1
|
Zhu X, Yin G, Xu J, Tang X, Yu F. Dendrobium huoshanense polysaccharide inhibits NSCLC proliferation and immune evasion via FXR1-IL-35 axis signaling pathway. J Nat Med 2025:10.1007/s11418-025-01894-7. [PMID: 40259042 DOI: 10.1007/s11418-025-01894-7] [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: 05/14/2024] [Accepted: 03/10/2025] [Indexed: 04/23/2025]
Abstract
Dendrobium huoshanense has received special attention for its advantages in the treatment of lung cancer, but the underlying molecular mechanisms are not yet well understood. First, we obtained 8 active ingredients and 159 effective action targets of Dendrobium huoshanense using network pharmacology, and searching target interactions through STRING, constructing the PPI network and KEGG, GO and Hallmark enrichment analysis. Then, we combined target's enrichment analysis and GSEA enrichment analysis of IL-35, indicating the mechanism of cDHPs for non-small cell lung cancer (NSCLC) may be related to tight junction and NSCLC pathway. Further, FXR1 and ACTR3 were identified as core therapeutic targets, and high expression of FXR1 or ACTR3 was significantly associated with poor prognosis of patients. The analysis of single-cell data also indicated that the percentage of CD4-CTLA4-Treg cells may be increased by the expression of IL-35, resulting in a suppressive immune microenvironment. Next, In vivo experiment, we detected iTr35 by flow cytometry, detected IL-35 level by RT-PCR, Western blotting and ELISA, and detected NK cell activity to explore the immunomodulatory effects and anti-tumor mechanism of cDHPs. After cDHPs administration, the conversion of CD4+ T cells to iTr35 is inhibited, p35 and EBI3 in both protein and mRNA levels, the levels of IL-35 and IL-4 in serum decreased. The levels of IFN-γ, while the activity of NK cells in mice increased, enhancing the anti-tumor immune effect of the organism. Finally, analysis of sequencing data from the immunotherapy cohort of tumor-bearing mice obtained from the TISMO database shows that the combination of cDHPs and PD-1/PD-L1 antibodies improves effector and thus PD-1/PD-L1 antibody efficacy. These findings suggest that cDHPs inhibit NSCLC proliferation and immune escape via the FXR1-IL-35 axis signaling pathway.
Collapse
Affiliation(s)
- Xinying Zhu
- Translational Medicine Center, The Second Affiliated Hospital of Wannan Medical College, Wuhu, 241000, Anhui Province, China
| | - Guoquan Yin
- Clinical Laboratory, Yangzhou Blood Center in Jiangsu Province, Yangzhou, 225007, Jiangsu Province, China
| | - Jiaqian Xu
- Department of Medical Microbiology and Immunology, School of Preclinical Medicine, Wannan Medical College, Wuhu, 241001, Anhui Province, China
| | - Xiaolei Tang
- Translational Medicine Center, The Second Affiliated Hospital of Wannan Medical College, Wuhu, 241000, Anhui Province, China
| | - Fangliu Yu
- Department of Medical Microbiology and Immunology, School of Preclinical Medicine, Wannan Medical College, Wuhu, 241001, Anhui Province, China.
| |
Collapse
|
2
|
Fan YW, Liu MH, Xu TJ, Fan RY, Xiang J, Wu JQ, He MF. Mechanism of etoposide resistance in small cell lung cancer and the potential therapeutic options. Med Oncol 2025; 42:167. [PMID: 40257680 DOI: 10.1007/s12032-025-02718-0] [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: 12/03/2024] [Accepted: 04/14/2025] [Indexed: 04/22/2025]
Abstract
Small cell lung cancer (SCLC) is a type of high-grade neuroendocrine malignancy with low gene mutation. Chemotherapy is the major treatment strategy, but long-term clinical application often leads to drug resistance. Etoposide is a first-line drug approved by the US Food and Drug Administration for SCLC treatment, but etoposide-resistance is a problem. In this study, a SCLC cell line with etoposide-acquired resistance, H1048-ER, was constructed through a concentration gradient increasing method, and its resistance to etoposide was investigated in vitro and in a zebrafish model. Through transcriptome sequencing, real-time reverse transcription-quantitative polymerase chain reaction, and bioinformatic analyses of H1048-ER vs. H1048 cells, 51 differentially expressed genes were found to be significantly enriched in "collagen degradation" and "MET/FAK signaling activation in ECM". Among them, six genes (COL11A1, COL26A1, COL4A3, COL4A4, LAMA4, and LAMC1) had strong correlations with the prognosis of lung cancer. They may be key factors in the acquired etoposide resistance of H1048-ER cells. H1048-ER cells showed cross-resistance to cisplatin but were sensitive to doxorubicin and temozolomide. Our study provides novel insights into etoposide resistance in SCLC and affords the potential treatment options after etoposide resistance.
Collapse
Affiliation(s)
- Yan-Wen Fan
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 30 Puzhu South Road, Nanjing, 211816, China
| | - Mei-Hui Liu
- Jiangsu Health Vocational College, Nanjing, China
| | - Tao-Jun Xu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 30 Puzhu South Road, Nanjing, 211816, China
| | - Ruo-Yue Fan
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 30 Puzhu South Road, Nanjing, 211816, China
| | - Jing Xiang
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Jia-Qi Wu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 30 Puzhu South Road, Nanjing, 211816, China.
| | - Ming-Fang He
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 30 Puzhu South Road, Nanjing, 211816, China.
| |
Collapse
|
3
|
Das S, Zea MP, Russon MP, Xing Z, Torregrosa-Allen S, Cervantes HE, Harper HA, Elzey BD, Tran EJ. Supinoxin blocks small cell lung cancer progression by inhibiting mitochondrial respiration through DDX5. iScience 2025; 28:112219. [PMID: 40224004 PMCID: PMC11987007 DOI: 10.1016/j.isci.2025.112219] [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: 08/07/2024] [Revised: 12/26/2024] [Accepted: 03/11/2025] [Indexed: 04/15/2025] Open
Abstract
DDX5 is a DEAD-box RNA helicase that is overexpressed and implicated in the progression of several cancers, including small cell lung cancer (SCLC). Our laboratory has demonstrated that DDX5 is essential for the invasive growth of SCLC and mitochondrial respiration. SCLC is an extremely lethal, recalcitrant tumor, and currently lacking effective treatments. Supinoxin (RX 5902), a compound having anti-cancer activity, is a known target of phosphor-DDX5. We now report that Supinoxin inhibits the proliferation of chemo-sensitive and chemo-resistant SCLC lines, H69 and H69AR, respectively. Additionally, Supinoxin mitigates both the growth of H69AR xenograft tumors and SCLC PDX tumors in vivo. Finally, we find that Supinoxin inhibits expression of mitochondrial genes and effectively blocks respiration. These studies suggest that Supinoxin functions in anti-tumor progression by reducing cellular energy levels through DDX5.
Collapse
Affiliation(s)
- Subhadeep Das
- Department of Biochemistry, Purdue University, BCHM A343, 175 S. University Street, West Lafayette, IN 47907-2063, USA
- Purdue University Institute for Cancer Research, Purdue University, Hansen Life Sciences Research Building, Room 141, 201 S. University Street, West Lafayette, IN 47907-2064, USA
| | - Maria P. Zea
- Department of Biochemistry, Purdue University, BCHM A343, 175 S. University Street, West Lafayette, IN 47907-2063, USA
| | - Matthew P. Russon
- Department of Biochemistry, Purdue University, BCHM A343, 175 S. University Street, West Lafayette, IN 47907-2063, USA
| | - Zheng Xing
- Department of Biochemistry, Purdue University, BCHM A343, 175 S. University Street, West Lafayette, IN 47907-2063, USA
| | - Sandra Torregrosa-Allen
- Purdue University Institute for Cancer Research, Purdue University, Hansen Life Sciences Research Building, Room 141, 201 S. University Street, West Lafayette, IN 47907-2064, USA
| | - Heidi E. Cervantes
- Purdue University Institute for Cancer Research, Purdue University, Hansen Life Sciences Research Building, Room 141, 201 S. University Street, West Lafayette, IN 47907-2064, USA
| | - Haley Anne Harper
- Purdue University Institute for Cancer Research, Purdue University, Hansen Life Sciences Research Building, Room 141, 201 S. University Street, West Lafayette, IN 47907-2064, USA
| | - Bennett D. Elzey
- Purdue University Institute for Cancer Research, Purdue University, Hansen Life Sciences Research Building, Room 141, 201 S. University Street, West Lafayette, IN 47907-2064, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | - Elizabeth J. Tran
- Department of Biochemistry, Purdue University, BCHM A343, 175 S. University Street, West Lafayette, IN 47907-2063, USA
- Purdue University Institute for Cancer Research, Purdue University, Hansen Life Sciences Research Building, Room 141, 201 S. University Street, West Lafayette, IN 47907-2064, USA
| |
Collapse
|
4
|
Lin G, Yao Z, Kang K, Luo R, Yi L, Lu Y. Dynamic evolution and antitumor mechanisms of CXCR6 +CD8 + T cells in small cell lung cancer treated with low-dose radiotherapy and immunotherapy. J Transl Med 2025; 23:453. [PMID: 40247265 PMCID: PMC12007177 DOI: 10.1186/s12967-025-06450-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 04/03/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Patients with small-cell lung cancer (SCLC) have the poor prognosis. Current research suggested that low-dose radiotherapy (LDRT) combined with immunotherapy can enhance the immunogenicity of tumor cells, thereby improving antigen presentation and promoting the intratumoral infiltration of CD8+ T cells, which significantly extends the survival of patients. However, the change trajectory of T cells, and the mechanisms underlying the promotion of intratumoral infiltration of CD8+ T cells, and the enhancement of their cytotoxic functions remain to be elucidated. METHODS To delineate the dynamic changes of T cells, we collected tumors from Kaede tumor-bearing mice that had undergone radioimmunotherapy. Using flow cytometry, we sorted intratumoral-infiltrating immune cells, which were required for single-cell RNA sequencing, at various time points (Kaede Red: derived from tumor-draining lymph node [TDLN]). The results obtained from the sequencing analysis were further validated through experiments, such as flow cytometry, immunofluorescence, and analysis of clinical cohort data. RESULTS Here, we observed stem-like T cells migrating from the TDLN to the tumor site and differentiating into effector phenotypes within the tumor. Dendritic cells (DCs) are the key cluster that induces the differentiation of stem-like T cell into effector phenotypes. Moreover, SCLC patients with a high infiltration of tumor-specific CXCR6+CD8+ T cells exhibited a supportive TME and longer survival time (P < 0.001). CONCLUSIONS This study delineates the change trajectory of CD8+ T cells, identifies the crucial role of DCs in T cell differentiation, and highlights the significance of tumor-specific CXCR6+CD8+ T cells in anti-tumor immunity. Future therapeutic strategies for SCLC could focus on enhancing the infiltration of activated DCs and CXCR6+CD8+ T cells within the tumor microenvironment to improve treatment efficacy.
Collapse
Affiliation(s)
- Guo Lin
- Division of Thoracic Tumor Multimodality Treatment and Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhuoran Yao
- Division of Thoracic Tumor Multimodality Treatment and Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Kang
- Division of Thoracic Tumor Multimodality Treatment and Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ren Luo
- Division of Thoracic Tumor Multimodality Treatment and Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Linglu Yi
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - You Lu
- Division of Thoracic Tumor Multimodality Treatment and Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China.
| |
Collapse
|
5
|
Oskolas H, Nogueira FCN, Domont GB, Yu KH, Semenov YR, Sorger P, Steinfelder E, Corps L, Schulz L, Wieslander E, Fenyö D, Kárpáti S, Holló P, Kemény LV, Döme B, Megyesfalvi Z, Pawłowski K, Nishimura T, Kwon H, Encarnación-Guevara S, Szasz AM, Veréb Z, Gyulai R, Németh IB, Appelqvist R, Rezeli M, Baldetorp B, Horvatovich P, Malmström J, Pla I, Sanchez A, Knudsen B, Kiss A, Malm J, Marko-Varga G, Gil J. Comprehensive biobanking strategy with clinical impact at the European Cancer Moonshot Lund Center. J Proteomics 2025; 316:105442. [PMID: 40246065 DOI: 10.1016/j.jprot.2025.105442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/26/2025] [Accepted: 04/12/2025] [Indexed: 04/19/2025]
Abstract
This white paper presents a comprehensive biobanking framework developed at the European Cancer Moonshot Lund Center that merges rigorous sample handling, advanced automation, and multi-omic analyses to accelerate precision oncology. Tumor and blood-based workflows, supported by automated fractionation systems and standardized protocols, ensure the collection of high-quality biospecimens suitable for proteomic, genomic, and metabolic studies. A robust informatics infrastructure, integrating LIMS, barcoding, and REDCap, supports end-to-end traceability and realtime data synchronization, thereby enriching each sample with critical clinical metadata. Proteogenomic integration lies at the core of this initiative, uncovering tumor- and blood-based molecular profiles that inform cancer heterogeneity, metastasis, and therapeutic resistance. Machine learning and AI-driven models further enhance these datasets by stratifying patient populations, predicting therapeutic responses, and expediting the discovery of actionable targets and companion biomarkers. This synergy between technology, automation, and high-dimensional data analytics enables individualized treatment strategies in melanoma, lung, and other cancer types. Aligned with international programs such as the Cancer Moonshot and the ICPC, the Lund Center's approach fosters open collaboration and data sharing on a global scale. This scalable, patient-centric biobanking paradigm provides an adaptable model for institutions aiming to unify clinical, molecular, and computational resources for transformative cancer research.
Collapse
Affiliation(s)
- Henriett Oskolas
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Sweden
| | - Fábio C N Nogueira
- Research Center for Precision Medicine, IBCCF & Chemistry Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gilberto B Domont
- Research Center for Precision Medicine, IBCCF & Chemistry Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kun-Hsing Yu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Yevgeniy R Semenov
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter Sorger
- Department of Systems Biology, Harvrad Medical School, Boston, MA, USA
| | | | - Les Corps
- Alderley Park, Macclesfield, Cheshire, England, United Kingdom
| | | | - Elisabet Wieslander
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Sweden
| | - David Fenyö
- Department of Biochemistry and Molecular Pharmacology, Institute for Systems Genetics, New York University Grossman School of Medicine, New York, USA
| | - Sarolta Kárpáti
- Department of Dermatology, Venerology and Dermato oncology, Semmelweis University, Budapest, Hungary
| | - Péter Holló
- Department of Dermatology, Venerology and Dermato oncology, Semmelweis University, Budapest, Hungary
| | - Lajos V Kemény
- Department of Dermatology, Venerology and Dermato oncology, Semmelweis University, Budapest, Hungary; Department of Physiology, HCEMM-SU Translational Dermatology Research Group, Semmelweis University, Budapest, Hungary
| | - Balazs Döme
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Sweden; Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria; Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary; National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Zsolt Megyesfalvi
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria; Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary; National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Krzysztof Pawłowski
- Department of Molecular Biology, University of Texas Southwestern Medical Center, TX, USA
| | | | - HoJeong Kwon
- Chemical Genomics Global Research Lab, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | | | - A Marcell Szasz
- Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Zoltán Veréb
- University Hospital Szeged Biobank, Szeged, Hungary
| | - Rolland Gyulai
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - István Balázs Németh
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - Roger Appelqvist
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, Sweden
| | - Melinda Rezeli
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, Sweden
| | - Bo Baldetorp
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Peter Horvatovich
- Department of Analytical Biochemistry, Faculty of Science and Engineering, University of Groningen, Groningen, the Netherlands
| | - Johan Malmström
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden; BioMS, Department of Mass Spectrometry, Lund University, Lund, Sweden
| | - Indira Pla
- Proteomics Center of Excellence, Northwestern University, Evanston, IL, USA
| | - Aniel Sanchez
- Proteomics Center of Excellence, Northwestern University, Evanston, IL, USA
| | - Beatrice Knudsen
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - András Kiss
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Johan Malm
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Sweden
| | - György Marko-Varga
- Board of Directors, Japan Society of Clinical Proteogenomics, Tokyo, Japan; Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary; 1st Department of Surgery, Tokyo Medical University, Tokyo, Japan
| | - Jeovanis Gil
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Sweden.
| |
Collapse
|
6
|
Chen C, Zhao H, Yuan W, Chen H, Wang P, Wang Q, Chen C, Song T. Tumoral Nanovesicles-Loaded Magnetotactic Bacteria for Tumor-Targeted Therapy under a Swing Magnetic Field. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40231984 DOI: 10.1021/acsami.5c02422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Tumor heterogeneity poses numerous challenges for targeted drug therapy. Although tumor cell-derived nanovesicles (NVs) have emerged as an intriguing method for tumor targeting, how to exert the antitumor effect after targeting remains a key concern. Magnetotactic bacteria (MTB) synthesize chain-like magnetite (Fe3O4) crystals with inherent magnetic moments, which could generate significant torque under a desired magnetic field and move along the magnetic field using their own flagella. Herein, a composite of MTB AMB-1 and NVs was fabricated via electrostatic adsorption where AMB-1 could transport NVs to the tumor site by a guiding magnetic field, while NVs also assist AMB-1 in binding to tumor cells. Subsequently, under the influence of a swing magnetic field (sMF), MTB exert physical stimuli on the cells, inducing the changes of mitochondrial membrane potential and cellular reactive oxygen species (ROS). Finally, it is revealed that the NVs-loaded AMB-1 induced a decrease in cellular viability and significantly inhibited the growth of tumors in vivo under the sMF. Therefore, by remote control of the guidance and stimuli production, the NVs-loaded AMB-1 was highly promising to advance the development of targeted therapeutic strategies for tumors under the context of tumor heterogeneity.
Collapse
Affiliation(s)
- Changyou Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Haoyu Zhao
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Yuan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical Collage, Beijing 100021, China
| | - Haitao Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Pingping Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Qingmeng Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Chuanfang Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Tao Song
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
7
|
Feizi I, Sedigh-Namin A, Kani A, Namin SS, Toularoud AB. A rare case report of triple primary malignancies: synchronous breast ductal invasive carcinoma and lung neuroendocrine tumor, followed by chronic myeloid leukemia. Int J Surg Case Rep 2025; 130:111305. [PMID: 40245679 DOI: 10.1016/j.ijscr.2025.111305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/27/2025] [Accepted: 04/13/2025] [Indexed: 04/19/2025] Open
Abstract
INTRODUCTION AND IMPORTANCE Multiple primary malignancies (MPMTs), the occurrence of two or more different primary cancers in a single person, are rare. These tumors can be synchronous or metachronous, with an incidence ranging from 0.73 % to 11.70 % in cancer patients. While invasive ductal carcinoma (IDC) is the most common form of breast cancer and lung neuroendocrine tumors (NETs) are rare, their co-occurrence as MPMT is extremely rare. In addition, chronic myeloid leukemia (CML) makes such cases even more complex. CASE PRESENTATION We report the case of a 59-year-old woman who presented with a lump in the left breast. Diagnostic examinations confirmed breast IDC. Metastatic examination identified a separate primary, well-differentiated NET of the left lung. Both malignancies were treated surgically followed by appropriate adjuvant therapy. A year later, routine follow-up revealed an elevated white blood cell count and a subsequent bone marrow biopsy confirmed the diagnosis of CML. CLINICAL DISCUSSION The diagnosis of IDC and pulmonary NET as primary tumors is rare and the subsequent development of CML in this setting has not been reported previously. This case highlights the critical importance of thorough diagnostic evaluations to accurately differentiate between metastatic disease and MPMTs. The occurrence of three different malignancies in a single patient presents significant treatment challenges and highlights the need for personalized treatment approaches. CONCLUSION This report emphasizes the need for comprehensive diagnostic protocols in the evaluation of multiple tumors and contributes to the growing body of knowledge about MPMT. The coexistence of IDC, pulmonary NET and subsequent CML represents a rare and complex clinical scenario that requires continuous research into optimal management strategies for such cases.
Collapse
Affiliation(s)
- Iraj Feizi
- Department of Surgery, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Atabak Sedigh-Namin
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - AmirAbbas Kani
- SHAFA PARTOO ARDEBIL Medical Center, Radiational Oncology Department, Ardabil, Iran
| | - Sonia Sharifi Namin
- Department of Anatomical Sciences and Pathology, School of Medicine, Imam Reza Hospital, Ardabil University of Medical Sciences, Ardabil, Iran
| | | |
Collapse
|
8
|
Jiang S, Guo F, Li L. Biological mechanisms and immunotherapy of brain metastases in non-small cell lung cancer. Biochim Biophys Acta Rev Cancer 2025:189320. [PMID: 40220878 DOI: 10.1016/j.bbcan.2025.189320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/07/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Non-small cell lung cancer (NSCLC) is a leading cause of cancer-related mortality worldwide, with Brain Metastases serving as a significant adverse prognostic factor. The blood-brain barrier poses a substantial challenge in the treatment of brain metastases, as it restricts the penetration of many anticancer agents. Novel immunotherapy, such as immune checkpoint inhibitors (ICIs) have emerged as promising treatment for NSCLC and its associated brain metastases. This review summarizes the biological mechanism underlying NSCLC brain metastases and provides an overview of the current landscape of immunotherapy, exploring the mechanism of action and clinical applications of these advanced treatments.
Collapse
Affiliation(s)
- Sitong Jiang
- Department of Medical Oncology, Beijing Hospital, The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, National Center of Gerontology, National Health Commission, PR China; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China
| | - Fengzhu Guo
- Department of Medical Oncology, Beijing Hospital, The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, National Center of Gerontology, National Health Commission, PR China; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China
| | - Lin Li
- Department of Medical Oncology, Beijing Hospital, The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, National Center of Gerontology, National Health Commission, PR China; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China.
| |
Collapse
|
9
|
Xu H, Zhao Q, Cai D, Chen X, Zhou X, Gao Y, Wu J, Yuan S, Li D, Zhang R, Peng W, Li G, Nan A. o8G-modified circKIAA1797 promotes lung cancer development by inhibiting cuproptosis. J Exp Clin Cancer Res 2025; 44:110. [PMID: 40176113 PMCID: PMC11963662 DOI: 10.1186/s13046-025-03365-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 03/13/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Lung cancer is a serious threat to human life and health, but effective screening and treatment methods are lacking. Circular RNAs (circRNAs) have important biological functions and are closely related to tumour development. Some studies have shown that the 8-oxo-7,8-dihydroguanosine (o8G) modification plays a key role in the disease process, but the effect of the o8G modification on circRNAs has not been elucidated. Moreover, cuproptosis is a novel mode of cell death in which copper ions directly promote protein aggregation and the disruption of cellular metabolic pathways. The present study revealed that the o8G modification of circKIAA1797 occurs and promotes lung cancer development by inhibiting cuproptosis, which provides new perspectives for epitranscriptomic studies and the development of novel therapeutic approaches for lung cancer. METHODS circRNA differential expression profiles in lung cancer were revealed via RNA high-throughput sequencing, and circKIAA1797 expression in lung cancer cell lines and tissues was detected using qPCR. Experiments such as o8G RNA immunoprecipitation (o8G RIP) and crosslinking immunoprecipitation (CLIP) were performed to explore the presence of o8G on circKIAA1797. The regulation of circKIAA1797 by the o8G reader Y-box binding protein 1 (YBX1) was explored using nuclear-cytoplasmic fractionation, actinomycin D (Act D) stability experiments and other experiments. circKIAA1797 silencing and overexpression systems were constructed for in vivo and in vitro experiments to study the role of circKIAA1797 in lung cancer development. Tagged RNA affinity purification (TRAP), RNA immunoprecipitation (RIP), coimmunoprecipitation (Co-IP), and immunofluorescence (IF) staining were subsequently conducted to reveal the molecular mechanism by which circKIAA1797 regulates cuproptosis and promotes lung cancer development. RESULTS This study is the first to reveal the presence of o8G on circKIAA1797 and that YBX1 is a reader that recognises ROS-induced circKIAA1797 o8G modifications and increases the stability and cytoplasmic expression of circKIAA1797. circKIAA1797, which is associated with the tumour stage and prognosis, has been shown to significantly promote the biological function of lung cancer development both in vivo and in vitro. This study revealed that circKIAA1797 inhibits intracellular cuproptosis by binding to the ferredoxin 1 (FDX1) mRNA, decreasing FDX1 mRNA stability, inhibiting FDX1 expression, and binding to the signal transducer and activator of transcription 1 (STAT1) protein and inhibiting lipoyltransferase 1 (LIPT1) transcription; moreover, circKIAA1797 promotes the closure of the mitochondrial permeability transition pore (mPTP), inhibits cuproptosis, and ultimately promotes lung cancer development. CONCLUSIONS This study revealed the presence of the o8G modification in circKIAA1797, which plays an important role in the development of lung cancer. circKIAA1797 can inhibit cuproptosis by inhibiting key cuproptosis proteins and promoting mPTP closure, ultimately promoting the development of lung cancer. This study provides not only a new theoretical basis for an in-depth understanding of the molecular mechanisms of lung cancer development but also a potential target for lung cancer treatment.
Collapse
Affiliation(s)
- Haotian Xu
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Qingyun Zhao
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Dunyu Cai
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Xingcai Chen
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Xiaodong Zhou
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Yihong Gao
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Jiaxi Wu
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Shengyi Yuan
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Deqing Li
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Ruirui Zhang
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Wenyi Peng
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Gang Li
- School of Public Health, Guangxi Medical University, Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China.
| | - Aruo Nan
- School of Public Health, Guangxi Medical University, Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
10
|
Csende K, Ferencz B, Boettiger K, Pozonec MD, Lantos A, Ferenczy A, Pipek O, Solta A, Ernhofer B, Laszlo V, Megyesfalvi E, Schelch K, Pozonec V, Skarda J, Skopelidou V, Lohinai Z, Lang C, Horvath L, Dezso K, Fillinger J, Renyi-Vamos F, Aigner C, Dome B, Megyesfalvi Z. Comparative profiling of surgically resected primary tumors and their lymph node metastases in small-cell lung cancer. ESMO Open 2025; 10:104514. [PMID: 40107154 PMCID: PMC11964634 DOI: 10.1016/j.esmoop.2025.104514] [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: 08/06/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Profiling studies in small-cell lung cancer (SCLC) have mainly focused on primary tumors, omitting the potential molecular changes that might occur during lymphatic metastasis formation. Here, we assessed the molecular discordance between primary SCLCs and corresponding lymph node (LN) metastases in the light of subtype distribution and expression of clinically relevant proteins. METHODS Comparative profiling of 32 surgically resected primary SCLCs and their LN metastases was achieved by RNA expression analysis and immunohistochemistry (IHC). In addition to subtype markers (ASCL1, NEUROD1, POU2F3, and YAP1), the expression of nine cancer-specific proteins was evaluated. RESULTS The selected clinically relevant molecules showed no significant differences in their RNA expression profile when assessing the primary tumors and their corresponding LN metastases. Nevertheless, IHC analyses revealed significantly higher DLL3 expression in the primary tumors than in the LN metastases (P = 0.008). In contrast, NEUROD1 expression was significantly lower in the primary tumors (versus LN metastases, P < 0.001). No statistically significant difference was found by IHC analysis in the case of other clinically relevant proteins. Concerning SCLC molecular subtypes, a change in subtype distribution was detected in 21 cases. Phenotype switching from neuroendocrine (NE) subtypes toward non-NE lesions and from non-NE landscape toward NE subtypes were both detected. CONCLUSIONS Although the molecular landscape of SCLC LN metastases largely resembles that of the tumor of origin, key differences exist in terms of DLL3 and NEUROD1 expression, and in subtype distribution. These diagnostic pitfalls should be considered when establishing the tumors' molecular profile for future clinical trials solely based on LN biopsies.
Collapse
Affiliation(s)
- K Csende
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary; National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - B Ferencz
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary; National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - K Boettiger
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - M D Pozonec
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary; National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - A Lantos
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - A Ferenczy
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary; Department of Obstetrics and Gynecology, South Buda Central Hospital, Saint Emeric University Teaching Hospital, Budapest, Hungary
| | - O Pipek
- Department of Physics of Complex Systems, Eotvos Lorand University, Budapest, Hungary
| | - A Solta
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - B Ernhofer
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - V Laszlo
- National Koranyi Institute of Pulmonology, Budapest, Hungary; Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - E Megyesfalvi
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary; Department of Thoracic and Abdominal Tumors and Clinical Pharmacology, National Institute of Oncology, Budapest, Hungary
| | - K Schelch
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - V Pozonec
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary; Multidisciplinary Centre of Head and Neck Tumors, National Institute of Oncology, Budapest, Hungary
| | - J Skarda
- Institute of Clinical and Molecular Pathology, Medical Faculty, Palacký University Olomouc, Olomouc, Czech Republic; Department of Pathology, University Hospital Ostrava, Ostrava, Czech Republic; Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - V Skopelidou
- Department of Pathology, University Hospital Ostrava, Ostrava, Czech Republic; Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Z Lohinai
- Torokbalint County Institute of Pulmonology, Torokbalint, Hungary
| | - C Lang
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - L Horvath
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - K Dezso
- Department of Pathology and Experimental Cancer Research, Budapest, Hungary
| | - J Fillinger
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - F Renyi-Vamos
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary; National Koranyi Institute of Pulmonology, Budapest, Hungary; National Institute of Oncology and National Tumor Biology Laboratory, Budapest, Hungary
| | - C Aigner
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - B Dome
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary; National Koranyi Institute of Pulmonology, Budapest, Hungary; Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Translational Medicine, Lund University, Lund, Sweden.
| | - Z Megyesfalvi
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary; National Koranyi Institute of Pulmonology, Budapest, Hungary; Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
11
|
Zhai X, Zhang Z, Chen Y, Wu Y, Zhen C, Liu Y, Lin Y, Chen C. Current and future therapies for small cell lung carcinoma. J Hematol Oncol 2025; 18:37. [PMID: 40170056 PMCID: PMC11959764 DOI: 10.1186/s13045-025-01690-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/14/2025] [Indexed: 04/03/2025] Open
Abstract
Small cell lung cancer (SCLC) is an aggressive malignancy characterized by rapid proliferation and high metastatic potential. It is characterized by universal inactivation of and RB1, overexpression of the MYC family and dysregulation of multiple oncogenic signaling pathways. Among different patients, SCLCs are similar at the genetic level but exhibit significant heterogeneity at the molecular level. The classification of SCLC has evolved from a simple neuroendocrine (NE)/non-neuroendocrine (non-NE) classification system to a transcription factor-based molecular subtype system; lineage plasticity adds further complexity and poses challenges for therapeutic development. While SCLC is initially sensitive to platinum-based chemotherapy, resistance develops rapidly, leading to a dismal prognosis. Various antibodies, including PD-1/PD-L1 inhibitors and antibody‒drug conjugates, have been introduced into clinical practice or are being evaluated in clinical trials. However, their therapeutic benefits for SCLC patients remain limited. This review summarizes SCLC carcinogenic mechanisms, tumor heterogeneity, and the immune microenvironment of SCLC, with a focus on recent advances in metastasis and resistance mechanisms. Additionally, the corresponding clinical progress in tackling these challenges is discussed.
Collapse
Affiliation(s)
- Xiaoqian Zhai
- Department of Medical Oncology, State Key Laboratory of Biotherapy and Cancer Center and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 1, Keyuan 4th Road, Gaopeng Avenue, Chengdu, 610041, Sichuan, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhengkun Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- College of Life Sciences, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuxin Chen
- West China School of Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yanmou Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- College of Life Sciences, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Cheng Zhen
- West China School of Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yu Liu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 1, Keyuan 4th Road, Gaopeng Avenue, Chengdu, 610041, Sichuan, China.
| | - Yiyun Lin
- Department of Medicine, Weill Cornell Medicine, East 69th Street, New York, NY, 10021, USA.
| | - Chong Chen
- Department of Medical Oncology, State Key Laboratory of Biotherapy and Cancer Center and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 1, Keyuan 4th Road, Gaopeng Avenue, Chengdu, 610041, Sichuan, China.
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
12
|
Xu J, Zhang W, Xie F, Wang C, Cheng F, Rao R, Chen Y, Zhang L, Wen W, Zhao Z, Yuan J, Zheng Y, Yu Z. Prognostic significance of ALK high expression in SCLC: a 9-year cohort analysis. Front Oncol 2025; 15:1530339. [PMID: 40201354 PMCID: PMC11975910 DOI: 10.3389/fonc.2025.1530339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/04/2025] [Indexed: 04/10/2025] Open
Abstract
Purpose The aim of this study was to investigate the prognostic value of the abnormal expression of anaplastic lymphoma kinase (ALK) protein in patients with small cell lung cancer (SCLC) based on 9-year data from our center. Methods A retrospective cohort study was conducted to assess the clinical outcomes of patients with ALK-positive SCLC diagnosed in our hospital over the past 9 years. We used public databases to analyze the expression of ALK in pan-cancer and its prognostic value and analyzed the correlation between ALK and SCLC prognosis-related genes. Results A total of 685 patients diagnosed with SCLC underwent ALK testing, and 59 patients were identified to have abnormal expression of the ALK protein, with 10 cases showing strong expression, 14 cases displaying moderate expression, and 35 cases exhibiting weak expression. The median age of the ALK-positive cohort was 64 years (range: 58-70 years), 91.5% (54/59) were male, 61.0% (36/59) were smokers, and the median overall survival (mOS) was 7.0 months (95% CI: 4.5-9.5 months). Within this cohort, the mOS for the ALK (+) subgroup was 4.0 months (95% CI: 2.9-5.1 months), the mOS for the ALK (++) subgroup was 10.0 months (95% CI: 4.9-15.1 months), and the mOS for the ALK (+++) subgroup was 12.0 months (95% CI: 7.4-16.6 months). Kaplan-Meier revealed that the mOS of the ALKLow group was significantly worse than that of the ALKHigh group [mOS: 4.0 months (95% CI: 2.9-5.1 months) versus 11.0 months (95% CI: 8.3-13.7 months), p = 0.009]. Following covariate adjustment using a Cox regression model, it was indicated that the level of abnormal expression of the ALK protein was an independent prognostic factor for patients with SCLC (HR: 0.486, 95% CI: 0.271-0.871, p = 0.015). Conclusion The prognosis for patients with SCLC with strong abnormal expression of the ALK protein was significantly better than those with weak expression.
Collapse
Affiliation(s)
- Jinhe Xu
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Wenting Zhang
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Feilai Xie
- Pathology Department, Dongfang Hospital of Xiamen University, Fuzhou General Hospital of Fujian Medical University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Chenxi Wang
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Feng Cheng
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Ruiying Rao
- Department of Respiratory and Critical Care Medicine, Fuzong Teaching Hospital, Fujian University of Traditional Chinese Medicine (900 Hospital), Fuzhou, China
| | - Ying Chen
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Lei Zhang
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Wen Wen
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Zhongquan Zhao
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Jialing Yuan
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Yuqin Zheng
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Zongyang Yu
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| |
Collapse
|
13
|
Zhang B, Shah KB, Parma M, Wang K, Singhi EK, Lewis W, Rivera M, Altan M, Pozadzides J, Le X, Vokes N, Fossella F, O'Brien B, Wang C, Tom MC, Beckham T, Swanson T, Bronk J, Lin SH, Vega MF, Jacome J, Halliday A, Negrao M, Zhang J, Gibbons DL, Heymach JV, Byers LA, Gay CM. Rapid Intracranial Response With Tarlatamab in Patients With Untreated Brain Metastases From SCLC-A Real-World Case Series: Case Report. J Thorac Oncol 2025:S1556-0864(25)00103-0. [PMID: 40126456 DOI: 10.1016/j.jtho.2025.02.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 02/13/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025]
Abstract
SCLC has the highest propensity for brain metastases among all malignancies. Systemic treatment for SCLC, particularly in the setting of brain metastases, is very limited. Tarlatamab, the CD3/delta-like ligand 3 bispecific T-cell engager, has changed the treatment landscape of relapsed SCLC since its Food and Drug Administration approval in May 2024. Patients with treated and stable brain metastases were included in the phase 1 DeLLphi-300 trial and phase 2 DeLLphi-301 trials of tarlatamab. Nevertheless, it remains unknown if tarlatamab is safe and efficacious in the setting of untreated, active or symptomatic brain metastases. Our case series provides, to our knowledge, the first reported evidence of the safety and efficacy of tarlatamab in patients with untreated brain metastases. In our cohort of 10 patients with relapsed SCLC and untreated brain metastases, including those with symptomatic intracranial disease and one with suspected leptomeningeal disease, clinical response or stability was seen in 90% of patients. We present several cases in which rapid, dramatic radiographic responses and clinical improvement were observed in patients with innumerable growing brain metastases (>20 lesions) who would otherwise require whole brain radiation, suggesting that tarlatamab can control intracranial metastases as monotherapy, potentially sparing or deferring the need for brain radiation.
Collapse
Affiliation(s)
- Bingnan Zhang
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Komal B Shah
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mitchell Parma
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kaiwen Wang
- Division of Pharmacy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eric K Singhi
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Whitney Lewis
- Division of Pharmacy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Melvin Rivera
- Division of Pharmacy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mehmet Altan
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jenny Pozadzides
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiuning Le
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Natalie Vokes
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Frank Fossella
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Barbara O'Brien
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chenyang Wang
- Department of CNS-Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Martin C Tom
- Department of CNS-Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Thomas Beckham
- Department of CNS-Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Todd Swanson
- Department of CNS-Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Julianna Bronk
- Department of Thoracic Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steven H Lin
- Department of Thoracic Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maria Franco Vega
- Department of Hospital Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joshua Jacome
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexa Halliday
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marcelo Negrao
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jianjun Zhang
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Don L Gibbons
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John V Heymach
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lauren A Byers
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Carl M Gay
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
14
|
Yu J, Xu BT, Li Q, Shang ZT. Tankyrase 2 as a therapeutic target in non-small cell lung cancer: Implications for apoptosis and migration. World J Clin Oncol 2025; 16:103234. [PMID: 40130048 PMCID: PMC11866084 DOI: 10.5306/wjco.v16.i3.103234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 01/21/2025] Open
Abstract
This letter addresses Wang and Zhang's investigation into the role of tankyrase 2 (TNKS2) as a pivotal driver of malignancy in non-small cell lung cancer (NSCLC) through mechanisms including apoptosis inhibition, enhanced cellular migration, and β-catenin pathway activation. Their study in NSCLC cell lines demonstrates that TNKS2 overexpression stabilizes β-catenin, subsequently triggering oncogenic gene expression and facilitating cellular migration-key attributes of metastatic potential. These insights position TNKS2 as a compelling target for therapy and a potential prognostic marker in NSCLC. Nevertheless, translating these in vitro findings to clinical practice requires validation in in vivo models. Additionally, further research should investigate TNKS2 expression in patient samples and assess its implications in therapy resistance and combination treatment strategies.
Collapse
Affiliation(s)
- Jing Yu
- Department of Respiratory Medicine, Zhuji people’s Hospital, Zhuji 311800, Zhejiang Province, China
| | - Bo-Tao Xu
- Department of Cardiothoracic Surgery, Zhuji People’s Hospital, Zhuji 311800, Zhejiang Province, China
| | - Qiu Li
- Department of Respiratory Medicine, Zhuji people’s Hospital, Zhuji 311800, Zhejiang Province, China
| | - Zhong-Tu Shang
- Department of Respiratory Medicine, Zhuji people’s Hospital, Zhuji 311800, Zhejiang Province, China
| |
Collapse
|
15
|
Liu X, Yin X, Zhuang L, Wen J, Wei Z, Cui W, Yu M, Zhao K, Liu L, Kong L, Jiang L, Jing X, Zhu H, Wang X, Dong X, Yu J, Meng X. Efficacy and safety of TQB2450 combined with anlotinib as maintenance therapy for LS-SCLC after definitive concurrent or sequential chemoradiotherapy: a prospective phase Ib study. BMC Cancer 2025; 25:509. [PMID: 40114144 PMCID: PMC11924736 DOI: 10.1186/s12885-025-13885-8] [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: 01/15/2025] [Accepted: 03/07/2025] [Indexed: 03/22/2025] Open
Abstract
PURPOSE There is a significant unmet need in treating patients with limited-stage small-cell lung cancer (LS-SCLC). The ETER701 study showed that Benmelstobart (TQB2450, an anti-PD-L1 antibody) combined with Anlotinib and chemotherapy achieved the longest progression-free survival (PFS) and overall survival (OS) as a first-line therapy in patients with extensive-stage small cell lung cancer (ES-SCLC). This suggests that TQB2450 and Anlotinib represent a promising treatment combination for LS-SCLC. This prospective study aimed to evaluate the efficacy and safety of TQB2450 combined with Anlotinib as maintenance therapy for LS-SCLC following concurrent or sequential chemoradiotherapy (CCRT or SCRT). METHODS Patients who did not show disease progression after chemoradiotherapy were enrolled. They received TQB2450 and Anlotinib every 3 weeks for up to 24 months. TQB2450 was intravenously administered at a dose of 1200 mg every 3 weeks. Anlotinib was initiated at a dose of 8 mg daily for days 1-14; if well tolerated, the dose was increased to 10 mg. Adverse events (AEs) were recorded using electronic data capture system. The trial was registered at the ClinicalTrials.gov (NCT05942508, 06/07/2023). RESULTS Fifteen patients were enrolled in the study between May 31, 2023 and October 13, 2023. As of October 31, 2024, the median follow-up time was 15.13 months. The 12-month PFS rate was 86.7% (95% CI, 71.1-100.0), and the OS rate at 12 months was 100%. The disease control rate was 100%. AEs were reported in 13 patients (86.67%), with fatigue being the most common treatment related AE (40.00%). And two SAEs were observed (elevation in cardiac troponin T and cerebral infarction), which were determined to be unlikely unrelated to the trial drugs. Radiation pneumonitis (RP) occurred in three patients, all classified as grade 2, and one patient developed grade 1 immune-related pneumonitis. No grade 5 AEs occurred, and no patients withdrew from the study due to AEs. CONCLUSIONS TQB2450 combined with Anlotinib showed promising efficacy and well tolerance in patients with LS-SCLC following first-line treatment. A randomized, double-blind, placebo-controlled Phase III clinical study (ClinicalTrials.gov Identifier: NCT06469879) is being conducted to further explore the efficacy and safety of TQB2450 combined with Anlotinib as maintenance therapy after definitive CCRT or SCRT for LS-SCLC. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT05942508. Date of registration: 7 June 2023.
Collapse
Affiliation(s)
- Xiaoli Liu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, Shandong, 250117, China
| | - Xiaoyan Yin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, Shandong, 250117, China
| | - Lulu Zhuang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, Shandong, 250117, China
| | - Junxu Wen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, Shandong, 250117, China
| | - Zhonghui Wei
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, Shandong, 250117, China
| | - Wenxing Cui
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, Shandong, 250117, China
| | - Minghao Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, Shandong, 250117, China
| | - Kaikai Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, Shandong, 250117, China
| | - Lanping Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, Shandong, 250117, China
| | - Lingling Kong
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, Shandong, 250117, China
| | - Liyang Jiang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, Shandong, 250117, China
| | - Xuquan Jing
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, Shandong, 250117, China
| | - Hui Zhu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, Shandong, 250117, China
| | - Xunqiang Wang
- Chia Tai Tianqing Pharmarceutical Group Co., Ltd., Nanjing, 211122, China
| | - Xinjun Dong
- Chia Tai Tianqing Pharmarceutical Group Co., Ltd., Nanjing, 211122, China
| | - Jinming Yu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, Shandong, 250117, China.
| | - Xiangjiao Meng
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, Shandong, 250117, China.
| |
Collapse
|
16
|
Lu C, Su Y, Xu Y, Sheng S, Chen T, Li J. A novel PAK1/TCF1 regulatory axis promotes non-small cell lung cancer progression. Discov Oncol 2025; 16:364. [PMID: 40111665 PMCID: PMC11926319 DOI: 10.1007/s12672-025-02110-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the leading cause of cancer death, necessitating the identification of novel therapeutic targets. P21-activated kinases-1 (PAK1) plays a crucial role in oncogenesis, including NSCLC. Recent findings have elucidated T cell factor 1 (TCF1) as an anti-tumour factor, influencing T cell biology. However, the precise mechanism by which PAK1 promotes NSCLC progression via TCF1 regulation remains unclear. METHODS We collected 23 pairs of NSCLC tissue samples and obtained NSCLC RNA sequencing data and corresponding clinicopathologic information from The Cancer Genome Atlas (TCGA). Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC) assessed PAK1 and TCF1 expression in NSCLC tissues and cells. Gain and loss-of-function experiments evaluated PAK1 and TCF1 effects on cell proliferation, invasion, migration, and apoptosis in vitro. Mechanistically, western blot (WB) and immunoprecipitation analysis evaluated the interaction between PAK1 and TCF1 in NSCLC. Finally, we assessed the clinical prognostic, disease progression, and immunotherapy response of PAK1 and TCF1 and their correlation with immune cell infiltration, immune checkpoint inhibitors (PD1, PDL1). RESULTS PAK1 expression was elevated in NSCLC tissues and cells, while TCF1 was significantly downregulated. PAK1 expression showed a significant inverse correlation with TCF1 mRNA in NSCLC. Silencing PAK1 (using shRNAs) and inhibiting PAK1 with the small molecule IPA-3 suppressed NSCLC cell malignancy in a dose-dependent manner, upregulating TCF1 expression, and vice versa. TCF1 amplification with the small molecule (TWS119) inhibited NSCLC cell proliferation, migration, and invasion in a dose-dependent manner without affecting PAK1 expression. Immunoprecipitation analysis confirmed PAK1 and TCF1 interaction in NSCLC. Joint survival analysis indicated that high PAK1 and low TCF1 expression were associated with unfavourable survival in patients with NSCLC. Lastly, the TCF1 was significantly correlated with immune cell infiltration [CD8+ T cell, and tumor infiltrating lymphocytes (TILs)], immune checkpoint inhibitors (PD1, PDL1), and can accurately predict the immunotherapeutic response. CONCLUSION This study demonstrates, for the first time, that PAK1 negatively regulates TCF1, contributing to NSCLC pathogenesis. The PAK1/TCF1 regulatory axis emerges as a critical determinant of carcinogenesis and a promising therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Chuangang Lu
- Department of Thoracic Surgery, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya, 572000, Hainan, People's Republic of China.
| | - Yuncong Su
- Department of Thoracic Surgery, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya, 572000, Hainan, People's Republic of China
| | - Youzhong Xu
- Department of Thoracic Surgery, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya, 572000, Hainan, People's Republic of China
| | - Siyuan Sheng
- Department of Medicine, Hunan University of Arts and Science, Changde, 415000, Hunan, People's Republic of China
| | - Taiting Chen
- Department of Gynaecology and Obstetrics, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya, 572000, Hainan, People's Republic of China
| | - Juan Li
- Department of Reproductive Medicine, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya, 572000, Hainan, People's Republic of China.
| |
Collapse
|
17
|
Kim M, Lee C, Yoon SY, Park SH, Hwang JH, Kang K, Park E, Choi S, Lee SY, Yoo SS, Chae YS, Park KS. Comparative analysis of the lumboperitoneal shunt versus ventriculoperitoneal shunt for leptomeningeal metastasis-associated hydrocephalus in non-small cell lung cancer. Acta Neurochir (Wien) 2025; 167:81. [PMID: 40106020 PMCID: PMC11922981 DOI: 10.1007/s00701-025-06490-y] [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: 01/13/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025]
Abstract
PURPOSE Leptomeningeal metastasis (LM)-associated hydrocephalus is a rare but severe complication of non-small cell lung cancer (NSCLC). The spread of malignant cells to the leptomeninges obstructs cerebrospinal fluid flow and increases intracranial pressure (ICP). This study compared the outcomes of lumboperitoneal (LP) and ventriculoperitoneal (VP) shunt surgeries in managing LM-associated hydrocephalus, focusing on symptom-free periods (SFPs) and overall survival (OS). METHODS A retrospective analysis was conducted on 43 NSCLC patients with LM-associated hydrocephalus who underwent shunt surgery between 2017 and 2024. Patients were classified into LP (n = 23) and VP (n = 20) groups. Clinical characteristics, surgical outcomes, and survival rates were analyzed using Kaplan-Meier survival and Cox regression analyses. Karnofsky performance status (KPS), increased intracranial pressure (IICP) symptoms, and postoperative epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) treatment were evaluated for prognostic significance. RESULTS No significant difference was observed between VP and LP shunt surgeries regarding SFP (VP: 5.02 ± 1.29 months, LP: 7.50 ± 2.86 months, p = 0.906) or OS (VP: 8.43 ± 1.89 months, LP: 9.62 ± 3.20 months, p = 0.820). High preoperative KPS, absence of ICP symptoms, and postoperative EGFR-TKI treatment were significantly associated with improved SFP and OS (p < 0.05). LP shunt surgery had shorter anesthesia and fewer complications compared to VP shunt surgery, representing a viable option for patients unsuitable for general anesthesia. CONCLUSION LP and VP shunt surgeries are equally effective for patients with LM-associated hydrocephalus in NSCLC. LP shunt surgery under local anesthesia could be recommended for its safety and patient acceptability. Favorable prognostic factors, including high KPS, absence of ICP, and postoperative EGFR-TKI treatment, should guide individualized treatment strategies to enhance patient outcomes and quality of life.
Collapse
Affiliation(s)
- Minjoon Kim
- Department of Neurosurgery, School of Medicine, Kyungpook National University, 101 Dongin-dong 2 Ga, Jung-gu, Daegu, 700-422, Republic of Korea
| | - Chaejin Lee
- Department of Neurosurgery, School of Medicine, Kyungpook National University, 101 Dongin-dong 2 Ga, Jung-gu, Daegu, 700-422, Republic of Korea
| | - Sang-Youl Yoon
- Department of Neurosurgery, School of Medicine, Kyungpook National University, 101 Dongin-dong 2 Ga, Jung-gu, Daegu, 700-422, Republic of Korea
| | - Seong-Hyun Park
- Department of Neurosurgery, School of Medicine, Kyungpook National University, 101 Dongin-dong 2 Ga, Jung-gu, Daegu, 700-422, Republic of Korea
| | - Jeong-Hyun Hwang
- Department of Neurosurgery, School of Medicine, Kyungpook National University, 101 Dongin-dong 2 Ga, Jung-gu, Daegu, 700-422, Republic of Korea
| | - Kyunghun Kang
- Department of Neurology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Eunhee Park
- Department of Rehabilitation Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Sunha Choi
- Division of Pulmonology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Shin Yup Lee
- Division of Pulmonology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Seung Soo Yoo
- Division of Pulmonology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Yee Soo Chae
- Department of Hemato/Oncology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Ki-Su Park
- Department of Neurosurgery, School of Medicine, Kyungpook National University, 101 Dongin-dong 2 Ga, Jung-gu, Daegu, 700-422, Republic of Korea.
| |
Collapse
|
18
|
Gao Y, Zhang L, Yan M, Sun Z, Zhao H, Zhao L. Development of a prognostic model for patients with extensive-stage small cell lung cancer undergoing immunotherapy and chemotherapy. Front Immunol 2025; 16:1561333. [PMID: 40124379 PMCID: PMC11926142 DOI: 10.3389/fimmu.2025.1561333] [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: 01/20/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
Purpose In this study, we aimed to develop a predictive model for patients receiving chemotherapy and immunotherapy for extensive-stage small cell lung cancer. Methods We retrospectively analyzed 112 extensive-stage small cell lung cancer patients treated with first-line immunotherapy and chemotherapy. The relevant clinical data were collected to evaluate the changes during the treatment. The best subset regression, univariate analysis, and LASSO regression with cross-validation were applied for variable selection and model establishment. The nomograms for 1- and 2-year survival probabilities were established, and the calibration curve was utilized to evaluate the correspondence between actual and predicted survival. The model prediction capacity was assessed using decision curve analysis, calibration curves, and receiver operating characteristic curves. Moreover, five-fold cross-validation was conducted for internal validation. According to risk score, the patients were assigned to high- and low-risk groups, and survival curves were generated for each group. Results The LASSO regression model was established based on the variables such as age, ECOG, metastatic sites, NLR, and immunotherapy cycles. This predictive model displayed robust performance, evidenced by the Area Under the Curve of 0.887 and concordance index of 0.759. The nomogram effectively predicted 1- and 2-year survival probabilities and demonstrated a high degree of calibration. The decision curve analysis displayed that the model possessed superior predictive capability. The risk stratification for patients with high- and low-risk categories facilitated more individualized survival assessment. Conclusion The study successfully developed a prognostic model for extensive-stage small cell lung cancer patients undergoing immunotherapy and chemotherapy, demonstrating the good accuracy and predictability.
Collapse
Affiliation(s)
- Yunbin Gao
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Department of Oncology, Jining No. 1 People’s Hospital, Jining, Shandong, China
| | - Lixia Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Meng Yan
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Zongwen Sun
- Department of Oncology, Jining No. 1 People’s Hospital, Jining, Shandong, China
| | - Haibo Zhao
- Department of Oncology, Jining No. 1 People’s Hospital, Jining, Shandong, China
| | - Lujun Zhao
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
19
|
Eldin MH, Arafa KK, Gamal O, Keshk S, El-Sherbiny IM. Inhalable pH-responsive core-shell nanocarriers with PEGylated chitosan/alginate layer-by-layer coating for sequential drug release in lung cancer therapy. Int J Biol Macromol 2025; 307:141851. [PMID: 40058440 DOI: 10.1016/j.ijbiomac.2025.141851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 03/01/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Lung cancer remains one of the most lethal malignancies globally, underscoring the dire need for effective therapy. Scheduled administration of gemcitabine (GMC) followed by docetaxel (DTX) is clinically employed. Yet, the detrimental systemic toxicity and pharmacokinetic inadequacies such as the short plasma half-life of the former and poor bioavailability of the latter limit their use. Herein, we report the development of a novel inhalable nanocarrier system (NC) to enable the sequential release of drugs as per the clinical protocol. The developed NC has core-shell structure, with aminated mesoporous silica (MSNs) homing DTX at the core; enclosed within the polyanionic alginate (Alg) to prevent premature DTX release and serve as an intermediary sellotape layer. The outermost shell is polycationic as-synthesized PEGylated-chitosan (PEG-CS) loaded with GMC, to ensure stealth characteristics and prompt release of GMC. The newly developed PEG-CS/Alg@MSNs core-shell nanocarriers were comprehensively characterized. Besides it was evaluated in-vitro on A549 cell line and its in-vivo biodistribution was determined using jet nebulizer. Physicochemical analysis confirmed spherical core-shell NCs, 150 nm in size with +32 ± 1.5 mV surface charge. Drug entrapment efficiency was 75.2 ± 2.1 % for DTX and 32.5 ± 6.5 % for GMC, with sequential release in physiological conditions. Next Generation Impactor (NGI) experiments showed effective lung deposition with favorable aerosolization behavior. In-vitro assays on A549 cells revealed enhanced lung cancer treatment. In-vivo biodistribution confirmed lung accumulation, and histopathology indicated safety of NC. Conclusively, inhalable targeted NCs deem promising for lung cancer treatment.
Collapse
Affiliation(s)
- Mariam Hossam Eldin
- Nanomedicine Laboratories, Center for Materials Science, Zewail City of Science and Technology, 6th of October City, 12578 Giza, Egypt
| | - Kholoud K Arafa
- Nanomedicine Laboratories, Center for Materials Science, Zewail City of Science and Technology, 6th of October City, 12578 Giza, Egypt
| | - Osman Gamal
- Nanomedicine Laboratories, Center for Materials Science, Zewail City of Science and Technology, 6th of October City, 12578 Giza, Egypt
| | - Sarah Keshk
- Center of Genomics, Zewail City of Science and Technology, 6th of October City, 12578 Giza, Egypt
| | - Ibrahim M El-Sherbiny
- Nanomedicine Laboratories, Center for Materials Science, Zewail City of Science and Technology, 6th of October City, 12578 Giza, Egypt.
| |
Collapse
|
20
|
Chen Y, Liu X, Sun Y, Liu K, Ding D, Song S, Tan W. Noninvasive molecular imaging using anti-Trop-2 aptamer for targeted therapy of small cell lung cancer. J Nanobiotechnology 2025; 23:182. [PMID: 40050871 PMCID: PMC11887224 DOI: 10.1186/s12951-025-03184-6] [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: 10/29/2024] [Accepted: 02/01/2025] [Indexed: 03/09/2025] Open
Abstract
Recent advancements in antibody-drug conjugates (ADCs) targeting trophoblast surface cell antigen 2 (Trop-2) have brought important progress in the field of targeted therapy. This progress also holds promise for the treatment of small cell lung cancer (SCLC) as anti-Trop-2 therapy appears to have a safe and effective clinical activity in metastatic SCLC patients. However, effective treatments of anti-Trop-2 ADCs rely on the comprehensive assessment of Trop-2 expression at the tumor sites, SCLC exhibits intratumoral heterogeneity, making the accurate acquisition of histological biopsies a challenge. To address this issue, we herein report the development of an anti-Trop-2 aptamer consisting of 76 bases is specifically bind to Trop-2-overexpressing SCLC cells. Further truncated anti-Trop-2 aptamer with 46 nucleotides also possesses excellent in vitro and in vivo binding affinity with Trop-2 antigens. After radiolabeling with gallium-68 radionuclide, an aptamer-based molecular imaging probe was successfully fabricated named [68Ga]Ga-NOTA-TRP-c. This imaging probe demonstrated effective and precise differentiation of Trop-2-positive tumors in both murine- and human-derived animal models, exhibiting favorable metabolic profiles. Furthermore, Trop-2-positive SCLC tumors recognized by anti-Trop-2 aptamer can be treated with anti-Trop-2 ADC sacituzumab govitecan (SG), either in vitro or in vivo. Importantly, SG induces DNA damage and cell apoptosis without affecting the expression of Trop-2 on the cell surface, which makes it possible to use anti-Trop-2 aptamer to monitor the expression of Trop-2 in SCLC. This study highlights the potential of aptamer-based molecular imaging and imaging-guided SG treatment as a promising option for targeted therapy in SCLC.
Collapse
Affiliation(s)
- Yamei Chen
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China
| | - Xuwei Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Yang Sun
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China
| | - Keying Liu
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China
| | - Ding Ding
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China.
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China.
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China.
| |
Collapse
|
21
|
Liu L, Wu P, Wang B, Dong J, Zhang C, Liu W, Ying J. Exploring the role of TIGIT in patients with Small Cell Lung Cancer as a novel predictor of prognosis and immunotherapy response. Cancer Immunol Immunother 2025; 74:134. [PMID: 40035834 PMCID: PMC11880484 DOI: 10.1007/s00262-025-03985-6] [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: 09/11/2024] [Accepted: 02/17/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domains (TIGIT) is a novel immune checkpoint playing a crucial role in immunosuppression and immune evasion. This study aims to elucidate the expression patterns, characteristics, and possible mechanisms of TIGIT in small cell lung cancer (SCLC). METHODS TIGIT expression was analyzed across various cancers and normal tissues using The Cancer Genome Atlas (TCGA). Transcriptomic data from SCLC patients, sourced from the Gene Expression Omnibus (GEO) and literature, were analyzed to assess TIGIT-related characteristics. Immunohistochemistry (IHC) was used to verify TIGIT expression in post-surgical and advanced SCLC samples, focusing on expression characteristics, prognostic value, and treatment response. RESULTS TIGIT was significantly overexpressed in various tumors, including SCLC (p < 0.05). Higher expression was associated with better overall survival (OS) (p < 0.05). Notably, a significant positive correlation was observed between TIGIT expression and immune-related metagenes, such as HCK, interferon, and LCK (p < 0.05). Immune infiltration analysis revealed a strong positive correlation between TIGIT expression and immune score in multiple cohorts. Additionally, TIGIT expression correlated positively with immune cells, including CD8 T cells, cytotoxic lymphocytes, and B cells (p < 0.05), and multiple immune checkpoints like BTLA, ICOS, and LAG3 (p < 0.05), while it had a significant negative correlation with the TIDE score (p < 0.05). In the validation section, patients with high TIGIT expression showed significantly prolonged disease-free survival (DFS) and OS (p < 0.05), and demonstrated a better response to adjuvant chemotherapy (ACT) and immunotherapy. CONCLUSION TIGIT serves as a biomarker in SCLC, with its high expression indicating favorable prognosis and treatment response. These effects may be due to TIGIT's unique immune landscape and its association with other immune checkpoints.
Collapse
Affiliation(s)
- Li Liu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peng Wu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Bingzhi Wang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jiyan Dong
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chaoqi Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenchao Liu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
22
|
Wu H, Gu Y, Xu L. Metallothionein 2A enhances the yes-associated protein 1 signaling pathway to promote small-cell lung cancer metastasis. Cytojournal 2025; 22:25. [PMID: 40260063 PMCID: PMC12010882 DOI: 10.25259/cytojournal_201_2024] [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: 09/26/2024] [Accepted: 01/10/2025] [Indexed: 04/23/2025] Open
Abstract
Objective Small-cell lung cancer (SCLC) remains challenging to treat due to its high invasiveness and propensity for drug resistance. Evidence suggests that the regulatory relationship between metallothionein 2A (MT2A) and the yes-associated protein 1 (YAP1) signaling pathway may influence the development of SCLC. Therefore, this study aims to explore the potential mechanisms affecting SCLC progression based on the regulatory interaction between YAP1 and MT2A. Material and Methods This study utilized reverse transcription quantitative polymerase chain reaction and Western blot analysis to analyze MT2A expression in cells. SCLC cell models with MT2A silencing and overexpression, as well as cotransfected cell models with YAP1 silencing and MT2A overexpression, were constructed. The effect of MT2A/YAP1 on cell growth, migration, and invasion was evaluated through a series of experiments, including cell viability assessment using cell counting kit-8 assay, colony formation examination, 5-ethynyl-2'-deoxyuridine staining, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining, and Transwell analysis. In addition, Western blot analysis was conducted to investigate alterations in crucial proteins associated with the YAP1 pathway and the epithelial-mesenchymal transition ( EMT) markers influenced by MT2A/YAP1. Lung metastasis and Ki67 expression were analyzed through hematoxylin and eosin staining and immunofluorescence analysis in vivo. Results In the SCLC cell line ( NCI-H69 cells), MT2A exhibits increased expression, facilitating cell growth, migration, and invasion. YAP1 expression decreases when MT2A is depleted. In addition, our findings validate that MT2A facilitates EMT progression and SCLC invasion and metastasis by upregulating YAP1 expression. In vitro, silencing MT2A inhibits lung metastasis and Ki67 expression. Conclusion MT2A facilitates the migration and invasion of SCLC cells by influencing the YAP1 signaling cascade. This investigation offers a fresh avenue for delving deeply into the potential mechanisms involved in the progression of SCLC.
Collapse
Affiliation(s)
- Hong Wu
- Department of Respiratory Medicine, The Second Hospital of Jiaxing, Jiaxing, China
| | - Yangyang Gu
- Department of Respiratory Medicine, The Second Hospital of Jiaxing, Jiaxing, China
| | - Lidong Xu
- Department of Cardiothoracic Surgery, The Second Hospital of Jiaxing, Jiaxing, China
| |
Collapse
|
23
|
Ramirez-Torres EE, Castañeda ARS, Rández L, Sisson SA, Cabrales LEB, Montijano JI. Proper likelihood functions for parameter estimation in S-shaped models of unperturbed tumor growth. Sci Rep 2025; 15:6598. [PMID: 39994407 PMCID: PMC11850645 DOI: 10.1038/s41598-025-91146-1] [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: 02/29/2024] [Accepted: 02/18/2025] [Indexed: 02/26/2025] Open
Abstract
The analysis of unperturbed tumor growth kinetics, particularly the estimation of parameters for S-shaped equations used to describe growth, requires an appropriate likelihood function that accounts for the increasing error in solid tumor measurements as tumor size grows over time. This study aims to propose suitable likelihood functions for parameter estimation in S-shaped models of unperturbed tumor growth. Five different likelihood functions are evaluated and compared using three Bayesian criteria (the Bayesian Information Criterion, Deviance Information Criterion, and Bayes Factor) along with hypothesis tests on residuals. These functions are applied to fit data from unperturbed Ehrlich, fibrosarcoma Sa-37, and F3II tumors using the Gompertz equation, though they are generalizable to other S-shaped growth models for solid tumors or analogous systems (e.g., microorganisms, viruses). Results indicate that error models with tumor volume-dependent dispersion outperform standard constant-variance models in capturing the variability of tumor measurements, particularly the Thres model, which provides interpretable parameters for tumor growth. Additionally, constant-variance models, such as those assuming a normal error distribution, remain valuable as complementary benchmarks in analysis. It is concluded that models incorporating volume-dependent dispersion are preferred for accurate and clinically meaningful tumor growth modeling, whereas constant-dispersion models serve as useful complements for consistency and historical comparability.
Collapse
Affiliation(s)
- Erick E Ramirez-Torres
- Instituto Universitario de Investigación de Matemáticas y Aplicaciones, Universidad de Zaragoza, Zaragoza, Spain
- Departamento de Biomédica, Facultad de Ingeniería en Telecomunicaciones, Informática y Biomédica, Universidad de Oriente, Santiago de Cuba, Cuba
| | - Antonio R Selva Castañeda
- Instituto Universitario de Investigación de Matemáticas y Aplicaciones, Universidad de Zaragoza, Zaragoza, Spain
| | - Luis Rández
- Instituto Universitario de Investigación de Matemáticas y Aplicaciones, Universidad de Zaragoza, Zaragoza, Spain
| | - Scott A Sisson
- UNSW Data Science Hub, and School of Mathematics and Statistics, University of New South Wales, Sydney, Australia
| | - Luis E Bergues Cabrales
- Departamento de Investigación e Innovación, Centro Nacional de Electromagnetismo Aplicado, Universidad de Oriente, Santiago de Cuba, Cuba.
| | - Juan I Montijano
- Instituto Universitario de Investigación de Matemáticas y Aplicaciones, Universidad de Zaragoza, Zaragoza, Spain.
| |
Collapse
|
24
|
Diao S, Wan Y, Huang D, Huang S, Sadiq T, Khan MS, Hussain L, Alkahtani BS, Mazhar T. Optimizing Bi-LSTM networks for improved lung cancer detection accuracy. PLoS One 2025; 20:e0316136. [PMID: 39992919 PMCID: PMC11849851 DOI: 10.1371/journal.pone.0316136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/05/2024] [Indexed: 02/26/2025] Open
Abstract
Lung cancer remains a leading cause of cancer-related deaths worldwide, with low survival rates often attributed to late-stage diagnosis. To address this critical health challenge, researchers have developed computer-aided diagnosis (CAD) systems that rely on feature extraction from medical images. However, accurately identifying the most informative image features for lung cancer detection remains a significant challenge. This study aimed to compare the effectiveness of both hand-crafted and deep learning-based approaches for lung cancer diagnosis. We employed traditional hand-crafted features, such as Gray Level Co-occurrence Matrix (GLCM) features, in conjunction with traditional machine learning algorithms. To explore the potential of deep learning, we also optimized and implemented a Bidirectional Long Short-Term Memory (Bi-LSTM) network for lung cancer detection. The results revealed that the highest performance using hand-crafted features was achieved by extracting GLCM features and utilizing Support Vector Machine (SVM) with different kernels, reaching an accuracy of 99.78% and an AUC of 0.999. However, the deep learning Bi-LSTM network surpassed both methods, achieving an accuracy of 99.89% and an AUC of 1.0000. These findings suggest that the proposed methodology, combining hand-crafted features and deep learning, holds significant promise for enhancing early lung cancer detection and ultimately improving diagnosis systems.
Collapse
Affiliation(s)
- Su Diao
- Department of Industrial & Systems Engineering, Auburn University, Auburn, Alabama, United States of America
| | - Yajie Wan
- Department of Computer Science, Brown University, Providence, RI, United States of America
| | - Danyi Huang
- Department of Chemical Engineering, Columbia University, New York City, NY, United States of America
| | - Shijia Huang
- Fu Foundation School of Engineering and Applied Science, Fu Foundation School of Engineering and Applied Science, Columbia University, New York, NY, United States of America
| | - Touseef Sadiq
- Department of Information and Communication Technology, Centre for Artificial Intelligence Research (CAIR), University of Agder, Grimstad, Norway
| | | | - Lal Hussain
- Department of Computer Science and Information Technology, The University of Azad Jammu and Kashmir, Chattar Kalas Campus, Muzaffarabad, Pakistan
- Department of Computer Science, Neelum Campus, The University of Azad Jammu and Kashmir, Azad Kashmir, Pakistan
| | - Badr S. Alkahtani
- Department of Mathematics, King Saud University, Riyadh, Saudi Arabia
| | - Tehseen Mazhar
- School of Computer Science, National College of Business Administration and Economics, Lahore, Pakistan
- Department of Computer Science and Information Technology, School Education Department, Government of Punjab, Layyah, Pakistan
| |
Collapse
|
25
|
Gao R, Wu P, Yin X, Zhuang L, Meng X. Deep analysis of the trials and major challenges in the first-line treatment for patients with extensive-stage small cell lung cancer. Int Immunopharmacol 2025; 148:114116. [PMID: 39847950 DOI: 10.1016/j.intimp.2025.114116] [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: 05/08/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 01/25/2025]
Abstract
The median overall survival (OS) is approximately 10 months when chemotherapy alone is the first-line treatment for extensive-stage small cell lung cancer (ES-SCLC). The approval of the two PD-L1 inhibitors, atezolizumab and durvalumab, marked the beginning of the immunotherapy era for ES-SCLC. Serplulimab, as the first PD-1 inhibitor to achieve success in the first-line treatment of ES-SCLC, has not only demonstrated significant improvements in patient survival outcomes but also ushered in a new era for PD-1 inhibitors in the treatment of ES-SCLC. Recently, antiangiogenic agents with chemo-immunotherapy have achieved breakthroughs in first-line ES-SCLC treatment. Improving the clinical benefits of individualized treatment for patients with ES-SCLC remains challenging. Challenges include identifying biomarkers for targeted therapy, exploring new treatments, developing new medicines, and classifying SCLC molecular subtypes. This review provides an in-depth analysis of research on first-line ES-SCLC treatment. Additionally, it discusses advances in ES-SCLC treatment.
Collapse
Affiliation(s)
- Ran Gao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China
| | - Peizhu Wu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China
| | - Xiaoyan Yin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China
| | - Lulu Zhuang
- Cheeloo College of Cancer Center, Shandong University, Jinan, Shandong, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China.
| |
Collapse
|
26
|
Benariba MA, Hannachi K, Wang S, Zhang Y, Wang X, Wang L, Zhou N. Liposome-encapsulated lambda exonuclease-based amplification system for enhanced detection of miRNA in platelet-derived microvesicles of non-small cell lung cancer. J Mater Chem B 2025; 13:2666-2673. [PMID: 39881659 DOI: 10.1039/d4tb02621g] [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: 01/31/2025]
Abstract
Platelet-derived microvesicles (PMVs) and their encapsulated microRNAs (miRNAs) hold immense potential as biomarkers for early non-small cell lung cancer (NSCLC) diagnosis. This study presents a pioneering liposome-based approach for enhanced miRNA detection within PMVs, employing a lambda exonuclease (λ EXO)-based amplification system encapsulated in immunoliposomes. The platform exploits the novel catalytic functionality of λ EXO, demonstrating its unprecedented capability to catalyze RNA-DNA hybrid substrates. The λ EXO-based amplification system exhibited high sensitivity and specificity in detecting miRNA-21, a key miRNA associated with NSCLC, demonstrating a limit of detection (LOD) of 33.11 fg mL-1. The system was successfully encapsulated within liposomes, which were then functionalized with CD41 antibody to facilitate targeted delivery and fusion with PMVs. The results reveal a significant difference in miRNA-21 levels between PMVs from NSCLC patients and healthy individuals, with a 2.06-fold higher abundance observed in NSCLC patients. This research presents a significant technological advancement in miRNA detection, paving the way for improved early diagnosis and personalized medicine approaches.
Collapse
Affiliation(s)
- Mohamed Aimene Benariba
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
- Bioengineering Laboratory, Ecole Nationale Supérieure de Biotechnologie, Ville Universitaire Ali Mendjeli, BP E66 25100, Constantine, Algeria
| | - Kanza Hannachi
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China.
| | - Sanxia Wang
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| | - Yuting Zhang
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| | - Xiaoli Wang
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| | - Li Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China.
| | - Nandi Zhou
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
27
|
Chen H, Liu L, Xing G, Zhang D, A. N, Huang J, Li Y, Zhao G, Liu M. Exosome tropism and various pathways in lung cancer metastasis. Front Immunol 2025; 16:1517495. [PMID: 40028322 PMCID: PMC11868168 DOI: 10.3389/fimmu.2025.1517495] [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: 10/26/2024] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Lung cancer, characterized by its high morbidity and mortality rates, has the capability to metastasize to various organs, thereby amplifying its detrimental impact and fatality. The metastasis of lung cancer is a complex biological phenomenon involving numerous physiological transformations. Exosomes, small membranous vesicles enriched with biologically active components, are pivotal in mediating intercellular communication and regulating physiological functions due to their specificity and stability. Extensive research has elucidated the production and functions of exosomes in cancer contexts. Multitude of evidence demonstrates a strong association between lung cancer metastasis and exosomes. Additionally, the concept of the pre-metastatic niche is crucial in the metastatic process facilitated by exosomes. This review emphasizes the role of exosomes in mediating lung cancer metastasis and their impact on the disease's development and the progression to other tissues. Furthermore, it explores the potential of exosomes as biomarkers for lung cancer metastasis, offering significant insights for future clinical advancements.
Collapse
Affiliation(s)
- Hui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Lin Liu
- Department of Drug Dispensing, The Third Hospital of Mianyang, Sichuan Mental Health Center, MianYang, China
| | - Gang Xing
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Dan Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Niumuqie A.
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jianlin Huang
- Department of Pharmacy, Luzhou Naxi District People’s Hospital, Luzhou, China
| | - Yaling Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ge Zhao
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Minghua Liu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
28
|
Zhang S, Liu K, Liu Y, Hu X, Gu X. The role and application of bioinformatics techniques and tools in drug discovery. Front Pharmacol 2025; 16:1547131. [PMID: 40017606 PMCID: PMC11865229 DOI: 10.3389/fphar.2025.1547131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
The process of drug discovery and development is both lengthy and intricate, demanding a substantial investment of time and financial resources. Bioinformatics techniques and tools can not only accelerate the identification of drug targets and the screening and refinement of drug candidates, but also facilitate the characterization of side effects and the prediction of drug resistance. High-throughput data from genomics, transcriptomics, proteomics, and metabolomics make significant contributions to mechanics-based drug discovery and drug reuse. This paper summarizes bioinformatics technologies and tools in drug research and development and their roles and applications in drug research and development, aiming to provide references for the development of new drugs and the realization of precision medicine.
Collapse
Affiliation(s)
- Shujun Zhang
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Kaijie Liu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Yafeng Liu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Xinjun Hu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
- Henan Medical Key Laboratory of Gastrointestinal Microecology and Hepatology, Luoyang, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| |
Collapse
|
29
|
Calles A, Navarro A, Doger de Speville Uribe BG, Colomé EÁ, de Miguel M, Álvarez R, Arregui M, Moreno V, Rocha P, Calvo E, Ramon-Patino J, Corral de la Fuente E, Alcalá-López D, Boix O, Fernández-Pinto M, Rodríguez-Morató J, Palmero R, Nadal E, Jove M, Felip E. Lurbinectedin Plus Pembrolizumab in Relapsed SCLC: The Phase I/II LUPER Study. J Thorac Oncol 2025:S1556-0864(25)00064-4. [PMID: 39938593 DOI: 10.1016/j.jtho.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/30/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
INTRODUCTION SCLC has limited second-line treatment options after chemotherapy. We assessed the efficacy and safety of lurbinectedin combined with pembrolizumab in relapsed SCLC patients who had not received prior immunotherapy, aiming to prevent early progression and achieve sustained responses. METHODS The LUPER trial (NCT04358237) is a phase I/II, single-arm, open-label, multicenter study. Phase I established the recommended phase II dose. The primary endpoint of phase II was the investigator-confirmed objective response rate. Secondary endpoints included duration of response, progression-free survival (PFS), overall survival (OS), and safety. Patients were categorized as platinum-sensitive (chemotherapy-free interval ≥ 90 d) or platinum-resistant (<90 d). RESULTS The recommended phase II dose was 3.2 mg/m2 lurbinectedin and 200 mg pembrolizumab IV every three weeks. Phase II included 28 patients, 50% of whom were platinum-resistant. The objective response rate was 46.4% (95% confidence interval: 27.5-66.1, p < 0.001), including three complete responses, with two complete metabolic responses post-treatment completion at 35 cycles. The median duration of response was 7.8 months, with 40% of patients maintaining responses for 12 months or longer. The median PFS was 4.6 months, and the median OS was 10.5 months. Platinum-sensitive patients had significantly better PFS (8.0 versus 2.8 mo, p = 0.012) and numerically superior OS (15.7 versus 7.1 mo, p = 0.058). Grade 3 or higher treatment-related adverse events occurred in 71.4% of patients, with transient neutropenia being the most common. Immune-related adverse events were consistent with prior pembrolizumab studies. CONCLUSIONS Lurbinectedin plus pembrolizumab reported promising efficacy in relapsed SCLC, particularly for platinum-sensitive patients, with a known and manageable safety profile. These results support further exploration of this combination in SCLC treatment.
Collapse
Affiliation(s)
- Antonio Calles
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.
| | - Alejandro Navarro
- Medical Oncology Department, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR), Barcelona (Spain), Ridgewood, New Jersey
| | | | - Enric Álvarez Colomé
- Medical Oncology Department, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - María de Miguel
- START Madrid-CIOCC, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Rosa Álvarez
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Marta Arregui
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Víctor Moreno
- START Madrid-FJD, Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Pedro Rocha
- Medical Oncology Department, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Emiliano Calvo
- START Madrid-CIOCC, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Jorge Ramon-Patino
- START Madrid-CIOCC, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | | | - Daniel Alcalá-López
- Medica Scientia Innovation Research (MEDSIR), Barcelona (Spain), Ridgewood, New Jersey
| | - Olga Boix
- Medica Scientia Innovation Research (MEDSIR), Barcelona (Spain), Ridgewood, New Jersey
| | | | - Jose Rodríguez-Morató
- Medica Scientia Innovation Research (MEDSIR), Barcelona (Spain), Ridgewood, New Jersey
| | - Ramón Palmero
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Barcelona, Spain
| | - Ernest Nadal
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Barcelona, Spain
| | - Maria Jove
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Barcelona, Spain
| | - Enriqueta Felip
- Medical Oncology Department, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
30
|
Nabipur L, Mouawad M, Venketaraman V. Therapeutic Applications of Programmed Death Ligand 1 Inhibitors in Small Cell Lung Cancer. Biomedicines 2025; 13:401. [PMID: 40002814 PMCID: PMC11852381 DOI: 10.3390/biomedicines13020401] [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: 12/26/2024] [Revised: 01/29/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Small cell lung cancer (SCLC) is an aggressive cancer with rapid progression, limited treatment success, and high relapse rates. Chemotherapy and radiation are standard treatments but often result in chemoresistance. PD-L1 inhibitors have gained attention for their role in enhancing tumor immunity. Methods: This review summarizes clinical trials involving PD-L1 inhibitors, such as atezolizumab, durvalumab, pembrolizumab, and nivolumab, in SCLC treatment. Key trials include IMpower133, CASPIAN, KEYNOTE-604, and CheckMate 331, focusing on survival outcomes and treatment efficacy. Results: Studies such as IMpower133 and CASPIAN demonstrate improved overall survival when PD-L1 inhibitors were added to platinum-based chemotherapy. However, outcomes in trials such as KEYNOTE-604 and CheckMate 331 varied, showing the need for refined patient selection. Adverse events (AEs) associated with these treatments were also noted. PD-L1 inhibitors offer promise in SCLC treatment, but efficacy varies across trials and patient groups. Future research should focus on better patient selection and overcoming resistance mechanisms. Addressing immune-related AEs is essential for optimizing treatment strategies.
Collapse
Affiliation(s)
| | | | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (L.N.); (M.M.)
| |
Collapse
|
31
|
Chen L, Qi T, Zhang B, Wang X, Zheng M. NT5E (CD73) as a prognostic biomarker and therapeutic target associated with immune infiltration in lung adenocarcinoma. Sci Rep 2025; 15:4340. [PMID: 39910337 PMCID: PMC11799229 DOI: 10.1038/s41598-025-88964-8] [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: 10/06/2024] [Accepted: 02/03/2025] [Indexed: 02/07/2025] Open
Abstract
Lung adenocarcinoma (LUAD), the most common type of lung cancer, is a leading cause of cancer-related mortality. NT5E, an ecto-5'-nucleotidase enzyme, has been implicated in cancer progression, particularly in efferocytosis. Despite its potential involvement, the prognostic significance of NT5E and relationship with immune cell infiltration in LUAD have not been extensively explored. In this study, we performed a comprehensive analysis to elucidate the expression patterns of NT5E and its prognostic implications in LUAD using data from diverse public databases. Multiple computational algorithms, including CIBERSORT, ESTIMATE, and xCell, were employed to assess the correlation between NT5E expression and immune cell infiltration. We found that NT5E was significantly overexpressed at both the mRNA and protein levels in LUAD tissues. Elevated NT5E expression was significantly linked to multiple clinicopathological factors, including metastasis and pathological stage, and served as a strong predictor of poor prognosis in LUAD patients. Gene Set Enrichment Analysis (GSEA) indicated that NT5E plays a crucial role in regulating immune responses, as evidenced by differential gene expression associated with NT5E levels. A strong positive correlation was observed between NT5E expression and the presence of immune cells, including dendritic cells, macrophages, and CD4+ T cells, as well as the expression of various immune cell markers, suggesting that NT5E may influence the prognosis of LUAD patients by regulating immune cell infiltration. Additionally, drug sensitivity analysis highlights the potential of selumetinib and PD318088, both MEK1/2 inhibitors, to target NT5E in LUAD treatment, suggesting their use as single agents or in combination with other therapies. Collectively, these findings establish NT5E as a promising prognostic biomarker and therapeutic target in LUAD, particularly in the context of immune cell infiltration.
Collapse
Affiliation(s)
- Leyan Chen
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Tuoya Qi
- Jinshan Hospital, Fudan University, Shanghai, China
| | - Bishu Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | - Mingfeng Zheng
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China.
- Wuxi Medical Center, Nanjing Medical University, Wuxi, China.
| |
Collapse
|
32
|
Qin L, Li Y, Liu J, An X. Advancements in cellular immunotherapy: overcoming resistance in lung and colorectal cancer. Front Immunol 2025; 16:1554256. [PMID: 39975543 PMCID: PMC11835964 DOI: 10.3389/fimmu.2025.1554256] [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: 01/01/2025] [Accepted: 01/17/2025] [Indexed: 02/21/2025] Open
Abstract
Immunotherapy has revolutionized cancer treatment, offering hope for patients with otherwise treatment-resistant tumors. Among the most promising approaches are cellular therapies, particularly chimeric antigen receptor T-cell (CAR-T) therapy, which has shown remarkable success in hematologic malignancies. However, the application of these therapies to solid tumors, such as lung and colorectal cancers, has faced significant challenges. Tumor resistance mechanisms-ranging from immune evasion, antigen loss, and immune checkpoint upregulation, to tumor microenvironment immunosuppression-remain major obstacles. This mini-review highlights the latest advancements in tumor immunotherapy, with a focus on cellular therapies, and addresses the resistance mechanisms that hinder their effectiveness in lung and colorectal cancers. We examine the evolution of CAR-T cell therapy, as well as the potential of engineered natural killer (NK) cells and macrophages in solid tumor treatment. The review also explores cutting-edge strategies aimed at overcoming resistance, including combination therapies, gene editing technologies, and nanotechnology for targeted drug delivery. By discussing the molecular, cellular, and microenvironmental factors contributing to resistance, we aim to provide a comprehensive overview of how these challenges can be overcome, paving the way for more effective, personalized immunotherapies in lung and colorectal cancer treatment.
Collapse
Affiliation(s)
- Lijuan Qin
- Department of Radiotherapy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yuan Li
- Department of Respiratory Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Juan Liu
- Department of Special needs Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoqin An
- Department of Respiratory Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
33
|
Iñañez A, del Rey-Vergara R, Quimis F, Rocha P, Galindo M, Menéndez S, Masfarré L, Sánchez I, Carpes M, Martínez C, Pérez-Buira S, Rojo F, Rovira A, Arriola E. The Potential of Single-Transcription Factor Gene Expression by RT-qPCR for Subtyping Small Cell Lung Cancer. Int J Mol Sci 2025; 26:1293. [PMID: 39941061 PMCID: PMC11818609 DOI: 10.3390/ijms26031293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/25/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Complex RNA-seq signatures involving the transcription factors ASCL1, NEUROD1, and POU2F3 classify Small Cell Lung Cancer (SCLC) into four subtypes: SCLC-A, SCLC-N, SCLC-P, and SCLC-I (triple negative or inflamed). Preliminary studies suggest that identifying these subtypes can guide targeted therapies and potentially improve outcomes. This study aims to evaluate whether the expression levels of these three key transcription factors can effectively classify SCLC subtypes, comparable to the use of individual antibodies in immunohistochemical (IHC) analysis of formalin-fixed, paraffin-embedded (FFPE) tumor samples. We analyzed preclinical models of increasing complexity, including eleven human and five mouse SCLC cell lines, six patient-derived xenografts (PDXs), and two circulating tumor cell (CTC)-derived xenografts (CDXs) generated in our laboratory. RT-qPCR conditions were established to detect the expression levels of ASCL1, NEUROD1, and POU2F3. Additionally, protein-level analysis was performed using Western blot for cell lines and IHC for FFPE samples of PDX and CDX tumors, following our experience with patient tumor samples from the CANTABRICO trial (NCT04712903). We found that the analyzed SCLC cell line models predominantly expressed ASCL1, NEUROD1, and POU2F3, or showed no expression, as identified by RT-qPCR, consistently matching the previously assigned subtypes for each cell line. The classification of PDX and CDX models demonstrated consistency between RT-qPCR and IHC analyses of the transcription factors. Our results show that single-gene analysis by RT-qPCR from FFPE-extracted RNA simplifies SCLC subtype classification. This approach provides a cost-effective alternative to IHC staining or expensive multi-gene RNA sequencing panels, making SCLC subtyping more accessible for both preclinical research and clinical applications.
Collapse
Affiliation(s)
- Albert Iñañez
- Cancer Research Program, Hospital del Mar Research Institute, 08003 Barcelona, Spain; (A.I.)
| | - Raúl del Rey-Vergara
- Cancer Research Program, Hospital del Mar Research Institute, 08003 Barcelona, Spain; (A.I.)
| | - Fabricio Quimis
- Cancer Research Program, Hospital del Mar Research Institute, 08003 Barcelona, Spain; (A.I.)
| | - Pedro Rocha
- Department of Medical Oncology, Hospital del Mar, 08003 Barcelona, Spain
| | - Miguel Galindo
- Cancer Research Program, Hospital del Mar Research Institute, 08003 Barcelona, Spain; (A.I.)
| | - Sílvia Menéndez
- Cancer Research Program, Hospital del Mar Research Institute, 08003 Barcelona, Spain; (A.I.)
| | - Laura Masfarré
- Cancer Research Program, Hospital del Mar Research Institute, 08003 Barcelona, Spain; (A.I.)
- Department of Medical Oncology, Hospital del Mar, 08003 Barcelona, Spain
| | - Ignacio Sánchez
- Department of Pathology, Hospital del Mar, 08003 Barcelona, Spain
| | - Marina Carpes
- Instituto Murciano de Investigación Biosanitaria IMIB-Pascual Parrilla, Pathology Core, 30120 Murcia, Spain
| | - Carlos Martínez
- Instituto Murciano de Investigación Biosanitaria IMIB-Pascual Parrilla, Pathology Core, 30120 Murcia, Spain
- Department of Anatomy and Comparative Pathology, Facultad de Veterinaria, Universidad de Murcia, 30100 Murcia, Spain
| | - Sandra Pérez-Buira
- Department of Pathology, IIS-Fundación Jiménez Díaz-CIBERONC, 28040 Madrid, Spain
| | - Federico Rojo
- Department of Pathology, IIS-Fundación Jiménez Díaz-CIBERONC, 28040 Madrid, Spain
| | - Ana Rovira
- Cancer Research Program, Hospital del Mar Research Institute, 08003 Barcelona, Spain; (A.I.)
- Department of Medical Oncology, Hospital del Mar, 08003 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Edurne Arriola
- Cancer Research Program, Hospital del Mar Research Institute, 08003 Barcelona, Spain; (A.I.)
- Department of Medical Oncology, Hospital del Mar, 08003 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
34
|
Mao G, Liu J. CALML3-AS1 enhances malignancies and stemness of small cell lung cancer cells through interacting with DAXX protein and promoting GLUT4-mediated aerobic glycolysis. Toxicol Appl Pharmacol 2025; 495:117177. [PMID: 39617259 DOI: 10.1016/j.taap.2024.117177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
The lncRNA CALML3 antisense RNA 1 (CALML3-AS1) is a biomarker for various cancers, including non-small cell lung cancer (NSCLC). However, the role of CALM3-AS1 in small cell lung cancer (SCLC) is still unclear. Here, we found that the CALML3-AS1 was upregulated in SCLC tissues and cells. SCLC cells (NCI-H69 and NCI-H466 cells) were transfected with small interfering RNA of CALML-AS1 (si-CALML3-AS1) and Death domain-associated protein (DAXX) (si-DAXX) or an overexpression vector of CALML-AS1 (dCas9-CALML3-AS1) and DAXX (dCas9-DAXX). The results showed that silencing CALML3-AS1 inhibited SCLC cell proliferation, colony formation, migration, invasion, and spheroid formation, and reduced the expression of stemness marker proteins (Nanog. Oct4, and Lin28). Moreover, silencing CALML3-AS1 reduced glycolysis rate, glucose utilization, and lactate production, and decreased the levels of key glycolytic regulatory proteins (GLUT1, GLUT4, HK2, and PKM2) in SCLC cells, while overexpression of CALML3-AS1 promoted malignant growth and stemness and enhanced glucose transporters type 4 (GLUT4)-mediated aerobic glycolysis by interacting with DAXX in NCI-H69 and NCI-H466 cells. Silencing DAXX or GLUT4, or treatment with 2-Deoxy-d-glucose (2-DG, a glycolysis inhibitor) reversed the effects of CALML3-AS1 overexpression on aerobic glycolysis, malignant growth, and stemness of SCLC cells. Finally, NCI-H69 cells transfected with CALML3-AS1, sh-CALML3-AS1, and sh-DAXX lentiviral vectors were subcutaneously injected into nude mice to construct xenograft models. Knockdown of CALML3-AS1 or DAXX inhibited tumor growth in SCLC in vivo. In conclusion, CALML3-AS1, an oncogene, promotes the malignancy and stemness of SCLC cells by interacting with DAXX to enhance GLUT4-mediated aerobic glycolysis, thereby promoting SCLC progression.
Collapse
Affiliation(s)
- Guangxian Mao
- Peking University Shenzhen Hospital Medical College, Anhui Medical University, Shenzhen 518036, People's Republic of China; Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, People's Republic of China
| | - Jixian Liu
- Peking University Shenzhen Hospital Medical College, Anhui Medical University, Shenzhen 518036, People's Republic of China; Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, People's Republic of China.
| |
Collapse
|
35
|
Zhang T, Han Z, Hou S, Song Y, Zhang Y, Wang M. Metastatic Small-Cell Lung Carcinoma Infiltrating the Heart: A Rare Case Diagnosed Using Imaging Data. JOURNAL OF CLINICAL ULTRASOUND : JCU 2025; 53:385-387. [PMID: 39364805 DOI: 10.1002/jcu.23853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 09/06/2024] [Indexed: 10/05/2024]
Abstract
Small-cell lung carcinoma is a high-grade aggressive disease that occurs most commonly in bronchial lung cancer, and metastasis to the heart is extremely rare. The diagnosis of metastatic small-cell lung carcinoma infiltrating the heart remains challenging because of the variability of its clinical presentation. Hereinafter, we reported a case of a 41-year-old man who suffered from small-cell lung cancer that invaded the pulmonary artery. The patient presented with chest tightness, dry cough, and deterioration in exercise tolerance and diagnosed at his imaging data, who had an uneventful recovery after surgical resection of the masses.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Ultrasound, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhengyang Han
- Department of Ultrasound, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Suyun Hou
- Department of Ultrasound, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Song
- Department of Ultrasound, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yongxiang Zhang
- Department of Ultrasound, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Menghe Wang
- Department of Ultrasound, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
36
|
Umemura S, Udagawa H, Ikeda T, Murakami H, Daga H, Toyozawa R, Kozuki T, Sakakibara-Konishi J, Ohe Y, Morise M, Kato T, Shingyoji M, Hara S, Furuya N, Teranishi S, Takata S, Miyamoto S, Nakachi I, Wakabayashi M, Nomura S, Sato A, Ishii G, Tsuchihara K, Sugiyama E, Kirita K, Sakai T, Shibata Y, Izumi H, Nosaki K, Zenke Y, Matsumoto S, Yoh K, Niho S, Goto K. Clinical Significance of a Prospective Large Genomic Screening for SCLC: The Genetic Classification and a Biomarker-Driven Phase 2 Trial of Gedatolisib. J Thorac Oncol 2025; 20:177-193. [PMID: 39395663 DOI: 10.1016/j.jtho.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 09/17/2024] [Accepted: 10/05/2024] [Indexed: 10/14/2024]
Abstract
INTRODUCTION SCLC has been treated as a single entity resulting in limited survival improvement. Developing effective tools for guiding appropriate therapeutic strategies is crucial. METHODS A total of 1035 SCLCs were prospectively analyzed by a genomic screening platform: LC-SCRUM-Asia. Fresh frozen tumor samples were subjected to a next-generation sequencing system enabling the integrative analysis of cancer-related genes. A phase 2 trial of gedatolisib for SCLC with PI3K/AKT/mTOR pathway mutations was conducted based on this screening. RESULTS On the basis of the treatment outcomes and therapeutic targets, the following five distinct genetic subgroups were identified in SCLC: NSCLC-subgroup (genetic alterations associated with NSCLC, 8.5%); Hotspot-subgroup (targetable hotspot mutations common in tumors, 3.0%); PI3K-subgroup (PI3K/AKT/mTOR pathway mutations, 7.4%); MYC-subgroup (MYC family amplifications, 13.0%); and HME-subgroup (mutations in the histone-modifying enzymes, 17.6%). The NSCLC-subgroup (hazard ratio = 1.57; 95% confidence interval: 1.22-2.03) and MYC-subgroup (hazard ratio = 1.56; 95% confidence interval: 1.26-1.93) had significantly shorter progression-free survivals after first-line platinum-based treatment. The Hotspot-subgroup and MYC-subgroup were candidates for novel targeted therapies. The HME-subgroup had a favorable survival in patients who received programmed cell death (ligand) 1 inhibitor-based therapies (p = 0.005, log-rank test) regardless of some overlap with other subgroups. There were 15 patients enrolled into the phase 2 trial of gedatolisib in the PI3K-subgroup, and the overall response rate and the disease control rate were 6.7% and 20%, respectively. The MYC-subgroup or NSCLC-subgroup was associated with unfavorable clinical outcomes in this trial. CONCLUSIONS Molecular classification of SCLC by genetic approach is beneficial for predicting the treatment outcomes and effectively guiding the clinical choices.
Collapse
Affiliation(s)
- Shigeki Umemura
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hibiki Udagawa
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takaya Ikeda
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Haruyasu Murakami
- Division of Thoracic Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Haruko Daga
- Department of Medical Oncology, Osaka City General Hospital, Osaka, Japan
| | - Ryo Toyozawa
- Department of Thoracic Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Toshiyuki Kozuki
- Department of Thoracic Oncology and Medicine, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Jun Sakakibara-Konishi
- Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Masahiro Morise
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Terufumi Kato
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | | | - Satoshi Hara
- Department of Respiratory Medicine, Itami City Hospital, Itami, Japan
| | - Naoki Furuya
- Division of Respiratory Medicine, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Shuhei Teranishi
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Saori Takata
- Department of Respiratory Medicine, Kyorin University School of Medicine, Mitaka, Japan
| | - Shingo Miyamoto
- Department of Medical Oncology, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Ichiro Nakachi
- Department of Internal Medicine, Saiseikai Utsunomiya Hospital, Utsunomiya, Japan
| | - Masashi Wakabayashi
- Clinical Research Support Office, National Cancer Center Hospital East, Chiba, Japan
| | - Shogo Nomura
- Clinical Research Support Office, National Cancer Center Hospital East, Chiba, Japan; Department of Biostatistics and Bioinformatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akihiro Sato
- Clinical Research Support Office, National Cancer Center Hospital East, Chiba, Japan
| | - Genichiro Ishii
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Chiba, Japan
| | - Katsuya Tsuchihara
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Eri Sugiyama
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Keisuke Kirita
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tetsuya Sakai
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yuji Shibata
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hiroki Izumi
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kaname Nosaki
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshitaka Zenke
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shingo Matsumoto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kiyotaka Yoh
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Seiji Niho
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Koichi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| |
Collapse
|
37
|
Shen Y, Liu Z, Chen Y, Shi X, Dong S, Wang B. Candidate Biomarker of Response to Immunotherapy In Small Cell Lung Cancer. Curr Treat Options Oncol 2025; 26:73-83. [PMID: 39841387 DOI: 10.1007/s11864-025-01292-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2025] [Indexed: 01/23/2025]
Abstract
OPINION STATEMENT Small-cell lung cancer accounts for about 15% of lung cancers with an extremely poor prognosis. The incorporation of immunotherapy to platinum-based chemotherapy offers sustained overall survival benefits and become the standard for the first-line setting of extensive-stage small-cell lung cancer. However, only a limited number of patients derive prolonged benefits. Although novel immunomodulatory agents and combination strategies are currently under investigation, identifying patients who are likely to obtain clinical benefits from this therapeutic approach is urgently needed. The modest therapeutic response to immunotherapy can be explained by various mechanisms. Traditional biomarkers do not guide immunotherapeutic decision-making in small-cell lung cancer. Notably, recent progress in the understanding of the molecular typing of small-cell lung cancer based on multi-omics data might bring new sights. This review summarizes the potential biomarkers for small-cell lung cancer immunotherapy based on clinical trials and preclinical studies. Moreover, important constraints in identifying biomarkers for small-cell lung cancer treatment are discussed.
Collapse
Affiliation(s)
- Yili Shen
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, Huzhou, Zhejiang, China
- Huzhou Key Laboratory of Precision Diagnosis and Treatment in Respiratory Diseases, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Zhicong Liu
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, Huzhou, Zhejiang, China
- Huzhou Key Laboratory of Precision Diagnosis and Treatment in Respiratory Diseases, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Yi Chen
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, Huzhou, Zhejiang, China
- Huzhou Key Laboratory of Precision Diagnosis and Treatment in Respiratory Diseases, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Xuefei Shi
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, Huzhou, Zhejiang, China.
- Huzhou Key Laboratory of Precision Diagnosis and Treatment in Respiratory Diseases, Huzhou Central Hospital, Huzhou, Zhejiang, China.
| | - Shunli Dong
- Huzhou Key Laboratory of Precision Diagnosis and Treatment in Respiratory Diseases, Huzhou Central Hospital, Huzhou, Zhejiang, China.
- Department of Central Laboratory, Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, Huzhou, Zhejiang, China.
| | - Bin Wang
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, Huzhou, Zhejiang, China.
- Huzhou Key Laboratory of Precision Diagnosis and Treatment in Respiratory Diseases, Huzhou Central Hospital, Huzhou, Zhejiang, China.
| |
Collapse
|
38
|
Yan HX, Zhang YZ, Niu YQ, Wang YW, Liu LH, Tang YP, Huang JM, Leung ELH. Investigating the interaction between calcium signaling and ferroptosis for novel cancer treatment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 137:156377. [PMID: 39798340 DOI: 10.1016/j.phymed.2025.156377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/02/2024] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Drug resistance in cancer is steadily rising, making the development of new therapeutic targets increasingly critical for improving treatment outcomes. PURPOSE The mutual regulation of ions is essential for cell growth. Based on this concept, ion interference strategies offer a highly effective approach for cancer treatment. Calcium ions (Ca2+), as major second messengers, are closely associated with ion exchange and homeostasis. Disruptions in this balance can lead to cell death. However, while iron ions are also crucial, the connection between Ca2+and iron-induced cell death (ferroptosis) has not been well established. Therefore, this study suggests that Ca2+ may play a role in the induction of ferroptosis, presenting a novel and efficient target for cancer therapy. STUDY DESIGN PubMed, Google Scholar, and Web of Science databases were systematically searched for articles published in the past 15 years on the mechanisms of calcium ion-induced ferroptosis in cancer and related drugs. RESULTS The analysis highlights how Ca2+regulate ferroptosis. The mechanisms by which Ca2+influence ferroptosis are summarized based on existing literature, and relevant drugs that act on Ca2+/ferroptosis axis are outlined. CONCLUSION Ca2+ regulate ferroptosis primarily through the modulation of reactive oxygen species (ROS) and glutathione (GSH) levels, a mechanism that applies to a wide range of cancer cells as well as paracancerous and normal cells in cancer treatment. Furthermore, plant-derived active compounds exhibit potent anticancer properties and often act on the Ca2+/ferroptosis axis. These natural compounds could play a significant role in the development of new cancer treatment strategies.
Collapse
Affiliation(s)
- Hao-Xin Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), PR China
| | - Yi-Zhong Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), PR China
| | - Yu-Qing Niu
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau (SAR), China. MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), PR China
| | - Yu-Wei Wang
- Key Laboratory of Shanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shanxi University of Chinese Medicine, Xianyang 712046, Shanxi Province, PR China
| | - Li-Hua Liu
- Economics and Management Yanbian University, Yanji, PR China
| | - Yu-Ping Tang
- Key Laboratory of Shanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shanxi University of Chinese Medicine, Xianyang 712046, Shanxi Province, PR China.
| | - Ju-Min Huang
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau (SAR), China. MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), PR China.
| | - Elaine Lai-Han Leung
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau (SAR), China. MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), PR China.
| |
Collapse
|
39
|
Tsao MS, Rosenthal A, Nicholson AG, Detterbeck F, Eberhardt WEE, Lievens Y, Lim E, Matilla JM, Yatabe Y, Filosso PL, Beyruti R, Nishimura KK, Travis WD, Osarogiagbon RU, Rami-Porta R, Rusch V, Asamura H. The International Association for the Study of Lung Cancer Staging Project: The Database and Proposal for the Revision of the Staging of Pulmonary Neuroendocrine Carcinoma in the Forthcoming Ninth Edition of the TNM Classification for Lung Cancer. J Thorac Oncol 2025:S1556-0864(25)00046-2. [PMID: 39864546 DOI: 10.1016/j.jtho.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 01/28/2025]
Abstract
INTRODUCTION Pulmonary high-grade neuroendocrine carcinoma (NEC) includes SCLC and large cell NEC (LCNEC). The seventh and eighth editions of the TNM classification for lung cancer confirmed the applicability of this staging system for SCLC. With the proposal of N2 and M1c subcategories for the ninth edition classification, we assessed the applicability to NECs. METHODS The database included NEC cases diagnosed between January 2011 and December 2019. Eligible cases, with valid survival time and eighth edition TNM stage, were classified as pure SCLC, combined SCLC with NSCLC, and LCNEC. Survival was calculated using the Kaplan-Meier method, pairwise differences using a log-rank test, and prognostic groups using a Cox regression analysis. RESULTS There were 6181 pure and combined SCLC and 697 LCNEC cases available. For SCLC, survival outcome analyses included 4453 cases with clinical stage and 583 with pathologic stage data. The corresponding numbers for LCNEC were 585 and 508. The SCLC data validated the ninth edition classification for lung cancer, including the proposed new subcategories, N2a, single-station ipsilateral mediastinal or subcarinal lymph node involvement, and N2b, involvement of multiple ipsilateral or subcarinal stations. The data also validated the subcategorization of M1c into M1c1 (multiple lesions in a single extrathoracic organ system) and M1c2 (involvement of multiple extrathoracic organ systems). The LCNEC data were insufficient for complete survival analysis, but the available data reported decreasing survival with increasing clinical and pathologic stages. CONCLUSIONS The ninth edition TNM classification applies to patients with NEC and is the appropriate standard for use in clinical practice.
Collapse
Affiliation(s)
- Ming Sound Tsao
- Princess Margaret Cancer Centre and University of Toronto, Toronto, Ontario, Canada.
| | | | - Andrew G Nicholson
- Department of Histopathology, Royal Brompton and Harefield NHS Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, United Kingdom
| | - Frank Detterbeck
- Department of Thoracic Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Wilfried E E Eberhardt
- Department of Medical Oncology, UniversitätsKlinikum Essen, Department of Thoracic Oncology, Ruhrlandklinik, Essen, Germany
| | - Yolande Lievens
- Department of Radiation Oncology, Ghent University Hospital and Ghent University, Ghent, Belgium
| | - Eric Lim
- Imperial College London and the Academic Division of Surgery, Royal Brompton Hospital, London, United Kingdom
| | - Jose-Maria Matilla
- Department of Thoracic Surgery, Hospital Clínico Universitario de Valladolid, University of Valladolid, Valladolid, Spain
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | | | - Ricardo Beyruti
- Department of Thoracic Surgery, University of São Paulo, São Paulo, Brazil
| | | | - William D Travis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Ramon Rami-Porta
- Department of Thoracic Surgery, Hospital Universitari Mútua Terrassa, University of Barcelona, Terrassa, Spain; Network of Centers for Biomedical Research in Respiratory Diseases (CIBERES) Lung Cancer Group, Terrassa, Barcelona, Spain
| | - Valerie Rusch
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hisao Asamura
- Division of Thoracic Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
40
|
Feng Y, Jiang Y, Yang L, Lu D, Li N, Zhang Q, Yang H, Qin H, Zhang J, Gou X, Jiang F. Interactions and communications in lung tumour microenvironment: chemo/radiotherapy resistance mechanisms and therapeutic targets. J Drug Target 2025:1-20. [PMID: 39815747 DOI: 10.1080/1061186x.2025.2453730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/28/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025]
Abstract
The lung tumour microenvironment (TME) is composed of various cell types, including cancer cells, stromal and immune cells, as well as extracellular matrix (ECM). These cells and surrounding ECM create a stiff, hypoxic, acidic and immunosuppressive microenvironment that can augment the resistance of lung tumours to different forms of cell death and facilitate invasion and metastasis. This environment can induce chemo/radiotherapy resistance by inducing anti-apoptosis mediators such as phosphoinositide 3-kinase (PI3K)/Akt, signal transducer and activator of transcription 3 (STAT3) and nuclear factor kappa B (NF-κB), leading to the exhaustion of antitumor immunity and further resistance to chemo/radiotherapy. In addition, lung tumour cells can resist chemo/radiotherapy by boosting multidrug resistance mechanisms and antioxidant defence systems within cancer cells and other TME components. In this review, we discuss the interactions and communications between these different components of the lung TME and also the effects of hypoxia, immune evasion and ECM remodelling on lung cancer resistance. Finally, we review the current strategies in preclinical and clinical studies, including the inhibition of checkpoint molecules, chemoattractants, cytokines, growth factors and immunosuppressive mediators such as programmed death 1 (PD-1), insulin-like growth factor 2 (IGF-2) for targeting the lung TME to overcome resistance to chemotherapy and radiotherapy.
Collapse
Affiliation(s)
- Yuan Feng
- Guangxi University of Chinese Medicine, Nanning, China
| | - Ying Jiang
- Department of Neurology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Lin Yang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Danni Lu
- Guangxi University of Chinese Medicine, Nanning, China
| | - Ning Li
- Guangxi University of Chinese Medicine, Nanning, China
| | - Qun Zhang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Haiyan Yang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Huiyuan Qin
- Guangxi University of Chinese Medicine, Nanning, China
| | - Jiaxin Zhang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Xinyun Gou
- Guangxi University of Chinese Medicine, Nanning, China
| | - Feng Jiang
- Science and Technology Department, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
41
|
Das S, Samaddar S. Recent Advances in the Clinical Translation of Small-Cell Lung Cancer Therapeutics. Cancers (Basel) 2025; 17:255. [PMID: 39858036 PMCID: PMC11764476 DOI: 10.3390/cancers17020255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/03/2025] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
Small-cell lung cancer (SCLC) is a recalcitrant form of cancer, representing 15% of lung cancer cases globally. SCLC is classified within the range of neuroendocrine pulmonary neoplasms, exhibiting shared morphologic, ultrastructural, immunohistochemical, and molecular genomic features. It is marked by rapid proliferation, a propensity for early metastasis, and an overall poor prognosis. The current conventional therapies involve platinum-etoposide-based chemotherapy in combination with immunotherapy. Nonetheless, the rapid emergence of therapeutic resistance continues to pose substantial difficulties. The genomic profiling of SCLC uncovers significant chromosomal rearrangements along with a considerable mutation burden, typically involving the functional inactivation of the tumor suppressor genes TP53 and RB1. Identifying biomarkers and evaluating new treatments is crucial for enhancing outcomes in patients with SCLC. Targeted therapies such as topoisomerase inhibitors, DLL3 inhibitors, HDAC inhibitors, PARP inhibitors, Chk1 inhibitors, etc., have introduced new therapeutic options for future applications. In this current review, we will attempt to outline the key molecular pathways that play a role in the development and progression of SCLC, together with a comprehensive overview of the most recent advancements in the development of novel targeted treatment strategies, as well as some ongoing clinical trials against SCLC, with the goal of improving patient outcomes.
Collapse
Affiliation(s)
- Subhadeep Das
- Department of Biochemistry, Purdue University, BCHM A343, 175 S. University Street, West Lafayette, IN 47907, USA
- Purdue University Institute for Cancer Research, Purdue University, Hansen Life Sciences Research Building, Room 141, 201 S. University Street, West Lafayette, IN 47907, USA
| | | |
Collapse
|
42
|
Liao Z, Jia P, Li Y, Zheng Z, Zhang J. Exploring potential therapeutic targets for small cell lung cancer based on transcriptomics combined with Mendelian randomization analysis. Front Immunol 2025; 15:1464259. [PMID: 39872525 PMCID: PMC11769988 DOI: 10.3389/fimmu.2024.1464259] [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: 07/13/2024] [Accepted: 12/27/2024] [Indexed: 01/30/2025] Open
Abstract
Objective The main objective of this study was to explore and identify new genetic targets in small-cell lung cancer (SCLC) through transcriptomics analysis and Mendelian randomization (MR) analysis, which will help in the subsequent development of new therapeutic interventions. Methods In this study, we extracted the SCLC dataset from the Gene Expression Omnibus (GEO) database, processed the data, and screened out differentially expressed genes (DEGs) using R software. Based on expression quantitative trait loci data and the genome-wide association study data of SCLC, MR analysis was used to screen the genes closely related to SCLC disease, which intersect with DEGs to obtain co-expressed genes (CEGs), and the biological functions and pathways of CEGs were further explored by enrichment analysis. In addition, the CIBERSORT algorithm was applied to assess the level of immune cell infiltration in SCLC and to analyze the correlation between CEGs and immune cells. Meanwhile, we performed a survival analysis on these five CEGs using an independent cohort of SCLC patients. Finally, the results for the target genes were validated. Results In this study, 857 DEGs were identified, including 443 up-regulated and 414 down-regulated genes, and 5 CEGs (PSAT1, PSRC1, COLEC12, PLLP, HP) that were significantly associated with SCLC were identified through further intersecting. The results of enrichment analyses indicated that CEGs play important roles in several key functions and pathways. Immune-cell-related analysis revealed the unique distribution of immune cell infiltration in SCLC and the mechanism of immune cell regulation by CEGs. Survival analysis results indicated that PSRC1 was significantly correlated with the overall survival of SCLC, and the survival rate of the high-expression group was markedly lower than that of the low-expression group. Finally, the consistency of the results between the validation group analyses and MR analysis confirmed that the results of this study is reliable. Conclusion The CEGs and their associated functions and pathways screened in this study may be potential targets of therapeutic intervention in SCLC by targeting specific molecular pathways.
Collapse
Affiliation(s)
| | | | | | | | - Jizhou Zhang
- Department of Medical Oncology, Wenzhou TCM Hospital of Zhejiang Chinese Medical
University, Wenzhou, China
| |
Collapse
|
43
|
Maleki AH, Rajabivahid M, Khosh E, Khanali Z, Tahmasebi S, Ghorbi MD. Harnessing IL-27: challenges and potential in cancer immunotherapy. Clin Exp Med 2025; 25:34. [PMID: 39797931 PMCID: PMC11724803 DOI: 10.1007/s10238-025-01562-w] [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: 12/05/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
IL-27 is structurally an immune-enhancing and pleiotropic two-chain cytokine associated with IL-12 and IL-6 families. IL-27 contains two subunits, namely IL-27p28 and EBI3. A heterodimer receptor of IL-27, composed of IL27Rα (WSX1) and IL6ST (gp130) chains, mediates the IL-27 function following the activation of STAT1 and STAT3 signaling pathways. Specifically, IL-27 is identified as augmenting cytokine of immune responses, including Th1 cell differentiation, TCd4 + cell proliferation, and IFN-γ production with the help of IL-12. According to several published studies, due to the pro-inflammatory or anti-inflammatory functions of cytokine related to the biological context in various disorders and diseases, IL-27 has been considered a complex regulator of the immune system. Surprisingly, the dual role of IL-27, the same as the double-edged sword, has also been evidenced in clinical models of various hematological or solid tumors. Predominantly, Il-27 applies anti-tumor functions by inducing the responses of a cytotoxic T lymphocyte (CTL) and Th1 and suppressing the growth, proliferation, angiogenesis, invasiveness, metastasis, and survival of tumor cells. On the other hand, IL-27 may also play a protumor role in cancers and induce tumor progression. The current update study aimed to summarize the protumor anti-tumor and biological functions of IL-27 in different hematological malignancies and solid tumors.
Collapse
Affiliation(s)
| | - Mansour Rajabivahid
- Department of Internal Medicine, Valiasr Hospital, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Elnaz Khosh
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Zeinab Khanali
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Safa Tahmasebi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mahmood Dehghani Ghorbi
- Department of Hematology-Oncology, Imam Hossein Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
44
|
Hao Y, Li M, Liu W, Ma Z, Liu Z. Autophagic flux modulates tumor heterogeneity and lineage plasticity in SCLC. Front Oncol 2025; 14:1509183. [PMID: 39850810 PMCID: PMC11754400 DOI: 10.3389/fonc.2024.1509183] [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: 10/14/2024] [Accepted: 12/12/2024] [Indexed: 01/25/2025] Open
Abstract
Introduction Small cell lung cancer (SCLC) is characterized by significant heterogeneity and plasticity, contributing to its aggressive progression and therapy resistance. Autophagy, a conserved cellular process, is implicated in many cancers, but its role in SCLC remains unclear. Methods Using a genetically engineered mouse model (Rb1fl/fl ; Trp53fl/fl ; GFP-LC3-RFP-LC3△G), we tracked autophagic flux in vivo to investigate its effects on SCLC biology. Additional in vitro experiments were conducted to modulate autophagic flux in NE and non-NE SCLC cell lines. Results Tumor subpopulations with high autophagic flux displayed increased proliferation, enhanced metastatic potential, and neuroendocrine (NE) characteristics. Conversely, low-autophagic flux subpopulations exhibited immune-related signals and non-NE traits. In vitro, increasing autophagy induced NE features in non-NE cell lines, while autophagy inhibition in NE cell lines promoted non-NE characteristics. Discussion This study provides a novel model for investigating autophagy in vivo and underscores its critical role in driving SCLC heterogeneity and plasticity, offering potential therapeutic insights.
Collapse
Affiliation(s)
- Yujie Hao
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Mingchen Li
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Wenxu Liu
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Cell Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Zhenyi Ma
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Cell Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Zhe Liu
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Cell Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
45
|
Pozonec V, Pozonec MD, Aigner C, Widder J, Boettiger K, Megyesfalvi Z, Dome B. Prophylactic cranial irradiation for small cell lung cancer in the era of immunotherapy and molecular subtypes. Curr Opin Oncol 2025; 37:27-34. [PMID: 39625049 PMCID: PMC11623382 DOI: 10.1097/cco.0000000000001111] [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: 12/08/2024]
Abstract
PURPOSE OF REVIEW Small cell lung cancer (SCLC) is an aggressive disease with a poor prognosis, whereas its metastatic capacity carries a predilection for the brain. Although prophylactic cranial irradiation (PCI) has been used to address this problem, upcoming alternatives might necessitate reflection of its application in SCLC treatment. RECENT FINDINGS The addition of immunotherapy to treatment guidelines has provided a new strategy for the management of brain metastases. Complementation of immunotherapy with active MRI surveillance could potentially replace PCI and avoid irradiation-related cognitive side effects. SCLC's molecular profile is heterogeneous, with differential response to treatment modalities between subgroups. Investigation of these variances might be essential to improve therapeutic outcomes in SCLC patients. SUMMARY The role of PCI in SCLC treatment must be examined in light of immunotherapy. We summarize recent results, bearing SCLC subtypes and therapeutic vulnerabilities in mind, to derive tailored treatment strategies for SCLC patients in future settings.
Collapse
Affiliation(s)
- Veronika Pozonec
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology
- Multidisciplinary Centre of Head and Neck Tumors, National Institute of Oncology
| | - Maria Dorothea Pozonec
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | | | - Joachim Widder
- Department of Radiation Oncology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | | | - Zsolt Megyesfalvi
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- Department of Thoracic Surgery
| | - Balazs Dome
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- Department of Thoracic Surgery
- Department of Translational Medicine, Lund University, Lund, Sweden
| |
Collapse
|
46
|
Dómine Gómez M, Subbiah V, Peters S, Sala MA, Trigo J, Paz-Ares L, Nieto Archilla A, Gomez Garcia J, Alvarez García C, López-Vilariño de Ramos JA, Kahatt Lopez C, Fernandez CM. Lurbinectedin is an effective alternative to platinum rechallenge and may restore platinum sensitivity in patients with sensitive relapsed small cell lung cancer. Expert Rev Anticancer Ther 2025; 25:27-40. [PMID: 39660812 DOI: 10.1080/14737140.2024.2438067] [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: 11/11/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
INTRODUCTION Platinum rechallenge is recommended for patients with small cell lung cancer (SCLC) who relapse ≥90 days after completing first-line chemotherapy, although it may not always be the most suitable option. AREAS COVERED Articles for review were identified via PubMed and ClinicalTrials.gov searches, supplemented with non-indexed publications (e.g. conference abstracts) known to the manufacturer. We examined evidence for platinum re-exposure in patients with sensitive relapsed SCLC, and present lurbinectedin as a potential alternative. The complementary mechanisms of action of lurbinectedin and platinum, owing to opposite sensitivity of SCLC cells, may resensitize tumor cells to platinum. As efficacy outcomes with lurbinectedin are equivalent or better than those with platinum rechallenge and its hematological safety profile is more favorable, achieving maximum dose intensity is more likely. The simpler dosing schedule of lurbinectedin (1 vs 3 days) and lack of need for granulocyte colony-stimulating factor primary prophylaxis lessens treatment burden. EXPERT OPINION Incorporation of lurbinectedin into therapeutic algorithms for relapsed SCLC has challenged long-established treatment paradigms. Initial evidence indicates that using lurbinectedin after failure of first-line platinum may prolong the platinum-free interval and reserve platinum for later use. Current evidence supports lurbinectedin as a second-line option in patients with sensitive relapsed SCLC.
Collapse
Affiliation(s)
- Manuel Dómine Gómez
- Hospital Universitario Fundación Jiménez Díaz, IIS-FJD, Universidad Autónoma de Madrid, Madrid, Spain
| | - Vivek Subbiah
- Early-Phase Drug Development, Sarah Cannon Research Institute, Nashville, TN, USA
| | - Solange Peters
- Oncology Department - CHUV, Lausanne University, Lausanne, Switzerland
| | - María Angeles Sala
- Medical Oncology Department, Hospital Universitario Basurto, Bilbao, Spain
| | - José Trigo
- Medical Oncology Department, Hospital HC Marbella, Málaga, Spain
| | - Luis Paz-Ares
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
47
|
Jin X, Zhao W, Li G, Jiang J. Immunotherapy for Limited-Stage Small Cell Lung Cancer: Innovative Treatments and Future Perspectives. Cancer Control 2025; 32:10732748251334434. [PMID: 40228829 DOI: 10.1177/10732748251334434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025] Open
Abstract
BackgroundLimited-stage small cell lung cancer (LS-SCLC) is a highly aggressive tumor characterized by a poor prognosis. While concurrent chemoradiotherapy (CCRT) remains the standard treatment, the high rates of recurrence and poor long-term survival highlight the pressing need for novel therapeutic approaches.PurposeIn recent years, the introduction of immunotherapy, particularly immune checkpoint inhibitors (ICIs), has opened new avenues for the treatment of LS-SCLC. This review highlights the clinical advancements of ICIs in CCRT, consolidation therapy, and neoadjuvant therapy, emphasizing their potential to improve progression-free survival (PFS) and overall survival (OS). This review also discusses management of immunotherapy-related side effects.Research DesignThis is a review article that synthesizes recent research findings on immunotherapy for LS-SCLC.Study SampleNot applicable (review of existing literature).Data Collection and/or AnalysisThis review summarizes key studies exploring the application of immunotherapy in limited-stage small cell lung cancer.Additionally, it examines the role of the tumor microenvironment, tumor mutation burden (TMB), and Programmed cell death 1 ligand 1(PD-L1) as biomarkers for predicting the efficacy of immunotherapy.ResultsThis review emphasizes their potential to improve PFS and OS.ConclusionsDespite the significant advancements in research, the use of ICIs in LS-SCLC continues to face challenges, including the identification of optimal treatment regimens, validation of long-term efficacy, and development of personalized predictive biomarkers. Future research should prioritize large-scale, multicenter clinical trials to refine combination therapy strategies, establish customized treatment approaches, and enhance patient outcomes.
Collapse
Affiliation(s)
- Xiaoni Jin
- Department of Oncology, Graduate School of Qinghai University, Xining, China
| | - Weixing Zhao
- Department of Oncology, Graduate School of Qinghai University, Xining, China
| | - Guoyuan Li
- Division Ⅱ, Department of Medical Oncology, Affiliated Hospital of Qinghai University, Xining, China
| | - Jun Jiang
- Division III, Department of Medical Oncology, Affiliated Hospital of Qinghai University, Xining, China
| |
Collapse
|
48
|
Cheng Y, Zhang W, Wu L, Zhou C, Wang D, Xia B, Bi M, Fu X, Li C, Lv D, Zhao Y, Chen G, Yi T, Huang J, Li M, Yang R, Huang X, Wang Y, Zhang M, Pan Y, Sun Y, Hu S, Zhang X, Zhou M, Fang J, Jin F, Liu Y, Li Y, Zhang Z, Hu J, Liu L, Wang R, Li Y, Gu K, Ding C, Fan Q, Zhang G, Chen Y, Jiang L, Zheng WE, Chen S, Huang C, Han Z, Yang H, Wang J, Wang B, Wu H, Bao Y, Li M, Luo X, Gu S, Yu W, Xu K, Zhang S, Yu J. Toripalimab Plus Chemotherapy as a First-Line Therapy for Extensive-Stage Small Cell Lung Cancer: The Phase 3 EXTENTORCH Randomized Clinical Trial. JAMA Oncol 2025; 11:16-25. [PMID: 39541202 PMCID: PMC11565370 DOI: 10.1001/jamaoncol.2024.5019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 07/05/2024] [Indexed: 11/16/2024]
Abstract
Importance Patients with extensive-stage small cell lung cancer (ES-SCLC) have poor prognoses and unmet medical needs. Objective To evaluate the efficacy and safety of toripalimab plus etoposide and platinum-based chemotherapy (EP) vs placebo plus EP as a first-line treatment for patients with ES-SCLC. Design, Setting, and Participants This multicenter, double-blind, placebo-controlled phase 3 randomized clinical trial (EXTENTORCH study) enrolled patients from September 26, 2019, to May 20, 2021, and was conducted at 49 sites in China. Eligible patients had histologically or cytologically confirmed ES-SCLC without previous systemic antitumor therapy for ES-SCLC. Data were analyzed between May 6, 2023, and June 1, 2024. Interventions Patients were randomized (1:1) to receive toripalimab, 240 mg, or placebo plus EP every 3 weeks for up to 4 to 6 cycles, followed by maintenance with toripalimab or placebo until disease progression, intolerable toxic effects, or up to 2 years of treatment. Main Outcomes and Measures The primary end points were investigator-assessed progression-free survival (PFS) and overall survival (OS). Whole-exome sequencing results identified correlative biomarkers for clinical efficacy. Results Among 595 screened patients, 442 eligible patients were randomized (median [range] age, 63 [30-77] years; 366 [82.8%] male); 223 patients were randomized to toripalimab plus EP, and 219 to placebo plus EP. By April 20, 2023, the median (range) survival follow-up was 13.7 (0.0-42.7) months. Compared with placebo, toripalimab improved investigator-assessed PFS (hazard ratio [HR], 0.67 [95% CI, 0.54-0.82]; P < .001), and significantly reduced the risk of death (HR, 0.80 [95% CI, 0.65-0.98]; P = .03). The median OS was 14.6 (95% CI, 12.9-16.6) months in the toripalimab group vs 13.3 (95% CI, 11.8-14.4) months in the placebo group. Whole-exome sequencing results from 300 patients identified low intratumor heterogeneity, HLA-A11+ HLA-B62- haplotype, wild-type KMT2D and COL4A4, or sequence variations in CTNNA2 or SCN4A correlated with favorable PFS and OS in the toripalimab group. No new safety signals were observed. Grade 3 or higher treatment-emergent adverse event incidence was similar between the toripalimab and placebo safety set groups (199 of 222 patients [89.6%] vs 193 of 216 patients [89.4%], respectively). Conclusions and Relevance In this phase 3 randomized clinical trial, adding toripalimab to first-line chemotherapy demonstrated significant improvements in PFS and OS for patients with ES-SCLC. The treatment exhibited an acceptable safety profile, supporting this combination regimen as a new treatment option for patients with ES-SCLC. Trial Registration ClinicalTrials.gov Identifier: NCT04012606.
Collapse
Affiliation(s)
- Ying Cheng
- Department of Oncology, Jilin Cancer Hospital, Changchun, China
| | - Wei Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lin Wu
- Second Department of Thoracic Medical Oncology, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Donglin Wang
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Bing Xia
- Department of Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minghong Bi
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Xiuhua Fu
- Department of Respiratory, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Chong Li
- Department of Respiration, First People’s Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Dongqing Lv
- Department of Respiratory Medicine, Taizhou Hospital, Taizhou, China
| | - Yanqiu Zhao
- Department of Respiratory Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Gongyan Chen
- First Department of Internal Medicine, Harbin Medical University Affiliated Cancer Hospital, Harbin, China
| | - Tienan Yi
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Sciences, Xiangyang, China
| | - Jianan Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Li
- Department of Respiratory Medicine, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China
| | - Runxiang Yang
- The Second Department of Medical Oncology, Yunnan Cancer Hospital, Kunming, China
| | - Xiaoping Huang
- Department of Medical Oncology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Ye Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Mingjun Zhang
- Department of Oncology, Second Affiliated Hospital of Anhui Medical University Hefei, Hefei, China
| | - Yueyin Pan
- Department of Tumor Chemotherapy, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, China
| | - Yilan Sun
- Department of Respiratory Disease, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Sheng Hu
- Ward I Chest Medicine, Hubei Cancer Hospital, Wuhan, China
| | - Xiqin Zhang
- Department of Respiratory Medicine, Shandong Cancer Hospital and Institute, Jinan, China
| | - Min Zhou
- Second Department of Internal Medicine, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Jian Fang
- Second Department of Thoracic Oncology, Beijing Cancer Hospital, Beijing, China
| | - Faguang Jin
- Department of Respiratory Medicine, Tangdu Hospital, The Second Affiliated Hospital of the Chinese People’s Liberation Army Air Force Medical University, Xi’an, China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Yinyin Li
- First Department of Oncology, Shenyang the Tenth People’s Hospital, Shenyang, China
| | - Zhihong Zhang
- Department of Respiratory Medicine, Anhui Provincial Cancer Hospital, Hefei, China
| | - Jie Hu
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Laiyu Liu
- Department of Respiratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Rui Wang
- Department of Medical Oncology, Anhui Chest Hospital, Hefei, China
| | - Yan Li
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Kangsheng Gu
- Department of Medical Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Cuimin Ding
- Department of Respiratory Medicine, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, China
| | - Qingxia Fan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guojun Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yongxing Chen
- Department of Respiratory, Hainan General Hospital, Haikou, China
| | - Liyan Jiang
- Department of Respiratory Medicine, Shanghai Chest Hospital, Jiao Tong University, Shanghai, China
| | - Wei-E. Zheng
- Department of Medical Oncology, Ruian People’s Hospital, Ruian, China
| | - Shaoshui Chen
- Department of Oncology, Binzhou Medical University Hospital, Binzhou, China
| | - Cheng Huang
- Department of Pneumology, Fujian Provincial Cancer Hospital, Fuzhou, China
| | - Zhigang Han
- Pulmonary Medicine, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, China
| | - Hong Yang
- Department of Oncology, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, China
| | - Jianfang Wang
- Department of Oncology, Shaoxing People’s Hospital, Shaoxing, China
| | - Baocheng Wang
- Department of Oncology, The 960th Hospital of People’s Liberation Army, Jinan, China
| | - Huita Wu
- Department of Oncology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Yongxing Bao
- Department of Oncology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Manxiang Li
- Department of Respiratory, First Affiliated Hospital of Xian Jiaotong University, Xi’an, China
| | - Xianming Luo
- Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Shanshan Gu
- Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Wenbo Yu
- Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Kai Xu
- Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Simo Zhang
- TopAlliance Biosciences Inc, Rockville, Maryland
| | - Jianjun Yu
- TopAlliance Biosciences Inc, Rockville, Maryland
| |
Collapse
|
49
|
Opee SA, Eva AA, Noor AT, Hasan SM, Mridha MF. ELW-CNN: An extremely lightweight convolutional neural network for enhancing interoperability in colon and lung cancer identification using explainable AI. Healthc Technol Lett 2025; 12:e12122. [PMID: 39845172 PMCID: PMC11751720 DOI: 10.1049/htl2.12122] [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: 11/03/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 01/24/2025] Open
Abstract
Cancer is a condition in which cells in the body grow uncontrollably, often forming tumours and potentially spreading to various areas of the body. Cancer is a hazardous medical case in medical history analysis. Every year, many people die of cancer at an early stage. Therefore, it is necessary to accurately and early identify cancer to effectively treat and save human lives. However, various machine and deep learning models are effective for cancer identification. Therefore, the effectiveness of these efforts is limited by the small dataset size, poor data quality, interclass changes between lung squamous cell carcinoma and adenocarcinoma, difficulties with mobile device deployment, and lack of image and individual-level accuracy tests. To overcome these difficulties, this study proposed an extremely lightweight model using a convolutional neural network that achieved 98.16% accuracy for a large lung and colon dataset and individually achieved 99.02% for lung cancer and 99.40% for colon cancer. The proposed lightweight model used only 70 thousand parameters, which is highly effective for real-time solutions. Explainability methods such as Grad-CAM and symmetric explanation highlight specific regions of input data that affect the decision of the proposed model, helping to identify potential challenges. The proposed models will aid medical professionals in developing an automated and accurate approach for detecting various types of colon and lung cancer.
Collapse
Affiliation(s)
- Shaiful Ajam Opee
- Department of Computer ScienceAmerican International University‐BangladeshDhakaBangladesh
| | - Arifa Akter Eva
- Department of Computer ScienceAmerican International University‐BangladeshDhakaBangladesh
| | - Ahmed Taj Noor
- Department of Computer Science EngineeringSoutheast UniversityDhakaBangladesh
| | - Sayem Mustak Hasan
- Department of Computer ScienceAmerican International University‐BangladeshDhakaBangladesh
| | - M. F. Mridha
- Department of Computer ScienceAmerican International University‐BangladeshDhakaBangladesh
| |
Collapse
|
50
|
Zheng R, Yu C, Yao D, Cai M, Zhang L, Ye F, Huang X. Engineering Stimuli-Responsive Materials for Precision Medicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406439. [PMID: 39444066 PMCID: PMC11707583 DOI: 10.1002/smll.202406439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Over the past decade, precision medicine has garnered increasing attention, making significant strides in discovering new therapeutic drugs and mechanisms, resulting in notable achievements in symptom alleviation, pain reduction, and extended survival rates. However, the limited target specificity of primary drugs and inter-individual differences have often necessitated high-dosage strategies, leading to challenges such as restricted deep tissue penetration rates and systemic side effects. Material science advancements present a promising avenue for these issues. By leveraging the distinct internal features of diseased regions and the application of specific external stimuli, responsive materials can be tailored to achieve targeted delivery, controllable release, and specific biochemical reactions. This review aims to highlight the latest advancements in stimuli-responsive materials and their potential in precision medicine. Initially, we introduce disease-related internal stimuli and capable external stimuli, elucidating the reaction principles of responsive functional groups. Subsequently, we provide a detailed analysis of representative pre-clinical achievements of stimuli responsive materials across various clinical applications, including enhancements in the treatment of cancers, injury diseases, inflammatory diseases, infection diseases, and high-throughput microfluidic biosensors. Finally, we discuss some clinical challenges, such as off-target effects, long-term impacts of nano-materials, potential ethical concerns, and offer insights into future perspectives of stimuli-responsive materials.
Collapse
Affiliation(s)
- Ruixuan Zheng
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
| | - Chang Yu
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
- Intervention DepartmentThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
| | - Dan Yao
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
| | - Mengsi Cai
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
| | - Lexiang Zhang
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiang325000China
| | - Fangfu Ye
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiang325000China
- Beijing National Laboratory for Condensed Matter PhysicsInstitute of PhysicsChinese Academy of SciencesBeijing100190China
| | - Xiaoying Huang
- Joint Centre of Translational MedicineDivision of Pulmonary MedicineThe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiang325000China
- Wenzhou Key Laboratory of Interdiscipline and Translational MedicineThe First Affiliated Hospital of Wenzhou Medical University WenzhouWenzhouZhejiang325000China
| |
Collapse
|