data from pre-treated advanced NSCLC patients who received a combination therapy of nab-ptx, anlotinib, and PD-1/L1 inhibitors as a second-line or later treatment. The primary endpoints for the study included the objective response rate (ORR), progression-free survival (PFS), disease control rate (DCR) and overall survival (OS), while adverse events (AEs) were also recorded. RESULTS Our findings revealed that the ORR of this regimen in pretreated NSCLC patients was 35.71%, with mean PFS of 5.0 months and mean OS of 10.0 months. Further analysis suggested correlations between the efficacy of the regimen and factors such as PD-L1 expression levels, the occurrence of certain types of adverse events, and the status of NK cell activity. Additionally, the tolerable toxicity profile of this regimen indicates its potential applicability in the treatment of pretreated advanced NSCLC. CONCLUSION Our study displayed that triple-drug combination of nab-ptx, anlotinib and PD-1/L1 inhibitors showed promising efficiency and tolerated cytotoxicity in the 2nd or above line treatment of advanced NSCLC, indicating the potential of such regimen as an important option for second-line treatment of advanced NSCLC. However, due to limitations in patient numbers, its actual clinical value awaits further research confirmation.
Collapse
Affiliation(s)
- Xiaobing Li
- Department of Thoracic Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - De Wu
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jing Tang
- Department of Lymphoma, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yuebing Wu
- Department of Lymphoma, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
41
|
Zhong Q, Zhang L, Wu L, Zhao J, Sun J, Fang Y, Zhou J, Chu Q, Shen Y, Yang Z, Chen L, Huang M, Lin X, Liu Z, Shen P, Wang Z, Wang X, Wang H, Han C, Liu A, Zhang H, Ye F, Gao W, Wu F, Song Z, Chen S, Zhou C, Huang D, Zhang Q, Zheng X, Zheng X, Miao Q, Jiang K, Zou Z, Xu Y, Wu S, Wang H, Hong Y, Lu T, Li C, Huang C, Chen C, Lin G. Pretreatment pulmonary tumor necrosis is a promising prognostic imaging biomarker for first-line anti-PD-1/PD-L1 therapy in advanced lung squamous cell carcinoma: a multi-institutional, propensity score-matching cohort analysis. Ther Adv Med Oncol 2024; 16:17588359241266188. [PMID: 39108839 PMCID: PMC11301739 DOI: 10.1177/17588359241266188] [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: 12/12/2023] [Accepted: 06/18/2024] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Tumor necrosis (TN) is a common feature in lung squamous cell carcinoma (LSCC), which could provide useful predictive and prognostic information. OBJECTIVES This study aimed to investigate the effect of pretreatment pulmonary TN (PTN) on the prognosis of first-line anti-programmed cell death 1 (PD-1)/PD ligand 1 (PD-L1) inhibitor in advanced LSCC. DESIGN We conducted a retrospective study to analyze the association between the presence of PTN and clinical outcomes in advanced LSCC patients treated with anti-PD-1/PD-L1 inhibitors. METHODS Data from 240 eligible patients were collected from 27 hospitals across China between 2016 and 2020. The presence of PTN was assessed using contrast-enhanced chest computed tomography (CT) imaging at baseline. We utilized the Cox proportional-hazards regression model to analyze the association between PTN and clinical outcomes. In addition, to account for potential confounding factors and ensure comparability between groups, we employed propensity score-matching (PSM) analysis. RESULTS In the overall patient cohort, the presence of PTN was 39.6%. The median follow-up duration was 20.3 months. The positive PTN group exhibited a notably inferior median progression-free survival (PFS; 6.5 months vs 8.6 months, p = 0.012) compared to the negative PTN group. Within the Cox proportional-hazards regression model, PTN emerged as an independent predictor of unfavorable PFS (hazard ratio (HR) = 1.354, 95% confidence interval (CI): 1.002-1.830, p = 0.049). After PSM, the median PFS for the positive PTN group (6.5 months vs 8.0 months, p = 0.027) remained worse than that of the negative PTN group. Multivariate analyses also further underscored that the presence of PTN independently posed a risk for shorter PFS (HR = 1.494, 95% CI: 1.056-2.112, p = 0.023). However, no statistically significant difference in overall survival was observed between the two groups. CONCLUSION Our study suggests that the presence of PTN on baseline contrast-enhanced chest CT is a potential negative prognostic imaging biomarker for the outcome of anti-PD-1/PD-L1 inhibitor therapy in advanced LSCC. Further studies are warranted to validate these findings and explore the underlying mechanisms.
Collapse
Affiliation(s)
- Qiaofeng Zhong
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, China
- Interdisciplinary Institute for Medical Engineering, Fuzhou University, Fuzhou, China
| | - Longfeng Zhang
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Lin Wu
- The Second Department of Thoracic Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, China
| | - Jun Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jianguo Sun
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yong Fang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Zhejiang, China
| | - Jin Zhou
- School of Medicine, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yihong Shen
- Department of Respiratory Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zhenzhou Yang
- Department of Cancer Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Lijin Chen
- Department of Oncology, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, China
| | - Meijuan Huang
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyan Lin
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhenhua Liu
- Department of Medical Oncology, Provincial Clinical College, Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Peng Shen
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhijie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xin Wang
- Department of Oncology, Zhongshan Hospital of Xiamen University, Xiamen, China
| | - Huijuan Wang
- Department of Respiratory Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Chengbo Han
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Anwen Liu
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hongmei Zhang
- Department of Oncology, Xijing Hospital, Air Force Military Medical University, Xian, Shanxi, China
| | - Feng Ye
- Department of Medical Oncology, Cancer Hospital, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, China
| | - Wen Gao
- Department of Medical Oncology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fang Wu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhengbo Song
- Department of Clinical Trial, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Shengchi Chen
- Department of Oncology, Nanping First Hospital Affiliated to Fujian Medical University, Nanping, China
| | - Chengzhi Zhou
- Respiratory Medicine Department, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dingzhi Huang
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Qiuyu Zhang
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Xinlong Zheng
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Xiaobin Zheng
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Qian Miao
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Kan Jiang
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Zihua Zou
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Yiquan Xu
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Shiwen Wu
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Haibo Wang
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Yaping Hong
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Tao Lu
- Department of Radiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Chao Li
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Cheng Huang
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Chuanben Chen
- Department of Radiation, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuma Road No. 420, Fuzhou 350014, China
| | - Gen Lin
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou 350014, China
| |
Collapse
|
42
|
McGovern J, O'Rourke F, Will S, Nguyen HTN, Cranfield E, Maseland C, MacLeod N, Maclay JD, Laird BJ, Dolan RD, McMillan DC. The prevalence and prognostic value of systemic inflammation in good performance status patients with advanced, inoperable non-small cell lung cancer receiving palliative radiotherapy: Comparison of composite ratios and cumulative scores. Cancer Med 2024; 13:e70139. [PMID: 39164973 PMCID: PMC11335809 DOI: 10.1002/cam4.70139] [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: 12/10/2023] [Revised: 08/04/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024] Open
Abstract
INTRODUCTION The present study sought to examine the relationships between systemic inflammatory composite ratios/cumulative scores, magnitude of systemic inflammatory response (SIR) and survival in good performance status patients (ECOG-PS 0/1) with advanced NSCLC receiving palliative radiotherapy. METHODS Systemic inflammatory composite ratios/cumulative scores included the neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), lymphocyte-monocyte ratio (LMR), C-reactive protein, (CRP)-albumin ratio (CAR), neutrophil- lymphocyte score (NLS), platelet-lymphocyte score (PLS), lymphocyte-monocyte score (LMS), neutrophil-platelet score (NPS), modified Glasgow prognostic score (mGPS). The magnitude of SIR was determined by serum CRP concentration, with a median CRP concentration of >10 m mg/L considered to be systemically inflamed. Relationships between systemic inflammatory composite ratios/ cumulative scores and clinicopathological characteristics were examined using chi-square analysis. Relationships between overall survival (OS) and systemic inflammatory composite ratios/ cumulative scores were examined using cox regression analysis. RESULTS 479 patients were included. 48% (n = 231) of patients were male and 70% (n = 338) were ≥65 years of age. 29% (n = 140) patients were ECOG-PS 0 and 71% (n = 339) were ECOG-PS 1. 98% (n = 469) of patients died during follow-up. The median survival was 5 months (2-11). A similar prevalence of systemic inflammation was noted across the various ratios/scores (NLR >3 68%; LMR <2.4 65%; PLR >150 70%; CAR >0.20 83%; NLS ≥1 66%; LMS ≥1 71%; NPS≥1 50%; PLS≥1 60% and mGPS≥1 75%). Despite not considered to be systemically inflamed, an NLR <3, LMR ≥2.4, PLR ≤150, NLS 0, LMS 0, NPS 0 and PLS 0 all had a median CRP concentration of >10 mg/L. When adjusted for ECOG-PS, CAR>0.40 (p < 0.001) and mGPS 2 (p < 0.05) remained significantly associated with OS. CONCLUSION Liver-based measures of systemic inflammation (CAR and mGPS) appear more reliable for the quantification of the magnitude of SIR and have prognostic value in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Josh McGovern
- Academic Unit of SurgeryUniversity of GlasgowGlasgowUK
| | | | - Sarah Will
- Academic Unit of SurgeryUniversity of GlasgowGlasgowUK
| | | | | | | | - Nicholas MacLeod
- Department of OncologyBeatson West of Scotland Cancer CentreGlasgowUK
| | - John D. Maclay
- Department of Respiratory MedicineGlasgow Royal InfirmaryGlasgowUK
| | - Barry J. Laird
- Institute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - Ross D. Dolan
- Academic Unit of SurgeryUniversity of GlasgowGlasgowUK
| | | |
Collapse
|
43
|
Till JE, McDaniel L, Chang C, Long Q, Pfeiffer SM, Lyman JP, Padrón LJ, Maurer DM, Yu JX, Spencer CN, Gherardini PF, Da Silva DM, LaVallee TM, Abbott C, Chen RO, Boyle SM, Bhagwat N, Cannas S, Sagreiya H, Li W, Yee SS, Abdalla A, Wang Z, Yin M, Ballinger D, Wissel P, Eads J, Karasic T, Schneider C, O'Dwyer P, Teitelbaum U, Reiss KA, Rahma OE, Fisher GA, Ko AH, Wainberg ZA, Wolff RA, O'Reilly EM, O'Hara MH, Cabanski CR, Vonderheide RH, Carpenter EL. Circulating KRAS G12D but not G12V is associated with survival in metastatic pancreatic ductal adenocarcinoma. Nat Commun 2024; 15:5763. [PMID: 38982051 PMCID: PMC11233636 DOI: 10.1038/s41467-024-49915-5] [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: 12/05/2023] [Accepted: 06/18/2024] [Indexed: 07/11/2024] Open
Abstract
While high circulating tumor DNA (ctDNA) levels are associated with poor survival for multiple cancers, variant-specific differences in the association of ctDNA levels and survival have not been examined. Here we investigate KRAS ctDNA (ctKRAS) variant-specific associations with overall and progression-free survival (OS/PFS) in first-line metastatic pancreatic ductal adenocarcinoma (mPDAC) for patients receiving chemoimmunotherapy ("PRINCE", NCT03214250), and an independent cohort receiving standard of care (SOC) chemotherapy. For PRINCE, higher baseline plasma levels are associated with worse OS for ctKRAS G12D (log-rank p = 0.0010) but not G12V (p = 0.7101), even with adjustment for clinical covariates. Early, on-therapy clearance of G12D (p = 0.0002), but not G12V (p = 0.4058), strongly associates with OS for PRINCE. Similar results are obtained for the SOC cohort, and for PFS in both cohorts. These results suggest ctKRAS G12D but not G12V as a promising prognostic biomarker for mPDAC and that G12D clearance could also serve as an early biomarker of response.
Collapse
Affiliation(s)
- Jacob E Till
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | | | - Changgee Chang
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Qi Long
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | | | - Jaclyn P Lyman
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Lacey J Padrón
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Deena M Maurer
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Jia Xin Yu
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | | | - Diane M Da Silva
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | | | | | | | - Neha Bhagwat
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | - Samuele Cannas
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | - Hersh Sagreiya
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | - Wenrui Li
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | - Stephanie S Yee
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | - Aseel Abdalla
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | - Zhuoyang Wang
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | - Melinda Yin
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | - Dominique Ballinger
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | - Paul Wissel
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer Eads
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas Karasic
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | - Charles Schneider
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | - Peter O'Dwyer
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | - Ursina Teitelbaum
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | - Kim A Reiss
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Andrew H Ko
- University of California, San Francisco, San Francisco, CA, USA
| | - Zev A Wainberg
- University of California, Los Angeles, Los Angeles, CA, USA
| | - Robert A Wolff
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Mark H O'Hara
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Erica L Carpenter
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
44
|
Shu M, Huang L, Chen Y, Wang Y, Xie Z, Li S, Zhou J, Wei L, Fu T, Liu B, Chen H, Tang K, Ke Z. Identification of a DNA-methylome-based signature for prognosis prediction in driver gene-negative lung adenocarcinoma. Cancer Lett 2024; 593:216835. [PMID: 38548216 DOI: 10.1016/j.canlet.2024.216835] [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: 11/30/2023] [Revised: 03/22/2024] [Accepted: 03/22/2024] [Indexed: 06/01/2024]
Abstract
"Driver gene-negative" lung adenocarcinoma (LUAD) was of rare treatment options and a poor prognosis. Presently, for them, few biomarkers are available for stratification analysis to make appropriate treatment strategy. This study aimed to develop a DNA-methylome-based signature to realize the precise risk-stratifying. Here, an Illumina MethylationEPIC Beadchip was applied to obtain differentially methylated CpG sites (DMCs). A four-CpG-based signature, named as TLA, was successfully established, whose prognosis-predicting power was well verified in one internal (n = 78) and other external (n = 110) validation cohorts. Patients with high-risk scores had shorter overall survival (OS) in all cohorts [hazard ratio (HR): 11.79, 5.16 and 2.99, respectively]. Additionally, it can effectively divide patients into low-risk and high-risk groups, with significantly different OS in the diverse subgroups stratified by the standard clinical parameters. As an independent prognostic factor, TLA may assist in improving the nomogram's 5-year OS-predicting ability (AUC 0.756, 95% CI:0.695-0.816), superior to TNM alone (AUC 0.644, 95% CI: 0.590-0.698). Additionally, the relationship of TLA-related genes, TAC1, LHX9, and ALX1, with prognosis and tumour invasion made them serve as potential therapy targets for driver gene-negative LUAD.
Collapse
Affiliation(s)
- Man Shu
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Leilei Huang
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Yu Chen
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China; Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, PR China
| | - Yanxia Wang
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Zhongpeng Xie
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Shuhua Li
- Molecular Diagnosis and Gene Test Centre, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, PR China
| | - Jianwen Zhou
- Molecular Diagnosis and Gene Test Centre, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, PR China
| | - Lihong Wei
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Tongze Fu
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China; Molecular Diagnosis and Gene Test Centre, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, PR China
| | - Bixia Liu
- Molecular Diagnosis and Gene Test Centre, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, PR China
| | - Honglei Chen
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan, Hubei, PR China.
| | - Kejing Tang
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China; Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, PR China.
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China; Molecular Diagnosis and Gene Test Centre, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
45
|
Luo L, Jiang M, Wu H, Liu Y, Wang H, Zhou C, Ren S, Chen X, Jiang T, Xu C. SIRPG expression positively associates with an inflamed tumor microenvironment and response to PD-1 blockade. Cancer Immunol Immunother 2024; 73:147. [PMID: 38833156 PMCID: PMC11150346 DOI: 10.1007/s00262-024-03737-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: 03/21/2024] [Accepted: 05/15/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND This study aimed to investigate the relationship between signal regulatory protein gamma (SIRPG) and tumor immune microenvironment phenotypes or T cell mediated-adaptive antitumor immunity, and its predictive value for response to PD-1 blockade in cancers. METHODS Pan-cancer analysis of SIRPG expression and immune deconvolution was performed using transcriptomic data across 33 tumor types. Transcriptomic and clinical data from 157 patients with non-small-cell lung cancer (NSCLC) and melanoma received PD-1 blockade were analyzed. Expression characteristics of SIRPG were investigated using single-cell RNA sequencing (scRNA-seq) data of 103,599 cells. The effect of SIRPG expression was evaluated via SIRPG knockdown or overexpression in Jurkat T cells. RESULTS The results showed that most cancers with high SIRPG expression had significantly higher abundance of T cells, B cells, NK cells, M1 macrophages and cytotoxic lymphocytes and increased expression level of immunomodulatory factors regulating immune cell recruitment, antigen presentation, T cell activation and cytotoxicity, but markedly lower abundance of neutrophils, M2 macrophages, and myeloid-derived suppressor cells. High SIRPG expression was associated with favorable response to PD-1 blockade in both NSCLC and melanoma. scRNA-seq data suggested SIRPG was mainly expressed in CD8+ exhausted T and CD4+ regulatory T cells, and positively associated with immune checkpoint expression including PDCD1 and CTLA4. In vitro test showed SIRPG expression in T cells could facilitate expression of PDCD1 and CTLA4. CONCLUSION High SIRPG expression is associated with an inflamed immune phenotype in cancers and favorable response to PD-1 blockade, suggesting it would be a promising predictive biomarker for PD-1 blockade and novel immunotherapeutic target.
Collapse
Affiliation(s)
- Libo Luo
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Thoracic Cancer Institute, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Minlin Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Thoracic Cancer Institute, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Hong Wu
- Department of Oncology and Cancer Institute, Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32 1st Ring Road, Chengdu, 610072, China
| | - Yiqiang Liu
- Department of Oncology and Cancer Institute, Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32 1st Ring Road, Chengdu, 610072, China
| | - Haowei Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Thoracic Cancer Institute, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Thoracic Cancer Institute, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Thoracic Cancer Institute, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Xiaoxia Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Thoracic Cancer Institute, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China.
| | - Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Thoracic Cancer Institute, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China.
| | - Chuan Xu
- Department of Oncology and Cancer Institute, Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32 1st Ring Road, Chengdu, 610072, China.
| |
Collapse
|
46
|
Rosca OC, Vele OE. Microsatellite Instability, Mismatch Repair, and Tumor Mutation Burden in Lung Cancer. Surg Pathol Clin 2024; 17:295-305. [PMID: 38692812 DOI: 10.1016/j.path.2023.11.011] [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: 05/03/2024]
Abstract
Since US Food and Drug Administration approval of programmed death ligand 1 (PD-L1) as the first companion diagnostic for immune checkpoint inhibitors (ICIs) in non-small cell lung cancer, many patients have experienced increased overall survival. To improve selection of ICI responders versus nonresponders, microsatellite instability/mismatch repair deficiency (MSI/MMR) and tumor mutation burden (TMB) came into play. Clinical data show PD-L1, MSI/MMR, and TMB are independent predictive immunotherapy biomarkers. Harmonization of testing methodologies, optimization of assay design, and results analysis are ongoing. Future algorithms to determine immunotherapy eligibility might involve complementary use of current and novel biomarkers. Artificial intelligence could facilitate algorithm implementation to convert complex genetic data into recommendations for specific ICIs.
Collapse
Affiliation(s)
- Oana C Rosca
- Molecular Pathologist/Cytopathologist, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell; Department of Pathology and Laboratory Medicine, 2200 Northern Boulevard, Suite 104, Greenvale, NY 11548, USA.
| | - Oana E Vele
- Molecular Pathologist/Cytopathologist, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell; Department of Pathology and Laboratory Medicine, Lenox Hill Hospital, New York, NY 10075, USA
| |
Collapse
|
47
|
Dong C, Hui K, Gu J, Wang M, Hu C, Jiang X. Plasma sPD-L1 and VEGF levels are associated with the prognosis of NSCLC patients treated with combination immunotherapy. Anticancer Drugs 2024; 35:418-425. [PMID: 38386011 DOI: 10.1097/cad.0000000000001576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
The clinical significance of plasma soluble programmed cell death ligand 1 (sPD-L1) and vascular endothelial growth factor (VEGF) for non-small cell lung cancer (NSCLC) treated with the combination of anti-angiogenic therapy and anti-PD-L1 antibody (Ab) remain unknown. This study aimed to explore the association between plasma sPD-L1 and VEGF levels and the prognosis of NSCLC patients treated with the combination of Envafolimab and Endostar. Peripheral blood samples were collected from 24 NSCLC patients at baseline and after 6 weeks of treatment and were detected for sPD-L1 and VEGF levels. Both baseline and posttreatment sPD-L1 were significantly higher in progressive disease (PD) group than in controlled disease (CD) group (median: 77.5 pg/ml vs. 64.6 pg/ml, P = 0.036, median: 8451 pg/ml vs. 5563 pg/ml, P = 0.012). In multivariate analysis, lower baseline sPD-L1 levels were significantly associated with longer progression-free survival (PFS) (HR = 6.834, 95% CI: 1.350-34.592, P = 0.020). There were significantly higher posttreatment VEGF levels in PD group compared with CD group (median: 323.7 pg/ml vs. 178.5 pg/ml, P = 0.009). Higher posttreatment VEGF levels were significantly associated with shorter PFS in multivariate analysis (HR = 5.911, 95% CI: 1.391-25.122, P = 0.016). Plasma sPD-L1 and VEGF levels are associated with the clinical response and prognosis of NSCLC patients treated with the combination of PD-L1 inhibitors and anti-angiogenetic therapy.
Collapse
Affiliation(s)
- Changhong Dong
- Department of Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, Jiangsu Province, China
| | | | | | | | | | | |
Collapse
|
48
|
Yu X, You Z, Liu Y, Fang J, Zhao Q, Sun Z, Song Y, Liu J, Sun C. Sex-based immune microenvironmental feature heterogeneity in response to PD-1 blockade in combination with chemotherapy for patients with untreated advanced non-small-cell lung cancer. Cancer Med 2024; 13:e7423. [PMID: 38899854 PMCID: PMC11188036 DOI: 10.1002/cam4.7423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND To investigate the sex-based heterogeneity of immune microenvironmental feature and its impact on the response to first-line PD-1 blockade plus chemotherapy in patients with driver-negative advanced or metastatic non-small-cell lung cancer (NSCLC). PATIENTS AND METHODS A total of 439 patients with advanced NSCLC treated with first-line PD-1 blockade plus chemotherapy or chemotherapy were identified. Differences in clinical outcomes between female and male patients were determined using Kaplan-Meier curves. Neoantigen burden and five immune microenvironmental markers expression including PD-L1, CD4, CD8, FOXP3, and CD68 were compared between two groups. RESULTS Of 175 eligible patients, 89 received PD-1 blockade plus chemotherapy and 86 received first-line chemotherapy. Forty five were women (25.7%) and 130 were men (74.3%). Female patients received first-line PD-1 blockade in combination with chemotherapy had dramatically better ORR (85.2% vs. 53.2%; p = 0.009), PFS (23.7 vs. 7.3 months; p = 0.013), and OS (46.2 vs. 20.0 months; p = 0.004) than males. Treatment outcomes were similar between females and males in chemotherapy group. Multivariate analyses showed that sex was the independent prognostic factor for patients received PD-1 blockade combined with chemotherapy. Although female patients had significantly lower tumor mutational and neoantigen burden than males, pretreatment tumor tissues of female patients had markedly higher CD4, CD4/FOXP3, and CD4/FOXP3/PD-L1 expression level than male patients. CONCLUSIONS Female patients with untreated advanced or metastatic NSCLC would derive a larger benefit from PD-1 blockade in combination with chemotherapy than males. The biological significances of heterogeneity of tumor immune microenvironmental features between them need further investigation.
Collapse
Affiliation(s)
- Xiaofeng Yu
- Department of Thoracic SurgeryYantai Yuhuangding HospitalYantaiChina
| | - Zhaolei You
- Department of Thoracic SurgeryYantai Yuhuangding HospitalYantaiChina
| | - Ying Liu
- Department of Medical OncologyYantai Yuhuangding HospitalYantaiChina
| | - Jian Fang
- Department of Thoracic SurgeryYantai Yuhuangding HospitalYantaiChina
| | - Qi Zhao
- Department of Clinical LaboratoryYantai Yuhuangding HospitalYantaiChina
| | - Zhihong Sun
- Department of Clinical LaboratoryYantai Yuhuangding HospitalYantaiChina
| | - Yingjian Song
- Department of Thoracic SurgeryYantai Yuhuangding HospitalYantaiChina
| | - Jie Liu
- Department of Clinical LaboratoryYantai Yuhuangding HospitalYantaiChina
| | - Chengming Sun
- Department of Clinical LaboratoryYantai Yuhuangding HospitalYantaiChina
| |
Collapse
|
49
|
Xu J, Wang P, Li Y, Shi X, Yin T, Yu J, Teng F. Development and validation of an MRI-Based nomogram to predict the effectiveness of immunotherapy for brain metastasis in patients with non-small cell lung cancer. Front Immunol 2024; 15:1373330. [PMID: 38686383 PMCID: PMC11057328 DOI: 10.3389/fimmu.2024.1373330] [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: 01/19/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
Introduction The variability and unpredictability of immune checkpoint inhibitors (ICIs) in treating brain metastases (BMs) in patients with advanced non-small cell lung cancer (NSCLC) is the main concern. We assessed the utility of novel imaging biomarkers (radiomics) for discerning patients with NSCLC and BMs who would derive advantages from ICIs treatment. Methods Data clinical outcomes and pretreatment magnetic resonance images (MRI) were collected on patients with NSCLC with BMs treated with ICIs between June 2019 and June 2022 and divided into training and test sets. Metastatic brain lesions were contoured using ITK-SNAP software, and 3748 radiomic features capturing both intra- and peritumoral texture patterns were extracted. A clinical radiomic nomogram (CRN) was built to evaluate intracranial progression-free survival, progression-free survival, and overall survival. The prognostic value of the CRN was assessed by Kaplan-Meier survival analysis and log-rank tests. Results In the study, a total of 174 patients were included, and 122 and 52 were allocated to the training and validation sets correspondingly. The intratumoral radiomic signature, peritumoral radiomic signature, clinical signature, and CRN predicted intracranial objective response rate. Kaplan-Meier analyses showed a significantly longer intracranial progression-free survival in the low-CRN group than in the high-CRN group (p < 0.001). The CRN was also significantly associated with progression-free survival (p < 0.001) but not overall survival. Discussion Radiomics biomarkers from pretreatment MRI images were predictive of intracranial response. Pretreatment radiomics may allow the early prediction of benefits.
Collapse
Affiliation(s)
- Junhao Xu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Peiliang Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yikun Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaonan Shi
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Tianwen Yin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Feifei Teng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
50
|
Li GX, Chang RZ, Liu TT, Jin GN, Lu K, Yong TY, Li Z, Liu JH, Zhang B, Zhang WG, Ding ZY. GRIN2A mutation is a novel indicator of stratifying beneficiaries of immune checkpoint inhibitors in multiple cancers. Cancer Gene Ther 2024; 31:586-598. [PMID: 38267623 DOI: 10.1038/s41417-024-00730-6] [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: 03/25/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/26/2024]
Abstract
Glutamate-NMDAR receptors (GRINs) have been reported to influence cancer immunogenicity; however, the relationship between GRIN alterations and the response to immune checkpoint inhibitors (ICIs) has not been determined. This study combined clinical characteristics and mutational profiles from multiple cohorts to form a discovery cohort (n = 901). The aim of this study was to investigate the correlation between the mutation status of the GRIN gene and the response to ICI therapy. Additionally, an independent ICI-treated cohort from the Memorial Sloan Kettering Cancer Center (MSKCC, N = 1513) was used for validation. Furthermore, this study explored the associations between GRIN2A mutations and intrinsic and extrinsic immunity using multiomics analysis. In the discovery cohort, patients with GRIN2A-MUTs had improved clinical outcomes, as indicated by a higher objective response rate (ORR: 36.8% vs 25.8%, P = 0.020), durable clinical benefit (DCB: 55.2% vs 38.7%, P = 0.005), prolonged progression-free survival (PFS: HR = 0.65; 95% CI 0.49 to 0.87; P = 0.003), and increased overall survival (OS: HR = 0.67; 95% CI 0.50 to 0.89; P = 0.006). Similar results were observed in the validation cohort, in which GRIN2A-MUT patients exhibited a significant improvement in overall survival (HR = 0.66; 95% CI = 0.49 to 0.88; P = 0.005; adjusted P = 0.045). Moreover, patients with GRIN2A-MUTs exhibited an increase in tumor mutational burden, high expression of costimulatory molecules, increased activity of antigen-processing machinery, and infiltration of various immune cells. Additionally, gene sets associated with cell cycle regulation and the interferon response were enriched in GRIN2A-mutated tumors. In conclusion, GRIN2A mutation is a novel biomarker associated with a favorable response to ICIs in multiple cancers.
Collapse
Affiliation(s)
- Gan-Xun Li
- Hepatic Surgery Center, and Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Rui-Zhi Chang
- Hepatic Surgery Center, and Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tong-Tong Liu
- Department of Anesthesiology, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guan-Nan Jin
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Kan Lu
- Hepatic Surgery Center, and Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tu-Ying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430071, Hubei, China
| | - Zifu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430071, Hubei, China
| | - Ji-Hong Liu
- Department and Institute of Urology, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bixiang Zhang
- Hepatic Surgery Center, and Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Wan-Guang Zhang
- Hepatic Surgery Center, and Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Ze-Yang Ding
- Hepatic Surgery Center, and Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|