1
|
Ma L, Li R, Li P, Yu W, Tang Z, Si L, Tian H. GINS1 facilitates the development of lung adenocarcinoma via Wnt/β-catenin activation. World J Surg Oncol 2025; 23:122. [PMID: 40197379 PMCID: PMC11974172 DOI: 10.1186/s12957-025-03786-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 03/29/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Lung adenocarcinoma(LUAD) is the primary reason for cancer-related deaths globally. GINS1 has a significant regulatory function in DNA replication. It is overexpressed in various malignant tumors, but the specific molecular mechanisms of GINS1 in LUAD pathogenesis are not fully elucidated. This is the first report that GINS1 enhances LUAD by activating Wnt/β-catenin signaling pathway, and may serve as a potential target for therapy. METHODS Bioinformatic analysis including analysis of difference, survival analysis and pathway enrichment, immunohistochemistry(IHC), western blotting(WB), and quantitative real time polymerase chain reaction(qRT-PCR) were used to detect GINS1 expression in LUAD cell lines and tissues. A range of in vivo and in vitro experiments, such as cck-8, EdU, cloning experiment, wound healing experiment and transwell experiment, confirmed that GINS1 facilitated the proliferation and migration of LUAD. Additionally, the potential mechanism of GINS1 was hypothesized through WB and transcriptome sequencing. The rescue experiment was used to verify our conclusion. RESULTS In this study, we discovered that GINS1 is significantly overexpressed in LUAD cell lines and tissues. Analysis of Kaplan - Meier survival data indicated that high levels of GINS1 expression are often linked to unfavorable survival outcomes. Additionally, a series of experiments showed that silencing GINS1 led to less proliferation and migration of LUAD cell lines, while its overexpression enhanced tumor progression. Furthermore, subcutaneous tumor experiments in nude mice supported the role of GINS1 in promoting tumor development in vivo. Lastly, transcriptome sequencing revealed that tumor progression is related to cell cycle (G1 to S phase transition associated with cyclinD) and β-catenin signaling pathway, which we subsequently validated using WB. A series of rescue experiment further confirmed that GINS1 facilitates the advancement of LUAD via the β-catenin signaling pathway. CONCLUSIONS Our findings suggest that GINS1 plays a critical role in the progression of LUAD by modulating key molecular pathways, particularly the β-catenin signaling pathway., and it might serve as a potential new target of β-catenin signaling pathway for treatment of LUAD.
Collapse
Affiliation(s)
- Luyuan Ma
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Rongyang Li
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Pengyong Li
- Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Wenhao Yu
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Zhanpeng Tang
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Libo Si
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| | - Hui Tian
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
2
|
Li W, Zhu K, Liu Y, Liu M, Chen Q. Recent advances in PKC inhibitor development: Structural design strategies and therapeutic applications. Eur J Med Chem 2025; 287:117290. [PMID: 39904144 DOI: 10.1016/j.ejmech.2025.117290] [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: 12/16/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Protein kinase C (PKC) isozymes play critical roles in diverse cellular processes and are implicated in numerous diseases, including cancer, diabetes, and autoimmune disorders. Despite extensive research efforts spanning four decades, only one PKC inhibitor has received clinical approval, highlighting the challenges in developing selective and efficacious PKC-targeting therapeutics. Here we review recent advances in the development of small-molecule PKC inhibitors, focusing on structural design strategies, pharmacological activities, and structure-activity relationships. We analyze emerging approaches including fragment-based drug design, allosteric targeting, and natural product derivatization that have yielded promising new scaffold classes. Special attention is given to innovations in achieving isozyme selectivity, particularly for PKCα and PKCβ, which have proven crucial for therapeutic applications. We discuss how integration of computational methods, structural biology insights, and rational design principles has advanced our understanding of PKC inhibition mechanisms. This comprehensive analysis reveals key challenges in PKC drug development, including the need for enhanced selectivity and reduced off-target effects, while highlighting promising directions for future therapeutic development. Our findings provide a framework for designing next-generation PKC inhibitors with improved clinical potential.
Collapse
Affiliation(s)
- Wen Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Kun Zhu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yuyin Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Meixi Liu
- Department of Endocrinology, Deyang Hospital Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, 618000, China
| | - Qiu Chen
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
3
|
Lin J, Lin S, Lin Z, Huang Z, Li D. Peroxynitrite as biomarker to evaluate the rehabilitation of cisplatin in the resistant cells with miR-125a-5p by using fluorescent assay. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 330:125663. [PMID: 39733707 DOI: 10.1016/j.saa.2024.125663] [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: 09/19/2024] [Revised: 12/11/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
Reactive oxygen species (ROS) play a dual role in the chemotherapy of cancer with cisplatin, providing both anti-tumor effects and contributing to drug resistance at various stages of treatment which seriously affects treatment effectiveness. The detailed mechanism of ROS is urgently necessary to be explored. To address this issue in the non-small-cell lung cancer (NSCLC) with cisplatin-resistance, a reliable assay was developed by synthesizing and characterizing an interesting near-infrared (NIR) ONOO- probe BPB with high specificity, quick response (<30 s) and excellent limitation of detection (59 nM), which was further convinced through living cell imaging techniques providing different fluorescence variation between cell and cuvette. All the results revealed that ONOO- might be a practical biomarker to comprehend the detail molecular mechanism in cisplatin-resistant A549 cells. The elevated levels of ONOO- in cisplatin-resistant A549 cells, accompanied by a significant reduction in fluorescence following the knockout of miR-125a-5p in these cells and strong fluorescence without knockout of miR-125a-5p ignoring the presence of cisplatin. Comparing with cisplatin-resistant cells, the control would provide a rational background and then showed distinct fluorescence from BPB with ONOO- toward cisplatin. This assay offers a promising tool for exploring the molecular mechanisms associated with miR-125a-5p and its potential linkage to therapeutic efficacy involving ONOO- signaling. By utilizing this innovative assay, researchers can gain valuable insights into the treatment strategies and underlying mechanisms of cisplatin-resistant cancers, which should be beneficial to the therapy of cancers.
Collapse
Affiliation(s)
- Jinpei Lin
- Department of Integrative Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province 350014, China
| | - Shufang Lin
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Zengyan Lin
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Zhengrong Huang
- Department of Integrative Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province 350014, China.
| | - Daliang Li
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China.
| |
Collapse
|
4
|
Meng W, Xu R, Miller E, Sun X, Thurmond J, Webb A, McElroy J, Palmer J, DiCostanzo DJ, Zhang S, Yamaguchi H, Haque SJ, Zhu J, Chakravarti A. Pilot Study of Metabolomic Biomarkers Associated with Outcomes in Patients with Lung Cancer Undergoing Radiation Therapy. J Proteome Res 2025; 24:1662-1671. [PMID: 40073233 PMCID: PMC11976842 DOI: 10.1021/acs.jproteome.4c00529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/26/2024] [Accepted: 12/20/2024] [Indexed: 03/14/2025]
Abstract
Lung cancer stands as the leading cause of cancer-related death worldwide, impacting both men and women in the United States and beyond. Radiation therapy (RT) serves as a key treatment modality for various lung malignancies. Our study aims to systematically assess the prognosis and influence of RT on metabolic reprogramming in patients diagnosed with nonsmall-cell lung cancer (NSCLC) through longitudinal metabolic profiling. A cohort of 54 NSCLC patients underwent thoracic radiotherapy, with 96% receiving a total radiation dose ranging from 40 to 70 Gy, averaging 56.3 Gy. Blood biospecimens were collected before RT, during RT, and at the first follow-up after RT, with a total of 126 serum samples randomized for liquid chromatography-mass spectrometry (LC-MS) metabolomics analysis using a high-performance LC (HPLC)-Q-Exactive mass spectrometry system. Our results indicated that the serum metabolite coumarin derivatives prior to radiotherapy exhibited the strongest unfavorable outcome with overall survival in these NSCLC cases. The metabolites in the blood samples can reflect the responses during RT. Notably, over half of the metabolites (12/23) were found to be fatty acids in the longitudinal analysis. This pilot study indicated that metabolic profiling of biofluids from NSCLC patients undergoing RT has the potential to assess the patient outcomes during and after treatment.
Collapse
Affiliation(s)
- Wei Meng
- Department
of Radiation Oncology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Rui Xu
- Department
of Human Sciences, The Ohio State University, Columbus, Ohio 43210, United States
| | - Eric Miller
- Department
of Radiation Oncology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Xiaowei Sun
- Department
of Human Sciences, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jennifer Thurmond
- Department
of Radiation Oncology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Amy Webb
- Center
for Biostatistics, The Ohio State University, Columbus, Ohio 43210, United States
| | - Joseph McElroy
- Center
for Biostatistics, The Ohio State University, Columbus, Ohio 43210, United States
| | - Joshua Palmer
- Department
of Radiation Oncology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Dominic J. DiCostanzo
- Department
of Radiation Oncology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Shiqi Zhang
- Department
of Human Sciences, The Ohio State University, Columbus, Ohio 43210, United States
| | - Hisashi Yamaguchi
- Department
of Radiation Oncology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Saikh Jaharul Haque
- Department
of Radiation Oncology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jiangjiang Zhu
- Department
of Human Sciences, The Ohio State University, Columbus, Ohio 43210, United States
| | - Arnab Chakravarti
- Department
of Radiation Oncology, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
5
|
Ogden J, Sellers R, Sahoo S, Oojageer A, Chaturvedi A, Dive C, Lopez-Garcia C. A human model to deconvolve genotype-phenotype causations in lung squamous cell carcinoma. Nat Commun 2025; 16:3215. [PMID: 40185723 PMCID: PMC11971459 DOI: 10.1038/s41467-025-58343-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
Tractable, patient-relevant models are needed to investigate cancer progression and heterogeneity. Here, we report an alternative in vitro model of lung squamous cell carcinoma (LUSC) using primary human bronchial epithelial cells (hBECs) from three healthy donors. The co-operation of ubiquitous alterations (TP53 and CDKN2A loss) and components of commonly deregulated pathways including squamous differentiation (SOX2), PI3K signalling (PTEN) and the oxidative stress response (KEAP1) is investigated by generating hBECs harbouring cumulative alterations. Our analyses confirms that SOX2-overexpression initiates early preinvasive LUSC stages, and co-operation with the oxidative stress response and PI3K pathways to drive more aggressive phenotypes, with expansion of cells expressing LUSC biomarkers and invasive properties. This cooperation is consistent with the classical LUSC subtype. Importantly, we connect pathway dysregulation with gene expression changes associated with cell-intrinsic processes and immunomodulation. Our approach constitutes a powerful system to model LUSC and unravel genotype-phenotype causations of clinical relevance.
Collapse
Affiliation(s)
- Julia Ogden
- Cancer Research UK Manchester Institute, Wilmslow Road, M20 4BX, Manchester, United Kingdom
| | - Robert Sellers
- Cancer Research UK Manchester Institute, Wilmslow Road, M20 4BX, Manchester, United Kingdom
| | - Sudhakar Sahoo
- Cancer Research UK Manchester Institute, Wilmslow Road, M20 4BX, Manchester, United Kingdom
| | - Anthony Oojageer
- Cancer Research UK Manchester Institute, Wilmslow Road, M20 4BX, Manchester, United Kingdom
| | - Anshuman Chaturvedi
- Department of Histopathology, The Christie Hospital, Wilmslow Road, Manchester, M20 4BX, United Kingdom
| | - Caroline Dive
- Cancer Research UK Manchester Institute, Wilmslow Road, M20 4BX, Manchester, United Kingdom
- Cancer Research UK, National Biomarker Centre, Wilmslow Road, M20 4BX, Manchester, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, Wilmslow Road, M20 4BX, Manchester, United Kingdom
| | - Carlos Lopez-Garcia
- Cancer Research UK Manchester Institute, Wilmslow Road, M20 4BX, Manchester, United Kingdom.
- Cancer Research UK Lung Cancer Centre of Excellence, Wilmslow Road, M20 4BX, Manchester, United Kingdom.
| |
Collapse
|
6
|
Korde A, Ramaswamy A, Anderson S, Jin L, Zhang JG, Hu B, Velasco WV, Diao L, Wang J, Pisani MA, Sauler M, Boffa DJ, Puchalski JT, Yan X, Moghaddam SJ, Takyar SS. Cigarette smoke induces angiogenic activation in the cancer field through dysregulation of an endothelial microRNA. Commun Biol 2025; 8:511. [PMID: 40155749 PMCID: PMC11953391 DOI: 10.1038/s42003-025-07710-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 02/10/2025] [Indexed: 04/01/2025] Open
Abstract
Cigarette smoke (CS) creates a "cancer field" in the lung that promotes malignant transformation. The molecular changes within this field are not fully characterized. We examined the significance of microRNA-1 (miR-1) downregulation as one of these changes. We found that tumor miR-1 levels in three non-small cell lung cancer cohorts show inverse correlations with the smoking burden. Lung MiR-1 levels follow a spatial gradient, have prognostic significance, and correlate inversely with the molecular markers of injury. In CS-exposed lungs, miR-1 is specifically downregulated in the endothelium. Exposure to CS induces angiogenesis by selectively degrading mature miR-1 via a vascular endothelial growth factor-driven pathway. Applying a multi-step molecular screen, we identified angiogenic genes regulated by miR-1 in the lungs of smokers. Knockdown of one of these genes, Notch homolog protein 3, simulates the anti-angiogenic effects of miR-1. These findings suggest that miR-1 can be used as an indicator of malignant transformation.
Collapse
Affiliation(s)
- Asawari Korde
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Anuradha Ramaswamy
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Seth Anderson
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Lei Jin
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Jian-Ge Zhang
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Buqu Hu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Walter V Velasco
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lixia Diao
- Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Wang
- Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Margaret A Pisani
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Maor Sauler
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Daniel J Boffa
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Jonathan T Puchalski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Xiting Yan
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Seyed Javad Moghaddam
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shervin S Takyar
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
7
|
Gawli CS, Patil CR, Patel HM. A clinical review on third and fourth generation EGFR tyrosine kinase inhibitors for the treatment of non-small cell lung cancer. Bioorg Med Chem 2025; 123:118146. [PMID: 40153991 DOI: 10.1016/j.bmc.2025.118146] [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: 01/16/2025] [Revised: 02/10/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
"Epidermal growth factor receptor (EGFR)" mutations are pivotal in the pathogenesis of "Non-Small Cell Lung Cancer (NSCLC)," which is associated with high morbidity and mortality rates. The advent of third and fourth-generation EGFR tyrosine kinase inhibitors (TKIs) has significantly advanced the therapeutic landscape for EGFR-mutant NSCLC, particularly in overcoming resistance mutations such as T790M and C797S. This review delves into the current clinical status, efficacy, safety profiles, and regulatory approvals of third-generation EGFR TKIs, including Osimertinib, Lazertinib, Furmonertinib, Aumolertinib, Rezivertinib, Befotertinib, Sunvozertinib. Furthermore, it explores emerging fourth-generation EGFR TKIs designed to address resistance mechanisms beyond those targeted by their predecessors. Notable fourth-generation candidates such as TQB3804, BPI-361175, BDTX-1535, WJ13404, QLH11811, H002, HS-10375, BBT-207, JIN-A02, and HS-10504 are highlighted for their potential to overcome the C797S mutation. The review emphasizes the importance of these advanced inhibitors in enhancing "progression-free survival and overall survival rates". By evaluating the therapeutic potential and limitations of these EGFR TKIs, this review aims to guide future research in the management of EGFR-mutant NSCLC. This acts as guiding beacon for the strategic design and development of third and fourth generation EGFR-TK inhibitors to overcome the drug resistance hurdles in the development of EGFR-TK inhibitors.
Collapse
Affiliation(s)
- Chandrakant S Gawli
- R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, District-Dhule 425405 Maharashtra, India
| | - Chandragouda R Patil
- R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, District-Dhule 425405 Maharashtra, India
| | - Harun M Patel
- R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, District-Dhule 425405 Maharashtra, India.
| |
Collapse
|
8
|
Waizman E, Dudnik E, Lavie I, Rotem O, Amiel A, Siegal T, Haim SE, Gal O, Limon D, Tschernichovsky R, Kanner AA, Laviv Y, Katz SY. The impact of brain MRI screening on stage IV NSCLC patients: A real world look at guidelines based care. J Neurol Sci 2025; 470:123398. [PMID: 39922142 DOI: 10.1016/j.jns.2025.123398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/15/2024] [Accepted: 01/18/2025] [Indexed: 02/10/2025]
Abstract
PURPOSE Brain metastases frequently occur in patients with non-small cell lung cancer (NSCLC) and are associated with poor prognosis and shortened overall survival, despite the advances in both imaging and therapeutic fields. Data are scarce regarding the utility of brain magnetic resonance imaging (MRI) screening in NSCLC patients. We aimed to characterize the impact of brain MRI screening on stage IV NSCLC patients and their survival. METHODS A retrospective analysis was performed in 609 newly-diagnosed patients with stage IV NSCLC treated at our center during 2019-2020. Patients with neurological symptoms at presentation were excluded (n = 230). The remaining 379 patients comprised the study group and were divided into two groups - those who underwent brain MRI screening within 12 weeks of diagnosis (n = 170), and those who did not (n = 209). The clinical data retrieved from patients' medical files included demographics, performance status (PS), brain metastases during follow-up, tumor molecular profiling, and oncology treatment. RESULTS Median survival among patients who underwent brain MRI screening was 24 months from diagnosis, versus 18 months for those who did not (p = 0.003). Among patients with good PS (ECOG 0-2), median OS was longer among patients who underwent screening MRI (25 months), versus those who did not (21 months) (p = 0.025). Among patients with low PS (ECOG 3-4), the between-group difference for OS did not reach statistical significance. CONCLUSIONS Our study supports the use of brain MRI screening among good PS patients diagnosed with stage IV NSCLC lung cancer. Brain MRI screening correlated with better survival among patients with good functional status in this patient population.
Collapse
Affiliation(s)
- Efraim Waizman
- Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.
| | | | - Inbar Lavie
- Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel; Brain Tumor Center, Davidoff Cancer Center, Rabin Medical Center - Beilinson Hospital, Petach Tikva, Israel
| | - Ofer Rotem
- Brain Tumor Center, Davidoff Cancer Center, Rabin Medical Center - Beilinson Hospital, Petach Tikva, Israel
| | - Alexandra Amiel
- Brain Tumor Center, Davidoff Cancer Center, Rabin Medical Center - Beilinson Hospital, Petach Tikva, Israel
| | - Tali Siegal
- Brain Tumor Center, Davidoff Cancer Center, Rabin Medical Center - Beilinson Hospital, Petach Tikva, Israel
| | - Shaked Even Haim
- Brain Tumor Center, Davidoff Cancer Center, Rabin Medical Center - Beilinson Hospital, Petach Tikva, Israel
| | - Omer Gal
- Brain Tumor Center, Davidoff Cancer Center, Rabin Medical Center - Beilinson Hospital, Petach Tikva, Israel
| | - Dror Limon
- Brain Tumor Center, Davidoff Cancer Center, Rabin Medical Center - Beilinson Hospital, Petach Tikva, Israel
| | - Roi Tschernichovsky
- Brain Tumor Center, Davidoff Cancer Center, Rabin Medical Center - Beilinson Hospital, Petach Tikva, Israel
| | - Andrew A Kanner
- Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel; Brain Tumor Center, Davidoff Cancer Center, Rabin Medical Center - Beilinson Hospital, Petach Tikva, Israel
| | - Yosef Laviv
- Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel; Brain Tumor Center, Davidoff Cancer Center, Rabin Medical Center - Beilinson Hospital, Petach Tikva, Israel
| | - Shlomit Yust Katz
- Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel; Brain Tumor Center, Davidoff Cancer Center, Rabin Medical Center - Beilinson Hospital, Petach Tikva, Israel
| |
Collapse
|
9
|
Zhang J, Wang X, Wang J, Wen X, Chen S, Wang T, Wang B, Hu W. ACSS3 promotes the tumorigenesis of non-small cell lung cancer via suppressing p53-mediated ferroptosis. Exp Cell Res 2025; 446:114438. [PMID: 39961466 DOI: 10.1016/j.yexcr.2025.114438] [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: 05/02/2024] [Revised: 11/22/2024] [Accepted: 02/09/2025] [Indexed: 02/27/2025]
Abstract
Non-small cell lung cancer (NSCLC) is a subtype of the most frequently diagnosed cancer, causing a considerable number of deaths globally. Mitochondrial dysfunction was found to promote malignant progression. However, the underlying mechanism remains unclear. Acyl-CoA synthetase short chain family member 3 (ACSS3) is mainly located in mitochondria, which abnormal regulation is usually accompanied by the occurrence and development of tumors. In this study, we found that the expression level of ACSS3 was correlated with poor prognosis in patients with NSCLC. Moreover, we demonstrated that ACSS3 knockdown led to mitochondrial contraction, increased reactive oxygen species levels, decreased mitochondrial membrane potential, and subsequently inhibited tumor growth of NSCLC cells in vitro and in vivo, whereas its overexpression promoted these processes. Mechanistically, ACSS3 knockdown promoted ferroptosis through transcriptional control of SLC7A11 and GPX4. Further investigations indicated that ACSS3 loss inhibited the SLC7A11/GPX4 axis by enhancing p53 stability. Taken together, our data confirmed that ACSS3 promotes NSCLC tumorigenesis through inhibiting the p53-mediated ferroptosis. Hence, ACSS3 emerges as a promising therapeutic target for NSCLC treatment.
Collapse
Affiliation(s)
- Jing Zhang
- Center for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), The Second Affiliated Hospital, Chongqing Medical University, Medical College Road No.1, Chongqing, 400016, PR China
| | - Xiuhong Wang
- Department of Pathology, China-Japan Friendship Hospital, Yinghua East Road No. 2, BeiJing, 100029, PR China
| | - Jingyi Wang
- Center for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), The Second Affiliated Hospital, Chongqing Medical University, Medical College Road No.1, Chongqing, 400016, PR China
| | - Xiao Wen
- Center for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), The Second Affiliated Hospital, Chongqing Medical University, Medical College Road No.1, Chongqing, 400016, PR China
| | - Siyuan Chen
- Center for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), The Second Affiliated Hospital, Chongqing Medical University, Medical College Road No.1, Chongqing, 400016, PR China
| | - Tao Wang
- Center for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), The Second Affiliated Hospital, Chongqing Medical University, Medical College Road No.1, Chongqing, 400016, PR China
| | - Bei Wang
- Department of Pathology, China-Japan Friendship Hospital, Yinghua East Road No. 2, BeiJing, 100029, PR China.
| | - Wenquan Hu
- Center for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), The Second Affiliated Hospital, Chongqing Medical University, Medical College Road No.1, Chongqing, 400016, PR China.
| |
Collapse
|
10
|
Kim K, Syeed S, Au T, Diaz A, Schabath MB, Cass A, Hall R, Pai L, Li C, Balmaceda N, Palumbo A, Carey A, Lalla M, Henry M, Brixner D, Stenehjem D. Real-world comparative outcomes of EGFR-TKIs for first-line treatment of EGFR+ metastatic non-small-cell lung cancer. Cancer Treat Res Commun 2025; 43:100898. [PMID: 40120239 DOI: 10.1016/j.ctarc.2025.100898] [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: 04/22/2024] [Revised: 10/28/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
PURPOSE Osimertinib is a third-generation EGFR-TKI and preferred first-line (1L) treatment for EGFR positive (EGFR+) metastatic non-small cell lung cancer (mNSCLC). This study compared real-world clinical outcomes of 1L osimertinib versus 1st or 2nd generation EGFR-TKIs (1/2G-TKIs) in patients with EGFR+ mNSCLC. METHODS Nine academic cancer centers in the US participated in the retrospective cohort study. Patients aged ≥18 years with EGFR+ mNSCLC and treated with 1L EGFR-TKI were included. Clinical outcomes included real-world progression-free survival (rwPFS), duration of treatment (DOT), time to next treatment (TTNT), central nervous system incidence-free survival (CNS-IFS), and overall survival (OS). Multivariable regression models were used to control for differences in patient characteristics (p < 0.1) between the osimertinib and 1/2G-TKI cohorts. RESULTS The study included 181 osimertinib patients and 171 1/2G-TKI patients. Osimertinib had a longer rwPFS compared to 1/2G-TKIs (median PFS, 95 % confidence interval [CI]: 16.2 months (13.2-19.7) vs. 10.8 months (9.5-12.7); hazard Ratio [HR], 95 % CI: 0.60 (0.44-0.82). DOT and TTNT were significantly longer in patients treated with osimertinib versus 1/2G-TKI (HR, 95 % CI: 0.51 (0.38-0.68) for DOT; 0.54 (0.39-0.74) for TTNT). The respective HR point estimate for CNF-IFS and OS of 0.62 and 0.83 preferred osimertinib. However, small patient counts and number of events posed challenges in drawing conclusion regarding the significance of the delayed CNS-IFS or OS. CONCLUSION Patients treated with osimertinib had a prolonged time to progression and longer time maintain the treatment compared to 1/2G-TKI. This real-world evidence is aligned with clinical trial results.
Collapse
Affiliation(s)
- Kibum Kim
- University of Utah, Department of Pharmacotherapy and Pharmacotherapy Outcomes Research Center, Salt Lake City, UT, USA; University of Illinois Chicago, Department of Pharmacy Systems, Outcomes and Policy, Chicago, IL, USA.
| | - Sakil Syeed
- University of Utah, Department of Pharmacotherapy and Pharmacotherapy Outcomes Research Center, Salt Lake City, UT, USA
| | - Trang Au
- University of Utah, Department of Pharmacotherapy and Pharmacotherapy Outcomes Research Center, Salt Lake City, UT, USA
| | - Amber Diaz
- Oregon Health & Science University, Portland, OR, USA
| | | | - Amanda Cass
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - Richard Hall
- University of Virginia Medical Center, Charlottesville, VA, USA
| | - Lori Pai
- Tufts Medical Center, Boston, MA, USA
| | - Chenghui Li
- University of Arkansas for Medical Sciences College of Pharmacy, Little Rock, AR, USA
| | | | | | - Autumn Carey
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | | | - Matthew Henry
- University of Arkansas for Medical Sciences College of Pharmacy, Little Rock, AR, USA
| | - Diana Brixner
- University of Utah, Department of Pharmacotherapy and Pharmacotherapy Outcomes Research Center, Salt Lake City, UT, USA
| | - David Stenehjem
- University of Utah, Department of Pharmacotherapy and Pharmacotherapy Outcomes Research Center, Salt Lake City, UT, USA; University of Minnesota, Department of Pharmacy Practice and Pharmaceutical Sciences, Duluth, MN, USA
| |
Collapse
|
11
|
Liu L, Gao C, Yang Y, Tang M, Zhao T, Chen D, Jin J, Xu Y, Li G, Zhong Q. Efficacy and safety of induction immunochemotherapy followed by radiotherapy for patients with unresectable locally advanced non-small cell lung cancer: A retrospective study. Radiat Oncol 2025; 20:37. [PMID: 40082897 PMCID: PMC11905732 DOI: 10.1186/s13014-025-02616-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 02/28/2025] [Indexed: 03/16/2025] Open
Abstract
OBJECTIVES Immune checkpoint inhibitor (ICI) has displayed considerable advantages in consolidation therapy of locally advanced non-small cell lung cancer (LA-NSCLC) after concurrent chemoradiotherapy (cCRT). However, many patients are considered unsuitable for cCRT owing to concerns with tolerability. In this study, we aimed to assess the efficacy and toxicity of induction immunochemotherapy followed by radiotherapy for unresectable LA-NSCLC who are not capable of receiving cCRT. METHODS From January 2019 and December 2022, LA-NSCLC patients treated with induction immunochemotherapy followed by radiotherapy as initial treatment at our institution were retrospectively reviewed. The short-term efficacy, overall survival (OS), progression free survival (PFS) and tolerability of induction immunochemotherapy followed by radiotherapy were evaluated in these patients. RESULTS Overall, 24 patients were enrolled (median age 64 years, 33.3% with ECOG performance status score 2, and 62.5% with stage IIIB-IIIC). Median follow-up from the start of induction immunochemotherapy was 30.5 months. Median number of induction immunochemotherapy was 4 cycles. A median radiotherapy dose of 60 Gy was delivered. After radiotherapy, 16 patients (66.6%) received consolidation immunotherapy. The overall response rate in these patients was 87.5%. The 1-year, 2-year and 3-year OS were 91.7%, 74.8% and 57.0%, respectively. The 1-year, 2-year and 3-year PFS were 87.0%, 54.1% and 37.1%, respectively. The incidence of grade ≥ 2 and grade ≥ 3 pneumonitis were 37.5% and 16.7%, respectively. Radiation pneumonitis of any grade occurred in 8 patients (33.3%), and the incidence of grade ≥ 2 and grade ≥ 3 radiation pneumonitis were 16.7% and 12.5%, respectively. CONCLUSION Induction immunochemotherapy followed by radiotherapy and consolidated immunotherapy had encouraging efficacy with acceptable toxicity for LA-NSCLC not capable of receiving cCRT.
Collapse
Affiliation(s)
- Lipin Liu
- Department of Radiation Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Cui Gao
- Department of Radiation Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Yufan Yang
- Department of Radiation Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Min Tang
- Department of Medical Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Ting Zhao
- Department of Radiation Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Dazhi Chen
- Department of Radiation Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Jingyi Jin
- Department of Radiation Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Yonggang Xu
- Department of Radiation Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Gaofeng Li
- Department of Radiation Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Qiuzi Zhong
- Department of Radiation Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P. R. China.
| |
Collapse
|
12
|
Vinjamuri S, Pant V. Demystifying the Role of Immuno PET-CT in Non-Small Cell Lung Cancer: Clinical Value and Research Trends. Semin Nucl Med 2025; 55:212-220. [PMID: 40016063 DOI: 10.1053/j.semnuclmed.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 03/01/2025]
Abstract
The management of Lung cancer, especially non-small cell lung cancer has undergone a paradigm shift recently with the advent of new treatment approaches including focused radiotherapy as well as evolution of a newer class of immunotherapy agents. Treatment efficacy and survival rates have improved and it is now even more important that patients are selected for appropriate interventions on the basis of a comprehensive assessment including a range of imaging as well as in-vitro tests such as immunohistochemistry. A new class of tracers targeting programmed cell death such as PD1 and PDL1 (broadly classed as Immuno PET) are being increasingly used in the molecular characterisation of patients deemed resistant to standard treatment approaches and being considered for additional interventions such as immunotherapy. In this review, we review the latest evidence in the field and propose a summary of clinical usefulness and provide a review of the research trends in this exciting and evolving field.
Collapse
Affiliation(s)
- Sobhan Vinjamuri
- Department of Nuclear Medicine, Royal Liverpool University Hospital, Liverpool, UK.
| | - Vineet Pant
- Department of Nuclear Medicine, Royal Liverpool University Hospital, Liverpool, UK
| |
Collapse
|
13
|
Dobkin J, Stanifer BP, Salvatore M, Eckhardt CM. Evaluating lung cancer risk factors in adults with interstitial lung disease. Lung Cancer 2025; 201:108416. [PMID: 39893773 PMCID: PMC11884992 DOI: 10.1016/j.lungcan.2025.108416] [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: 01/07/2025] [Revised: 01/27/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Adults with interstitial lung disease (ILD) have a higher risk of developing lung cancer compared to the general population. We aimed to identify ILD-specific risk factors that can be used to improve lung cancer detection in this high-risk population. METHODS Adults ≥21 years who received at least two chest CT scans at an academic medical center between 2005 and 2020 and were found to have ILD were studied retrospectively. Lung cancer diagnoses were adjudicated based on pathology reports from lung biopsies. Logistic regression was used to evaluate associations of clinical variables with comorbid lung cancer. RESULTS Among 1,366 adults with ILD, the mean age was 67.2 ± 12.4 years and 639 (46.8 %) were men. In total, 227 adults (16.6 %) had a lung nodule on CT imaging, of whom 55 (24.3 %) were diagnosed with lung cancer. Radiographic usual interstitial pneumonia (UIP) (OR 3.00, 95 % CI 1.43-6.33) was independently associated with increased odds of lung cancer. Risk factors including age, sex, smoking status, pack-years, use of immunosuppression, and radiographic fibrosis pattern collectively demonstrated high discriminative accuracy in predicting comorbid lung cancer, even among adults who would not have qualified for lung cancer screening based on current guidelines (AUC 0.80, 95 % CI 0.72-0.88). CONCLUSIONS In a large study of adults with ILD, radiographic UIP was independently associated with comorbid lung cancer even after adjusting for established risk factors. Our results suggest radiographic UIP is an independent lung cancer risk factor and support the development of targeted lung cancer screening guidelines in adults with UIP.
Collapse
Affiliation(s)
- Jane Dobkin
- Columbia University Irving Medical Center, Department of Medicine, New York, NY, USA
| | - B Payne Stanifer
- Columbia University Vagelos College of Physicians & Surgeons, Department of Surgery, New York, NY, USA
| | - Mary Salvatore
- Jacobi Medical Center, Department of Radiology, Bronx, NY, USA
| | - Christina M Eckhardt
- Columbia University Vagelos College of Physicians & Surgeons, Department of Medicine, New York, NY, USA; Columbia University Mailman School of Public Health, Department of Environmental Health Sciences, New York, NY, USA.
| |
Collapse
|
14
|
Sultana A, Alam MS, Khanam A, Liang H. Unraveling the molecular landscape of non-small cell lung cancer: Integrating bioinformatics and statistical approaches to identify biomarkers and drug repurposing. Comput Biol Med 2025; 187:109744. [PMID: 39914199 DOI: 10.1016/j.compbiomed.2025.109744] [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: 08/28/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/21/2025]
Abstract
Non-small-cell lung cancer (NSCLC) is one of the most malignant tumors and the leading cause of death from cancer worldwide and is increasing at a massive rate every year. Most NSCLC patients are diagnosed at advanced stages, which is associated with a poor prognosis and a very low 5-year survival rate. Therefore, the purpose of this study is to investigate molecular markers for early diagnosis, prognosis and therapy of NSCLC through the integration of bioinformatics and statistical methods. A total of 93 overlapping differentially expressed genes (oDEGs) were identified between NSCLC and normal samples through Linear Models for Microarray (LIMMA) and Significance Analysis of Microarrays (SAM) methods. Six top-degree oDEGs (CCNA2, CDC6, AURKA, CCNB1, MKI67, and PRC1) were identified as key genes (KGs) through the protein-protein interaction (PPI) network. The predictive accuracy analysis of the identified KGs revealed an accuracy of 96.92 %, with a sensitivity of 91.73 % and a specificity of 98.15 %. KGs associated with 3 molecular functions (MFs), 5 cellular components (CCs), 3 biological processes (BPs), and 4 pathways were identified through FunRich software. Analysis of expression levels using the UALCAN database revealed that KGs are significantly associated with potential early diagnostic biomarkers. Survival analysis using the GEPIA database demonstrated that the KGs possessed strong prognostic power for NSCLC. Finally, seven repurposed candidate drugs ENTRECTINIB, SORAFENIB, CHEMBL1765740, TOZASERTIB, NERVIANO, AZD-1152-HQPA, and SELICICLIB were proposed through molecular docking analysis. In conclusion, the findings of this study have the potential to significantly impact the early diagnosis, prognosis, and treatment of NSCLC.
Collapse
Affiliation(s)
- Adiba Sultana
- Medical Big Data Center, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, 510080, China
| | - Md Shahin Alam
- Medical Big Data Center, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, 510080, China; Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, China.
| | - Alima Khanam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Huiying Liang
- Medical Big Data Center, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, 510080, China.
| |
Collapse
|
15
|
ALMatrafi TA. Deciphering the role of TMEM164 in autophagy-mediated ferroptosis and immune modulation in non-small cell lung cancer. Cell Immunol 2025; 409-410:104915. [PMID: 39798196 DOI: 10.1016/j.cellimm.2024.104915] [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: 08/04/2024] [Revised: 11/13/2024] [Accepted: 12/25/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) remains one of the most prevalent and deadly malignancies. Despite advancements in molecular therapies and diagnostic methods, the 5-year survival rate for lung adenocarcinoma patients remains unacceptably low, highlighting the urgent need for novel therapeutic strategies. Ferroptosis, a distinct form of regulated cell death, has emerged as a promising target in cancer treatment. This study investigates the role of TMEM164, a membrane protein, in promoting ferroptosis and modulating anti-tumor immunity in NSCLC, aiming to elucidate its therapeutic potential. METHODS Using publicly available datasets, we performed bioinformatics analyses to identify TMEM164-regulated genes involved in ferroptosis. In addition, in vitro and in vivo assays were conducted to assess the impact of TMEM164 on cellular functions in NSCLC. RESULTS Functional assays demonstrated that TMEM164 overexpression significantly inhibited invasion, migration, and cell proliferation in both in vitro and in vivo models. TMEM164 was also found to induce ferroptosis in NSCLC cells by promoting autophagy. Specifically, we identified a mechanism whereby TMEM164 mediates ATG5-dependent autophagosome formation, leading to the degradation of ferritin, GPX4, and lipid droplets. This degradation facilitated iron accumulation and lipid peroxidation, which triggered iron-dependent cell death. Notably, co-administration of TMEM164 upregulation and anti-PD-1 antibodies exhibited synergistic anti-tumor effects in a mouse model. CONCLUSION These findings suggest that targeting TMEM164 to enhance ferroptosis and stimulate anti-tumor immunity may inhibit NSCLC progression. Consequently, TMEM164 holds promise as a new therapeutic target for NSCLC treatment.
Collapse
|
16
|
Wang Y, Li Z, Lin C, Zhou J, Cai X, Lv F, Yang W, Ji L. Revisiting the association between sodium-glucose cotransporter-2 inhibitors and the risk of neoplasm in patients with type 2 diabetes: new insights from an updated systematic review and meta-analysis of randomized controlled trials. Expert Rev Clin Pharmacol 2025; 18:165-173. [PMID: 39886909 DOI: 10.1080/17512433.2024.2439970] [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: 09/25/2024] [Revised: 11/16/2024] [Accepted: 12/05/2024] [Indexed: 02/01/2025]
Abstract
OBJECTIVE To evaluate the association between sodium-glucose cotransporter-2 inhibitors (SGLT-2i) and the risk of neoplasm in patients with Type 2 diabetes (T2D). METHODS Literature retrieval was conducted using databases from inception to June 2024. Randomized controlled trials (RCTs) comparing SGLT-2i with placebo or other treatments in patients with T2D, and with reports of neoplasm events were included. Results were computed as the risk ratio (RR) with 95% confidence intervals (CI). RESULTS A total of 53 RCTs with 126,232 participants were included. No significant differences were found for the risk of overall neoplasm (RR = 1.08, 95% CI: 0.99 to 1.19, I2 = 23%) in patients with SGLT-2i treatment compared with non-users. However, decreased risk of pulmonary neoplasm (RR = 0.83, 95% CI: 0.69 to 0.99, I2 = 0.0%) was observed in SGLT-2i users compared to non-users, while increased risk of prostate neoplasm in SGLT-2i users was found (RR = 1.21, 95% CI: 1.00 to 1.47, I2 = 0.0%). CONCLUSION Compared with non-users, the use of SGLT-2i was not associated with the risk of overall neoplasm. However, pulmonary neoplasms were less frequent in SGLT-2i users, while an increased risk of prostate neoplasm was observed in SGLT-2i users compared to non-users. PROTOCOL REGISTRATION www.crd.york.ac.uk/prospero identifier is CRD42021273681.
Collapse
Affiliation(s)
- Yiran Wang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Zonglin Li
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Chu Lin
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Jinyu Zhou
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Xiaoling Cai
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Fang Lv
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Wenjia Yang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| |
Collapse
|
17
|
Yin J, Huang J, Ren M, Tang R, Xie L, Xue J. A Bayesian network meta-analysis of EGFR-tyrosine kinase inhibitor treatments in patients with EGFR mutation-positive non-small cell lung cancer. CANCER PATHOGENESIS AND THERAPY 2025; 3:135-146. [PMID: 40182124 PMCID: PMC11963207 DOI: 10.1016/j.cpt.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/11/2024] [Accepted: 06/18/2024] [Indexed: 04/05/2025]
Abstract
Background To date, no direct comparisons have been performed to compare the effectiveness of all epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) against EGFR mutation-positive non-small cell lung cancer (NSCLC). This study aimed to investigate the efficacy and safety of EGFR-TKIs in patients with EGFR mutation-positive NSCLC. Methods We conducted a network meta-analysis of randomized controlled trials comparing osimertinib, lazertinib, aumolertinib, befotertinib, furmonertinib, dacomitinib, afatinib, erlotinib, gefitinib, icotinib, and chemotherapy. Pooled estimations of progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and toxicity (grade ≥ 3 adverse events) were performed within the Bayesian framework. Results Twenty-three trials involving 11 treatments were included. All EGFR-TKIs improved PFS relative to chemotherapy, except for icotinib (hazard ratio [HR] = 0.61, 95% confidence interval [CI]: 0.26-1.44). All EGFR-TKIs demonstrated significant ORR benefits over chemotherapy. Osimertinib seemed to prolong PFS compared with icotinib (HR = 0.29, 95% CI: 0.1-0.86), gefitinib (HR = 0.39, 95% CI: 0.21-0.74), and erlotinib (HR = 0.53, 95% CI: 0.29-1.0). In addition, osimertinib showed favorable superiority in improving OS compared with chemotherapy (HR = 0.6, 95% CI: 0.43-0.82), gefitinib (HR = 0.61, 95% CI: 0.45-0.83), erlotinib (HR = 0.65, 95% CI: 0.48-0.89), and afatinib (HR = 0.65, 95% CI: 0.44-0.94). Among these regimens, afatinib showed the highest ORR (cumulative probability: 96.96%). Icotinib was associated with minimal toxicity among the EGFR-TKIs, followed by furmonertinib and osimertinib. Moreover, the toxicity spectra differed among the EGFR-TKIs. Subgroup analyses of patients with two common types of EGFR mutations indicated that furmonertinib possessed the greatest PFS benefit in patients with exon 19 deletion, and lazertinib showed the greatest PFS benefit in patients with Leu858Arg mutation. We also identified differences between EGFR-TKIs in prolonging PFS in patients with brain metastasis. Conclusions Osimertinib is the first choice of treatment with considerable efficacy and safety for EGFR mutation-positive NSCLC. The treatments associated with the best PFS in patients with exon 19 deletions and Leu858Arg mutations were furmonertinib and lazertinib, respectively.
Collapse
Affiliation(s)
- Jianqiong Yin
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jing Huang
- Department of Ultrasound, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Min Ren
- Abdominal Oncology Ward, Division of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Rui Tang
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Linshen Xie
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianxin Xue
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
18
|
Thapa R, Marianesan AB, Rekha A, Ganesan S, Kumari M, Bhat AA, Ali H, Singh SK, Chakraborty A, MacLoughlin R, Gupta G, Dua K. Hypoxia-inducible factor and cellular senescence in pulmonary aging and disease. Biogerontology 2025; 26:64. [PMID: 40011266 DOI: 10.1007/s10522-025-10208-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025]
Abstract
Cellular senescence and hypoxia-inducible factor (HIF) signaling are crucial in pulmonary aging and age-related lung diseases such as chronic obstructive pulmonary disease idiopathic pulmonary fibrosis and lung cancer. HIF plays a pivotal role in cellular adaptation to hypoxia, regulating processes like angiogenesis, metabolism, and inflammation. Meanwhile, cellular senescence leads to irreversible cell cycle arrest, triggering the senescence-associated secretory phenotype which contributes to chronic inflammation, tissue remodeling, and fibrosis. Dysregulation of these pathways accelerates lung aging and disease progression by promoting oxidative stress, mitochondrial dysfunction, and epigenetic alterations. Recent studies indicate that HIF and senescence interact at multiple levels, where HIF can both induce and suppress senescence, depending on cellular conditions. While transient HIF activation supports tissue repair and stress resistance, chronic dysregulation exacerbates pulmonary pathologies. Furthermore, emerging evidence suggests that targeting HIF and senescence pathways could offer new therapeutic strategies to mitigate age-related lung diseases. This review explores the intricate crosstalk between these mechanisms, shedding light on how their interplay influences pulmonary aging and disease progression. Additionally, we discuss potential interventions, including senolytic therapies and HIF modulators, that could enhance lung health and longevity.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - A Rekha
- Dr D Y Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Mukesh Kumari
- NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo, NSW, 2007, Australia
| | - Amlan Chakraborty
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Ronan MacLoughlin
- Aerogen, IDA Business Park, Dangan, Galway, H91 HE94, Ireland
- School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- School of Pharmacy & Pharmaceutical Sciences, Trinity College, Dublin, D02 PN40, Ireland
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo, NSW, 2007, Australia.
- Discipline of Pharmacy, Graduate School of Health, University of Technology, Sydney, Ultimo, NSW, 2007, Australia.
- Woolcock Institute of Medical Research, Macquarie University, Sydney, Australia.
| |
Collapse
|
19
|
Fisher WS, Douglas J, Roshan S, Perez R, Wei S, Roberts L, Ewert KK, Safinya CR. Acidic Conditions Promote Clustering of Cancer Cell Derived Extracellular Vesicles and Enhance their Fusion with Synthetic Liposomes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:4533-4544. [PMID: 39943777 DOI: 10.1021/acs.langmuir.4c04297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
Extracellular vesicles (EVs) are endogenous vesicles secreted by cells. Exosomes (30-150 nm), are a subset of EVs playing key roles in intercellular communication. Exosomes show promise as cancer chemotherapeutic drug delivery vehicles given their low immunogenicity and cell-specific cytosolic delivery of their contents. However, inefficient drug loading limits their therapeutic application. To address this, methods for the fusion of EVs with therapeutic drug-loaded synthetic liposomes have been developed. While more efficient than passive incubation of EVs with liposomes, these risk either damage to EV membrane proteins or contamination of the EV-liposome hybrids with residual depletant molecules, which can cause side effects or hinder content delivery. Here, we present a new, weakly perturbative method, which uses acidic conditions (pH 5) to enhance the fusion of EVs and synthetic, neutral liposomes (NLs) compared to passive incubation in pH 7.4 at 37 °C. An adapted Forster resonance energy transfer (FRET) based lipid mixing assay confirms that fusion is enhanced with this method, albeit less efficiently than with depletant-induced fusion. This significant finding implies that lipid-only synthetic liposomes can fuse with EVs, creating EV-liposome hybrids under relevant temperature and pH conditions, without nonlipidic components, such as fusogenic amphipathic peptides, added to the synthetic liposomes. Remarkably, differential interference contrast (DIC) and fluorescence microscopy show that this enhanced fusion corresponds with the clustering of mixtures of EVs and NLs, or EVs alone, in acidic but not neutral pH conditions. The findings support a hypothesis that content release from EVs in early to late endocytic environments may be a combination of protein-protein clustering interactions and a lipidic component. Further, this study provides a novel method for enhanced fusion of EVs and liposomes, which is expected to preserve EV membrane proteins and functionality toward the development of therapeutic hybrid drug delivery vehicles in nanomedicine applications.
Collapse
Affiliation(s)
- William S Fisher
- Materials Department, Molecular, Cellular, and Developmental Biology Department, Physics Department, and Biomolecular Science and Engineering Program, University of California, Santa Barbara, California 93106, United States
| | - Jessica Douglas
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, United States
| | - Sherwin Roshan
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, United States
| | - Ramon Perez
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, United States
| | - Sophia Wei
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, United States
| | - Logan Roberts
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, United States
| | - Kai K Ewert
- Materials Department, Molecular, Cellular, and Developmental Biology Department, Physics Department, and Biomolecular Science and Engineering Program, University of California, Santa Barbara, California 93106, United States
| | - Cyrus R Safinya
- Materials Department, Molecular, Cellular, and Developmental Biology Department, Physics Department, and Biomolecular Science and Engineering Program, University of California, Santa Barbara, California 93106, United States
| |
Collapse
|
20
|
Iemwimangsa N, Anantaya D, Oranratnachai S, Thamrongjirapat T, Lumjiaktase P, Teoh VH, Khiewngam K, Monnamo N, Sanvarinda P, Incharoen P, Charoenyingwattan A, Sensorn I, Dejthevaporn T, Sirachainan E, Chantratita W, Reungwetwattana T, Trachu N. Dynamic changes in immune repertoire profiles in patients with stage III unresectable non-small cell lung cancer during consolidation treatment with immunotherapy. BMC Cancer 2025; 25:333. [PMID: 39994571 PMCID: PMC11853222 DOI: 10.1186/s12885-025-13716-w] [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: 09/05/2023] [Accepted: 02/11/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND One-year of immune checkpoint inhibitor (ICI) treatment after concurrent chemoradiation (CCRT) in unresectable stage III non-small cell lung cancer (NSCLC) is a standard of care. The precise predictive biomarkers are under investigations either immunological markers or clinical characteristics. Here, we explored immune repertoire of T cell receptor β-chain (TCRβ) during ICI treatment. METHODS During August 2019 and September 2021, stage III NSCLC, post CCRT patients from Ramathibodi Hospital was enrolled. All patients were treated by durvalumab after CCRT. Blood samples were collected together with clinical data and tumor assessment every 3-4 months until disease progression or discontinuation of treatment due to adverse events. CDR3 region and TCRΒ polymorphisms was explored by RNA sequencing using Next-Generation Sequencing (NGS) TCR beta short-read assay. Bioinformatic analysis was performed to analyze clonal diversity, TCR convergence frequency and the Shannon diversity from each timepoint. Immune repertoire and clinical correlation were explored using Spearman's correlation and Pearson's correlation. RStudio software version 2021 build 372 was used for analyses. A significance level was at P < 0.05. RESULTS Forty-four blood samples from 12 patients were analyzed. Mean duration of durvalumab treatment was 284 days. After durvalumab treatment, increasing of TCR convergence frequency was found compared to baseline (R = 0.36). Interestingly, it was also significantly higher in non-progressive disease (non-PD) patients compared with progressive disease (PD) patients (P = 0.011). Furthermore, Shannon diversity was higher increasing in PD patients compared with non-PD patients. Taken together, our study found that increasing of TCR convergence with less T-cell diversity in non-PD patients probably demonstrated a T cell-specific clonal expansion response to durvalumab treatment in this population. CONCLUSIONS TCRβ repertoire is the potential biomarker for predicting durvalumab treatment response in post CCRT stage III NSCLC patients. However, a larger cohort with long-read assay should be explored.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/therapy
- Lung Neoplasms/pathology
- Lung Neoplasms/genetics
- Male
- Female
- Middle Aged
- Aged
- Neoplasm Staging
- Immunotherapy/methods
- Immune Checkpoint Inhibitors/therapeutic use
- Chemoradiotherapy/methods
- Antibodies, Monoclonal/therapeutic use
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Biomarkers, Tumor/blood
- Adult
- High-Throughput Nucleotide Sequencing
Collapse
Affiliation(s)
- Nareenart Iemwimangsa
- Center for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Dulyathat Anantaya
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Faculty of Medicine Ramathibodi Hospital, Ramathibodi Lung Cancer Consortium (RLC), Mahidol University, Bangkok, Thailand
| | - Songporn Oranratnachai
- Oncology Unit Sriphat Medical Center, Faculty of Medicine, Chiangmai University, Chiangmai, Thailand
| | - Thanaporn Thamrongjirapat
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Faculty of Medicine Ramathibodi Hospital, Ramathibodi Lung Cancer Consortium (RLC), Mahidol University, Bangkok, Thailand
| | - Putthapoom Lumjiaktase
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | | | - Khantong Khiewngam
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Nanamon Monnamo
- Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Tungpayathai, Rajathewee, Bangkok, 10400, Thailand
| | | | - Pimpin Incharoen
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Angkana Charoenyingwattan
- Center for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Insee Sensorn
- Center for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Thitiya Dejthevaporn
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Faculty of Medicine Ramathibodi Hospital, Ramathibodi Lung Cancer Consortium (RLC), Mahidol University, Bangkok, Thailand
| | - Ekaphop Sirachainan
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Faculty of Medicine Ramathibodi Hospital, Ramathibodi Lung Cancer Consortium (RLC), Mahidol University, Bangkok, Thailand
| | - Wasun Chantratita
- Center for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Thanyanan Reungwetwattana
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Faculty of Medicine Ramathibodi Hospital, Ramathibodi Lung Cancer Consortium (RLC), Mahidol University, Bangkok, Thailand
| | - Narumol Trachu
- Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Tungpayathai, Rajathewee, Bangkok, 10400, Thailand.
| |
Collapse
|
21
|
Shen Y, Dong X, Li X, Shi Z, Shao T, Jiang J, Song J. WNT inhibitor SP5-mediated SERPING1 suppresses lung adenocarcinoma progression via TSC2/mTOR pathway. Cell Death Dis 2025; 16:103. [PMID: 39962118 PMCID: PMC11832940 DOI: 10.1038/s41419-025-07440-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/25/2025] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
The long-term outlook for patients grappling with lung cancer (LC) remains bleak, with lung adenocarcinoma (LUAD) emerging as the most predominant histological subtype. Our Mendelian randomization (MR) investigation spotlighted that heightened levels of the circulating protein serpin peptidase inhibitor family G1 (SERPING1) substantially mitigated LC risk. The fusion of multi-omics strategies unveiled that SERPING1 exhibited diminished expression in LUAD patients compared to healthy individuals both in tissues and serum, with LUAD individuals showcasing elevated SERPING1 expression demonstrating improved prognoses. Furthermore, SERPING1 expression exhibited a robust correlation with the efficacy of immunotherapy. Through meticulous in vivo and in vitro analyses, we unraveled that SERPING1 impeded the proliferation, migration, invasion and wound healing of LUAD cells via the tuberous sclerosis 2 (TSC2)/mammalian target of rapamycin (mTOR) pathway. Mechanistically, WNT inhibitor- Specificity Protein (SP5) was delineated as facilitator of SERPING1 transcription by binding to the SERPING1 gene promoter. Intriguingly, aside from the association between SERPING1 and systolic blood pressure, glycosylated hemoglobin (HbA1c), type I diabetes, no discernible link between SERPING1 overexpression and heightened risks of other cardiometabolic conditions and diseases was evident. In summary, SERPING1 emerges as a novel tumor suppressor gene and SP5/SERPING1/TSC2 is a promising therapeutic target in the context of LUAD.
Collapse
Affiliation(s)
- Yefeng Shen
- Institute of Cardiovascular Sciences, Guangxi Academy of Medical Sciences, Nanning, China
- Department of Thoracic Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaofeng Dong
- Department of Hepatobiliary, Pancreas and Spleen Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region (Guangxi Academy of Medical Sciences), Nanning, China
| | - Xujia Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhiyuan Shi
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
| | - Tingting Shao
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Junlan Jiang
- Department of Pathology, the First Affiliated Hospital, Anhui Medical University, Hefei, China
- Pathology Center, Anhui Medical University, Hefei, China
| | - Jian Song
- Institute of Cardiovascular Sciences, Guangxi Academy of Medical Sciences, Nanning, China.
- Department of Radiation Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
22
|
Kharat M, Nookala H, John JD, Patel T, Kalyandrug P, Emmanuel S, Noor A, Halani DJK, Goyal A, Anand N, Gadad B, Millis RM. A comparative evaluation of alectinib for ALK-positive non-small-cell lung cancer: A systematic review. Medicine (Baltimore) 2025; 104:e41506. [PMID: 39960949 PMCID: PMC11835117 DOI: 10.1097/md.0000000000041506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/23/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Lung cancer, a significant global health challenge, notably the non-small-cell lung cancer (NSCLC) subtype, is a topic of utmost importance. The continuous advancements in NSCLC treatment, especially in the context of anaplastic lymphoma kinase (ALK)-positive NSCLC, are of great interest. A thorough review of alectinib's comparative efficacy and safety with other treatment modalities is a crucial step, and the role of clinicians and surgeons is integral in optimizing patient care. This review can also inform neoadjuvant therapies and enhance surgical education, facilitating more informed decision-making processes between surgeons and patients. METHODS This comprehensive systematic review results from rigorous screening. Following a rigorous screening process of the PubMed, PubMed Central, and Medline databases by quality assessment and application of inclusion/exclusion criteria filters, 9 relevant articles were identified that directly addressed the research question and provided a holistic understanding of it. The analysis included a total of 1403 patients from 9 different studies. Alectinib was given to 836 patients, while 567 patients received other chemotherapeutic drugs. The primary objective of this study was to evaluate and compare the efficacy of alectinib with other treatment modalities. RESULTS The analysis revealed that alectinib is promising for ALK-positive NSCLC cases, with significantly better efficacy and a positive impact on limiting central nervous system metastases. Alectinib's favorable safety profile, with medically manageable adverse events, provides reassurance about its safety compared with other treatment modalities. CONCLUSIONS Alectinib has emerged as a viable, significantly superior treatment option for patients with ALK-positive NSCLC. The superior efficacy and manageable safety profile are significant; it remains a novel therapy with much potential, such as neoadjuvant therapy, which will make significant strides in patient care of ALK-positive NSCLC. Therefore, it is crucial for healthcare professionals, including surgeons, to be well-versed in alectinib and its potential. This knowledge will empower them and instill confidence in their ability to provide the best care for their patients.
Collapse
Affiliation(s)
- Monica Kharat
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Queens Hospital Center, New York, NY
| | - Harshitha Nookala
- Department of Medicine, Mamata Academy of Medical Sciences, Telangana, Hyderabad, India
| | - Jabez David John
- Department of Medicine, Malla Reddy Institute of Medical Sciences, Telangana, Hyderabad, India
| | - Tirath Patel
- Department of Medicine, Trinity Medical Sciences University School of Medicine, Ribishi, Saint Vincent and Grenadines
| | - Pragnesh Kalyandrug
- Department of Medicine, Trinity Medical Sciences University School of Medicine, Ribishi, Saint Vincent and Grenadines
| | - Sonya Emmanuel
- Department of Medicine, California Unit of Behavioral Science and Psychology, Fairfield, CA
| | - Anaya Noor
- Department of Medicine, Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | | | - Abhishek Goyal
- Department of Neurology, JFK University Medical Center, Edison, NJ
| | - Nikhilesh Anand
- Department of Medical Education, University of Texas Rio Grande Valley, Edinburg, TX
| | - Bharathi Gadad
- Department of Medical Education, University of Texas Rio Grande Valley, Edinburg, TX
| | - Richard M. Millis
- Department of Pathophysiology, American University of Antigua, Antigua and Barbuda, Saint John
| |
Collapse
|
23
|
Shi Z, Hu C, Liu J, Cheng W, Chen X, Liu X, Bao Y, Tian H, Yu B, Gao F, Ye F, Jin X, Sun C, Li Q. Single-Cell Sequencing Reveals the Role of Radiation-Induced Stemness-Responsive Cancer Cells in the Development of Radioresistance. Int J Mol Sci 2025; 26:1433. [PMID: 40003899 PMCID: PMC11855645 DOI: 10.3390/ijms26041433] [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: 01/16/2025] [Revised: 01/29/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Increased stemness of cancer cells exacerbates radioresistance, thereby greatly limiting the efficacy of radiotherapy. In order to study the changes in cancer cell stemness during radiotherapy, we established a radioresistance model of human non-small cell lung cancer A549 cells and obtained A549 radioresistant cells (A549-RR). We sampled the cells at different time points during the modeling process and investigated the heterogeneity of each group of cells using single-cell sequencing. Cells in the early stages of fractionated irradiation were found to be significantly up-regulated in stemness, and a subpopulation of cells producing this response was screened and referred to as "radiation-induced stemness-responsive cancer cells". They were undergoing stemness response, energy metabolism reprogramming, and progressively differentiating into cells with more diverse and malignant phenotypes in order to attenuate the killing effect of radiation. Furthermore, we demonstrated that such responses might be driven by the activation of the EGFR-Hippo signaling pathway axis, which also plays a crucial role in the development of radioresistance. Our study reveals the dynamic evolution of cell subpopulation in cancer cells during fractionated radiotherapy; the early stage of irradiation can determine the destiny of the radiation-induced stemness-responsive cancer cells. The activation of stemness-like phenotypes during the development of radioresistance is not the result of dose accumulation but occurs during the early stage of radiotherapy with relatively low-dose irradiation. The degree of the radiation-induced stemness response of cancer cells mediated by the EGFR-Hippo signaling pathway might be a potential predictor of the efficacy of radiotherapy and the development of radioresistance.
Collapse
Affiliation(s)
- Zheng Shi
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (Z.S.); (C.H.); (J.L.); (W.C.); (X.C.); (X.L.); (Y.B.); (H.T.); (B.Y.); (F.G.); (F.Y.); (X.J.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Gansu Provincial Key Laboratory of Ion Beam Medicine Research, Lanzhou 730000, China
- College of Biopharmaceutical and Engineering, Lanzhou Jiaotong University, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Cuilan Hu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (Z.S.); (C.H.); (J.L.); (W.C.); (X.C.); (X.L.); (Y.B.); (H.T.); (B.Y.); (F.G.); (F.Y.); (X.J.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Gansu Provincial Key Laboratory of Ion Beam Medicine Research, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Jiadi Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (Z.S.); (C.H.); (J.L.); (W.C.); (X.C.); (X.L.); (Y.B.); (H.T.); (B.Y.); (F.G.); (F.Y.); (X.J.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Gansu Provincial Key Laboratory of Ion Beam Medicine Research, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Wei Cheng
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (Z.S.); (C.H.); (J.L.); (W.C.); (X.C.); (X.L.); (Y.B.); (H.T.); (B.Y.); (F.G.); (F.Y.); (X.J.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Gansu Provincial Key Laboratory of Ion Beam Medicine Research, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xiaohua Chen
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (Z.S.); (C.H.); (J.L.); (W.C.); (X.C.); (X.L.); (Y.B.); (H.T.); (B.Y.); (F.G.); (F.Y.); (X.J.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Gansu Provincial Key Laboratory of Ion Beam Medicine Research, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xiongxiong Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (Z.S.); (C.H.); (J.L.); (W.C.); (X.C.); (X.L.); (Y.B.); (H.T.); (B.Y.); (F.G.); (F.Y.); (X.J.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Gansu Provincial Key Laboratory of Ion Beam Medicine Research, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Yanyu Bao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (Z.S.); (C.H.); (J.L.); (W.C.); (X.C.); (X.L.); (Y.B.); (H.T.); (B.Y.); (F.G.); (F.Y.); (X.J.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Gansu Provincial Key Laboratory of Ion Beam Medicine Research, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Haidong Tian
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (Z.S.); (C.H.); (J.L.); (W.C.); (X.C.); (X.L.); (Y.B.); (H.T.); (B.Y.); (F.G.); (F.Y.); (X.J.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Gansu Provincial Key Laboratory of Ion Beam Medicine Research, Lanzhou 730000, China
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China
| | - Boyi Yu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (Z.S.); (C.H.); (J.L.); (W.C.); (X.C.); (X.L.); (Y.B.); (H.T.); (B.Y.); (F.G.); (F.Y.); (X.J.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Gansu Provincial Key Laboratory of Ion Beam Medicine Research, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Feifei Gao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (Z.S.); (C.H.); (J.L.); (W.C.); (X.C.); (X.L.); (Y.B.); (H.T.); (B.Y.); (F.G.); (F.Y.); (X.J.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Gansu Provincial Key Laboratory of Ion Beam Medicine Research, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Fei Ye
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (Z.S.); (C.H.); (J.L.); (W.C.); (X.C.); (X.L.); (Y.B.); (H.T.); (B.Y.); (F.G.); (F.Y.); (X.J.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Gansu Provincial Key Laboratory of Ion Beam Medicine Research, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (Z.S.); (C.H.); (J.L.); (W.C.); (X.C.); (X.L.); (Y.B.); (H.T.); (B.Y.); (F.G.); (F.Y.); (X.J.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Gansu Provincial Key Laboratory of Ion Beam Medicine Research, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Chao Sun
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (Z.S.); (C.H.); (J.L.); (W.C.); (X.C.); (X.L.); (Y.B.); (H.T.); (B.Y.); (F.G.); (F.Y.); (X.J.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Gansu Provincial Key Laboratory of Ion Beam Medicine Research, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (Z.S.); (C.H.); (J.L.); (W.C.); (X.C.); (X.L.); (Y.B.); (H.T.); (B.Y.); (F.G.); (F.Y.); (X.J.)
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Gansu Provincial Key Laboratory of Ion Beam Medicine Research, Lanzhou 730000, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| |
Collapse
|
24
|
Chen L, Yang R, Li W, Wen X, Li Y, Tang J, Hu J, Kou Q. Nanoparticle-aptamer based cytosensing for the detection of human non-small cell lung cancer cells. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2025; 17:1354-1361. [PMID: 39835521 DOI: 10.1039/d4ay02183e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
This study reports a simple and rapid aptamer-based sensor platform designed for the sensitive and selective detection of human non-small cell lung cancer (NSCLC) cells. Under standard conditions, gold nanoparticles (AuNPs) remain dispersed and exhibit a characteristic peak at 520 nm. However, the addition of sodium chloride (NaCl) destabilizes the charge of the solution, leading to the aggregation of AuNPs. The AS1411 aptamer can adsorb onto the surface of AuNPs, effectively preventing their aggregation. In the presence of A549 cells, the AS1411 aptamer is induced to form stable G-tetrads, which allows for specific binding to the cells and results in the aggregation of AuNPs in the NaCl solution. This proposed aptasensor platform demonstrates high specificity for A549 cells when compared to other control human normal cells. The method exhibits a dynamic range of 101 to 106 cells per mL, with a detection limit of 7 cells per mL.
Collapse
Affiliation(s)
- Lianju Chen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.
- Jilin Medical University, Jilin, 132013, China
| | - Rong Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Wenwei Li
- College of Life Sciences, Guizhou University, Guiyang City, Guizhou Province, 550000, China
| | - Xu Wen
- College of Food Engineering, Chongqing Vocational College of Light Industry, Chongqing, 401329, China
| | - Yue Li
- Sichuan University, Chengdu, Sichuan, 610065, China
| | - Jiaming Tang
- Sichuan University, Chengdu, Sichuan, 610065, China
| | - Jing Hu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Qiming Kou
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.
- Jilin Medical University, Jilin, 132013, China
- Sichuan University, Chengdu, Sichuan, 610065, China
| |
Collapse
|
25
|
Villalona-Calero MA, Tian L, Li X, Palmer JM, Aceves C, Meisen H, Cortez C, Synold TW, Egelston C, VanDeusen J, Bruno I, Zhang L, Romeu-Bonilla E, Butt O, Forman SJ, Caligiuri MA, Yu J. Interim report on engineered NK cell trial in lung cancer refractory to immune checkpoint inhibitors. JCI Insight 2025; 10:e186890. [PMID: 39903538 PMCID: PMC11949060 DOI: 10.1172/jci.insight.186890] [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] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) remains the leading cause of cancer-related mortality, necessitating the exploration of alternate therapeutic approaches. Tumor-reactive or activated-by-cytokine killers (TRACK) are PD-L1+, highly cytolytic NK cells derived from umbilical cord blood NK cells and engineered to express soluble IL-15 (sIL15), and these cells show promise in preclinical studies against NSCLC. METHODS We assessed safety, persistence, homing, and cytotoxic activity in 6 patients with advanced, refractory, and progressing NSCLC who received a low dose of unmatched, allogeneic, off-the-shelf sIL15_TRACK NK cells. We evaluated NK cell presence and persistence with droplet digital PCR (ddPCR), flow cytometry, and immunofluorescence staining. RESULTS sIL15_TRACK NK cells had peak measurements at 1 hour and became undetectable 4 hours after each infusion. Cognate ligands to activating NK cell receptors were found in NSCLC. sIL15_TRACK NK cells were observed in a lung tumor biopsy 7 days after the final infusion, confirming their sustainment and tumor-homing ability. They retained cytolytic function following isolation from the lung tumor. Three of 6 patients achieved disease stabilization on repeat imaging, while the others progressed. CONCLUSION Unmatched, allogeneic, cryopreserved, off-the-shelf sIL15_TRACK NK cells express activating receptors, home to tumor sites that express their cognate ligands, and retain cytolytic activity after infusion, underscoring their potential as a therapeutic approach in solid tumors. At low doses, the therapy was safely administered and showed preliminary evidence of activity in 3 of 6 patients with advanced and progressive NSCLC. Additional dose escalation cohorts and coadministration with atezolizumab are planned. TRIAL REGISTRATION CLINICALTRIALS gov NCT05334329. FUNDING Funding was provided by CytoImmune Therapeutics and grants from the National Cancer Institute (CA266457, CA033572, and CA210087).
Collapse
Affiliation(s)
- Miguel A. Villalona-Calero
- The Department of Medical Oncology and Experimental Therapeutics, Beckman Research Institute and Comprehensive Cancer Center
| | - Lei Tian
- Hematologic Malignancies Research Institute, Department of Hematology and Hematopoietic Stem Cell Transplantation
| | - Xiaochen Li
- Division of Biostatistics, Department of Computational and Quantitative Medicine
| | - Joycelynne M. Palmer
- Hematologic Malignancies Research Institute, Department of Hematology and Hematopoietic Stem Cell Transplantation
- Division of Biostatistics, Department of Computational and Quantitative Medicine
- Beckman Research Institute and Comprehensive Cancer Center, and
| | - Claudia Aceves
- Beckman Research Institute, City of Hope National Medical Center, Los Angeles, California, USA
| | - Hans Meisen
- Beckman Research Institute, City of Hope National Medical Center, Los Angeles, California, USA
| | - Catherine Cortez
- Beckman Research Institute, City of Hope National Medical Center, Los Angeles, California, USA
| | - Timothy W. Synold
- Beckman Research Institute, City of Hope National Medical Center, Los Angeles, California, USA
| | - Colt Egelston
- Beckman Research Institute, City of Hope National Medical Center, Los Angeles, California, USA
| | | | - Ivone Bruno
- CytoImmune Therapeutics, Los Angeles, California, USA
| | - Lei Zhang
- CytoImmune Therapeutics, Los Angeles, California, USA
| | | | - Omer Butt
- CytoImmune Therapeutics, Los Angeles, California, USA
| | - Stephen J. Forman
- Hematologic Malignancies Research Institute, Department of Hematology and Hematopoietic Stem Cell Transplantation
- Beckman Research Institute and Comprehensive Cancer Center, and
| | - Michael A. Caligiuri
- Hematologic Malignancies Research Institute, Department of Hematology and Hematopoietic Stem Cell Transplantation
- Beckman Research Institute and Comprehensive Cancer Center, and
| | - Jianhua Yu
- Hematologic Malignancies Research Institute, Department of Hematology and Hematopoietic Stem Cell Transplantation
- Beckman Research Institute and Comprehensive Cancer Center, and
- Institute for Precision Cancer Therapeutics and Immuno-Oncology, Chao Family Comprehensive Cancer Center, Orange, California, USA
- The Clemons Family Center for Transformative Cancer Research, University of California, Irvine, California, USA
| |
Collapse
|
26
|
Li M, Gao J, Qi X, Li B, Zhao Y, Liu X, Zhang G, Wang H, Tong T. Super-Resolution Fluorescence Imaging Reveals the Mechanism of NRP1 Clustering on Non-Small-Cell Lung Cancer Membranes. Anal Chem 2025; 97:2326-2334. [PMID: 39841049 DOI: 10.1021/acs.analchem.4c05675] [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: 01/23/2025]
Abstract
Neuropilin 1 (NRP1) is upregulated in various types of malignant tumors, especially non-small-cell lung cancer (NSCLC). However, the precise mechanisms for membrane localization and regulation are not fully understood. Observations from super-resolution microscopy have revealed that NRP1 tends to form nanoscale clusters on the cell membrane, with these clusters varying significantly in size and density across different regions. Further research has shown that stimulation by hepatocyte growth factor (HGF) can reorganize the distribution of NRP1, reducing the number of small clusters while promoting the formation of larger ones. This suggests a propensity for internalization after activation. Additionally, dual-color dSTORM imaging has demonstrated a certain degree of colocalization between NRP1 and c-MET, indicating that c-MET plays an important role in stabilizing NRP1 clusters. This study provides new insights into the mechanism behind NRP1's clustered distribution on cell membranes and paves the way for developing more effective therapeutic strategies targeting NRP1 within tumors.
Collapse
Affiliation(s)
- Meng Li
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130041, P. R. China
| | - Jing Gao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Xiao Qi
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130041, P. R. China
| | - Baofeng Li
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130041, P. R. China
| | - Yinghao Zhao
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130041, P. R. China
| | - Xiangyu Liu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Guangxin Zhang
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130041, P. R. China
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
- University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Ti Tong
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130041, P. R. China
| |
Collapse
|
27
|
Yu F, Zheng S, Yu C, Gao S, Shen Z, Nar R, Liu Z, Huang S, Wu L, Gu T, Qian Z. KRAS mutants confer platinum resistance by regulating ALKBH5 posttranslational modifications in lung cancer. J Clin Invest 2025; 135:e185149. [PMID: 39960727 PMCID: PMC11910214 DOI: 10.1172/jci185149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 01/24/2025] [Indexed: 03/18/2025] Open
Abstract
Constitutively active mutations of KRAS are prevalent in non-small cell lung cancer (NSCLC). However, the relationship between these mutations and resistance to platinum-based chemotherapy and the underlying mechanisms remain elusive. In this study, we demonstrate that KRAS mutants confer resistance to platinum in NSCLC. Mechanistically, KRAS mutants mediate platinum resistance in NSCLC cells by activating ERK/JNK signaling, which inhibits AlkB homolog 5 (ALKBH5) N6-methyladenosine (m6A) demethylase activity by regulating posttranslational modifications (PTMs) of ALKBH5. Consequently, the KRAS mutant leads to a global increase in m6A methylation of mRNAs, particularly damage-specific DNA-binding protein 2 (DDB2) and XPC, which are essential for nucleotide excision repair. This methylation stabilized the mRNA of these 2 genes, thus enhancing NSCLC cells' capability to repair platinum-induced DNA damage and avoid apoptosis, thereby contributing to drug resistance. Furthermore, blocking KRAS-mutant-induced m6A methylation, either by overexpressing a SUMOylation-deficient mutant of ALKBH5 or by inhibiting methyltransferase-like 3 (METTL3) pharmacologically, significantly sensitizes KRAS-mutant NSCLC cells to platinum drugs in vitro and in vivo. Collectively, our study uncovers a mechanism that mediates KRAS-mutant-induced chemoresistance in NSCLC cells by activating DNA repair through the modulation of the ERK/JNK/ALKBH5 PTM-induced m6A modification in DNA damage repair-related genes.
Collapse
MESH Headings
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/drug therapy
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Drug Resistance, Neoplasm/genetics
- Proto-Oncogene Proteins p21(ras)/genetics
- Proto-Oncogene Proteins p21(ras)/metabolism
- AlkB Homolog 5, RNA Demethylase/metabolism
- AlkB Homolog 5, RNA Demethylase/genetics
- Protein Processing, Post-Translational
- Animals
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Mice
- Mutation
- Cell Line, Tumor
- MAP Kinase Signaling System/drug effects
- Mice, Nude
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
Collapse
Affiliation(s)
- Fang Yu
- Department of Medicine, University of Florida Health Cancer Center and
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| | - Shikan Zheng
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
| | - Chunjie Yu
- Department of Medicine, University of Florida Health Cancer Center and
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| | - Sanhui Gao
- Department of Medicine, University of Florida Health Cancer Center and
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| | - Zuqi Shen
- Department of Medicine, University of Florida Health Cancer Center and
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| | - Rukiye Nar
- Department of Medicine, University of Florida Health Cancer Center and
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| | - Zhexin Liu
- Department of Medicine, University of Florida Health Cancer Center and
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| | - Shuang Huang
- Department of Anatomy & Cell Biology, University of Florida, Gainesville, Florida, USA
| | - Lizi Wu
- Department of Molecular Genetics and Microbiology, University of Florida Health Cancer Center, University of Florida Genetic Institute, University of Florida, Gainesville, Florida, USA
| | - Tongjun Gu
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
- Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Zhijian Qian
- Department of Medicine, University of Florida Health Cancer Center and
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
28
|
Jin D, Ni X, Tan Y, Yin H, Fan G. Radiomics based on dual-layer spectral detector CT for predicting EGFR mutation status in non-small cell lung cancer. J Appl Clin Med Phys 2025; 26:e14616. [PMID: 39673508 PMCID: PMC11799912 DOI: 10.1002/acm2.14616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 11/22/2024] [Accepted: 11/28/2024] [Indexed: 12/16/2024] Open
Abstract
OBJECTIVE To explore the value of dual-layer spectral computed tomography (DLCT)-based radiomics for predicting epidermal growth factor receptor (EGFR) mutation status in patients with non-small cell lung cancer (NSCLC). METHODS DLCT images and clinical information from 115 patients with NSCLC were collected retrospectively and randomly assigned to a training group (n = 81) and a validation group (n = 34). A radiomics model was constructed based on the DLCT radiomic features by least absolute shrinkage and selection operator (LASSO) dimensionality reduction. A clinical model based on clinical and CT features was established. A nomogram was built combining the radiomic scores (Radscores) and clinical factors. Receiver operating characteristic (ROC) analysis and decision curve analysis (DCA) were used for the efficacy and clinical value of the models assessment. RESULTS A total of six radiomic features and two clinical features were screened for modeling. The AUCs of the radiomic model, clinical model, and nomogram were 0.909, 0.797, and 0.922, respectively, in the training group and 0.874, 0.691, and 0.881, respectively, in the validation group. The AUCs of the nomogram and the radiomics model were significantly higher than that of the clinical model, but no significant difference was found between them. DCA revealed that nomogram had the greatest clinical benefit at most threshold intervals. CONCLUSION Nomogram integrating clinical factors and pretreatment DLCT radiomic features can help evaluate the EGFR mutation status of patients with NSCLC in a noninvasive way.
Collapse
Affiliation(s)
- Dan Jin
- Department of RadiologyThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouChina
| | - Xiaoqiong Ni
- Department of RadiologyThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yanhuan Tan
- Department of RadiologyChangshu Hospital Affiliated to Nanjing University of Chinese MedicineSuzhouChina
| | - Hongkun Yin
- Department of Advanced ResearchInfervision Medical Technology Co. LtdBeijingChina
| | - Guohua Fan
- Department of RadiologyThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouChina
| |
Collapse
|
29
|
Subramanian J, Gregg J, Berktas M, Li J, Leighl NB. EGFR testing practices, treatment choice and clinical outcomes in advanced NSCLC in a real-world setting: A retrospective analysis of a US-based electronic health records database. Lung Cancer 2025; 201:108412. [PMID: 39933217 DOI: 10.1016/j.lungcan.2025.108412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 02/13/2025]
Abstract
OBJECTIVES Guidelines recommend epidermal growth factor receptor (EGFR) mutation testing for patients with advanced non-small cell lung cancer (NSCLC) and initiation of first-line EGFR tyrosine kinase inhibitors (EGFR-TKIs) for EGFR mutation-positive (EGFRm) NSCLC. We analyzed a nationwide electronic health record-derived de-identified database to describe EGFR testing practices, treatment choice, and outcomes in patients from the United States (US) with advanced NSCLC. MATERIALS AND METHODS Adults diagnosed with stage IIIB-IV NSCLC January 2015-January 2020, who received first-line treatment from a network of ∼280 US cancer clinics were included. Demographics/characteristics, EGFR status, time from advanced diagnosis to EGFR test result, first-line treatment, time from treatment initiation to discontinuation/death (TTD), next treatment/death (TTNTD), and overall survival (OS) were extracted. RESULTS 12,577/16,309 (77 %) eligible patients had an EGFR test recorded; 1,914/12,577 (15 %) patients had EGFRm NSCLC. Of 1,778 patients with confirmed EGFRm NSCLC before first-line treatment, 75 % received first-line EGFR-TKIs, 11 % chemotherapy, 9 % immunotherapy, and 4 % other treatment. Of 136 patients with an EGFRm result after initiating first-line treatment, 13 % received EGFR-TKIs, 50 % chemotherapy, 19 % immunotherapy, and 18 % other treatment in first-line. Among patients with EGFRm NSCLC, median time from advanced diagnosis to EGFR test result was shorter in patients who received first-line EGFR-TKIs versus first-line chemotherapy/immunotherapy/other treatment. Patients treated with first-line EGFR-TKIs had significantly improved TTD/TTNTD versus those who received first-line chemotherapy/immunotherapy/other treatment (p < 0.001). OS was significantly longer in patients receiving treatment ≥21 versus <21 days after index (p < 0.001). CONCLUSIONS Nearly one-quarter of patients with advanced NSCLC in a US health network were not tested for EGFR mutations. Of patients who received a EGFRm result after initiating first-line treatment, 13 % received first-line EGFR-TKIs. These real-world data support the need to improve EGFR testing implementation and time to result to optimize first-line treatment for advanced NSCLC.
Collapse
Affiliation(s)
- Janakiraman Subramanian
- Saint Luke's Cancer Institute, Kansas City, MO, USA; Inova Schar Cancer Institute, Fairfax, VA, USA.
| | - Jeffrey Gregg
- Department of Pathology, University of Nevada, Reno, School of Medicine, Reno, NV, USA.
| | - Mehmet Berktas
- Oncology Outcome Research, AstraZeneca, Cambridge, United Kingdom.
| | - Jingyi Li
- Global Medical Affairs, AstraZeneca, Gaithersburg, MD, USA.
| | | |
Collapse
|
30
|
Rao Z, Wang Z, Deng H, Su W, Huang X, Xu Z. Role of Traditional Chinese Medicine in Lung Cancer Management: A Review. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2025; 53:97-117. [PMID: 39880665 DOI: 10.1142/s0192415x25500053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
With the continuous advancements in modern medicine, significant progress has been made in the treatment of lung cancer. Current standard treatments, such as surgery, chemotherapy, radiotherapy, targeted therapy, and immunotherapy, have notably improved patient survival. However, the adverse effects associated with these therapies limit their use and impact the overall treatment process. Traditional Chinese medicine (TCM) has shown holistic, multi-target, and multi-level therapeutic effects. Numerous studies have highlighted the importance of TCM's role in the comprehensive management of lung cancer, demonstrating its benefits in inhibiting tumor growth, reducing complications, mitigating side effects, and enhancing the efficacy of conventional treatments. Here, we review the main mechanisms of TCM in combating lung cancer, inducing cancer cell cycle arrest and apoptosis. These include inhibiting lung cancer cell growth and proliferation, inhibiting cancer cell invasion and metastasis, suppressing angiogenesis and epithelial-mesenchymal transition (EMT), and modulating antitumor inflammatory responses and immune evasion. This paper aims to summarize recent advancements in the application of TCM for lung cancer, emphasizing its unique advantages and distinctive features. In promoting the benefits of TCM, we seek to provide valuable insights for the integrated treatment of lung cancer.
Collapse
Affiliation(s)
- Zhijing Rao
- Oncology Department, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine Shanghai, P. R. China
| | - Zhongqi Wang
- Oncology Department, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine Shanghai, P. R. China
| | - Haibin Deng
- Oncology Department, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine Shanghai, P. R. China
| | - Wan Su
- Oncology Department, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine Shanghai, P. R. China
| | - Xiaowei Huang
- Oncology Department, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine Shanghai, P. R. China
| | - Zhenye Xu
- Oncology Department, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine Shanghai, P. R. China
| |
Collapse
|
31
|
Stawarska A, Bamburowicz-Klimkowska M, Pisklak DM, Gawlak M, Grudzinski IP. A Pilot Study on Qualitative Metabolomics to Characterize Lewis Lung Carcinoma in Mice. Life (Basel) 2025; 15:202. [PMID: 40003611 PMCID: PMC11857005 DOI: 10.3390/life15020202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/18/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Metabolomics is a powerful tool that can be used to identify different stages in cancer development. In this study, the metabolomic profile of Lewis lung carcinoma (LLC) was characterized in C57BL/6 mice bearing LLC tumors. Magnetic resonance spectroscopy (nuclear magnetic resonance-NMR) was applied using a 400 MHz 1H NMR spectrometer. Two types of metabolites (polar and non-polar) were identified on LLC based on the analysis of methanol/water and chloroform extracts collected from lung cancer samples in mice. The investigated metabolomics show that the neoplastic processes of growing LLC on mice may affect carbohydrate; alanine and glutamate; leucine and isoleucine; lysine; creatine; and choline metabolism, whereas hypoxia states were identified due to elevated lactate in lung cancer tissues. The metabolomic profile of Lewis lung carcinoma could be considered to be a valuable biomarker in translational lung cancer research.
Collapse
Affiliation(s)
- Agnieszka Stawarska
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland;
| | | | - Dariusz Maciej Pisklak
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland;
| | - Maciej Gawlak
- Department of Pharmacotherapy and Pharmaceutical Care, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland;
| | - Ireneusz P. Grudzinski
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland;
| |
Collapse
|
32
|
Moon GY, Dalkiran B, Park HS, Shin D, Son C, Choi JH, Bang S, Lee H, Doh I, Kim DH, Jeong WJ, Bu J. Dual Biomarker Strategies for Liquid Biopsy: Integrating Circulating Tumor Cells and Circulating Tumor DNA for Enhanced Tumor Monitoring. BIOSENSORS 2025; 15:74. [PMID: 39996976 PMCID: PMC11852634 DOI: 10.3390/bios15020074] [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: 12/28/2024] [Revised: 01/21/2025] [Accepted: 01/26/2025] [Indexed: 02/26/2025]
Abstract
The liquid biopsy has gained significant attention in cancer diagnostics, with circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) being recognized as key biomarkers for tumor detection and monitoring. However, each biomarker possesses inherent limitations that restrict its standalone clinical utility, such as the rarity and heterogeneity of CTCs and the variable sensitivity and specificity of ctDNA assays. This highlights the necessity of integrating both biomarkers to maximize diagnostic and prognostic potential, offering a more comprehensive understanding of the tumor biology and therapeutic response. In this review, we summarize clinical studies that have explored the combined analysis of CTCs and ctDNA as biomarkers, providing insights into their synergistic value in diverse tumor types. Specifically, this paper examines the individual advantages and limitations of CTCs and ctDNA, details the findings of combined biomarker studies across various cancers, highlights the benefits of dual biomarker approaches over single-biomarker strategies, and discusses future prospects for advancing personalized oncology through liquid biopsies. By offering a comprehensive overview of clinical studies combining CTCs and ctDNA, this review serves as a guideline for researchers and clinicians aiming to enhance biomarker-based strategies in oncology and informs biosensor design for improved biomarker detection.
Collapse
Affiliation(s)
- Ga Young Moon
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Basak Dalkiran
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Hyun Sung Park
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Dongjun Shin
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Chaeyeon Son
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Jung Hyun Choi
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
- Division of Biomedical Metrology, Korea Research Institute of Standards and Science, 267 Gajeongno, Yuseong-gu, Daejeon 34113, Republic of Korea; (I.D.); (D.H.K.)
| | - Seha Bang
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Hosu Lee
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Il Doh
- Division of Biomedical Metrology, Korea Research Institute of Standards and Science, 267 Gajeongno, Yuseong-gu, Daejeon 34113, Republic of Korea; (I.D.); (D.H.K.)
| | - Dong Hyung Kim
- Division of Biomedical Metrology, Korea Research Institute of Standards and Science, 267 Gajeongno, Yuseong-gu, Daejeon 34113, Republic of Korea; (I.D.); (D.H.K.)
| | - Woo-jin Jeong
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
- Department of Biological Engineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| | - Jiyoon Bu
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
- Department of Biological Engineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
- Biohybrid Systems Research Center, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| |
Collapse
|
33
|
Huang Y, Xia M, Xu C, Lin Z, Chen M, Shi X, Ding Y, Xiao Y, Zhao C. A GSH-responsive oxidative stress nanoamplifier for self-augmented chemo/chemodynamic therapy to reverse cisplatin resistance. Acta Biomater 2025; 193:440-454. [PMID: 39706539 DOI: 10.1016/j.actbio.2024.12.041] [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: 09/09/2024] [Revised: 11/27/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Drug resistance and off-target toxicity of cisplatin (CDDP) pose significant challenges in effectively treating non-small cell lung cancer (NSCLC). Recently, chemodynamic therapy (CDT), an emerging reactive oxygen species (ROS)-mediated tumor-specific therapeutic modality, has shown great potential in sensitizing multidrug resistance tumor cells. Herein, a glutathione (GSH)-responsive Pt(IV) prodrug-based oxidative stress nanoamplifier (CuBSO@PtC16) was developed for effective chemo/chemodynamic therapy to reverse CDDP resistance in NSCLC. CuBSO@PtC16, a lipid-coated nanoagent, was constructed by coordinating Cu2+ with l-buthioninesulfoximine (BSO) as the core framework, and Pt(IV) prodrug (PtC16) was concurrently loaded on the outer lipid bilayer. With appropriate particle size (∼35 nm) and good physiological stability, CuBSO@PtC16 efficiently accumulated at tumor tissue. Under high intracellular GSH levels, PtC16 was reduced to generate cytotoxic CDDP that induced cell-killing and boosted intracellular H2O2 levels, and the CuBSO core was disassembled to release Cu ions and BSO simultaneously. The released BSO could efficiently reduce the intracellular GSH content to weaken its detoxification effect on CDDP, leading to more Pt-DNA adduct formation and more severe DNA damage. Meanwhile, Cu ions catalyzed the intracellular elevated H2O2 into highly lethal •OH through Fenton-like reactions, and the reduction of GSH weakened the •OH elimination, which jointly amplified the intracellular oxidative stress levels, finally achieving enhanced chemo/chemodynamic therapeutic effect and reversing CDDP resistance in NSCLC. Therefore, this work offers an inspirational idea for effectively treating drug-resistant cancers. STATEMENT OF SIGNIFICANCE: Cisplatin (CDDP) faces challenges in treating non-small cell lung cancer (NSCLC) due to drug resistance and off-target toxicity. Herein, a GSH-responsive nanoreactor (CuBSO@PtC16) was developed for effective chemo/chemodynamic therapy to address CDDP resistance. CuBSO@PtC16 could efficiently traffic to tumor site and response to high GSH levels in tumor cells to release CDDP, Cu ions and buthioninesulfoximine (BSO) simultaneously. CDDP could induce DNA damage and boost intracellular H2O2 levels, which then served as the substrate of Cu to induce •OH generation through Fenton-like reactions. Meanwhile, the released BSO efficiently reduced the intracellular GSH content to weaken its detoxification effect on CDDP and the elimination of the •OH, leading to amplified intracellular oxidative stress and more severe damage to induce cell death.
Collapse
Affiliation(s)
- Yanjuan Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Meng Xia
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Congjun Xu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Zijun Lin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Meixu Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Xianmin Shi
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Yaqing Ding
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Yan Xiao
- Laboratory Animal Center, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Chunshun Zhao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China.
| |
Collapse
|
34
|
Kashyap P, Raj KV, Sharma J, Dutt N, Yadav P. Classification of NSCLC subtypes using lung microbiome from resected tissue based on machine learning methods. NPJ Syst Biol Appl 2025; 11:11. [PMID: 39824879 PMCID: PMC11742043 DOI: 10.1038/s41540-025-00491-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 01/07/2025] [Indexed: 01/20/2025] Open
Abstract
Classification of adenocarcinoma (AC) and squamous cell carcinoma (SCC) poses significant challenges for cytopathologists, often necessitating clinical tests and biopsies that delay treatment initiation. To address this, we developed a machine learning-based approach utilizing resected lung-tissue microbiome of AC and SCC patients for subtype classification. Differentially enriched taxa were identified using LEfSe, revealing ten potential microbial markers. Linear discriminant analysis (LDA) was subsequently applied to enhance inter-class separability. Next, benchmarking was performed across six different supervised-classification algorithms viz. logistic-regression, naïve-bayes, random-forest, extreme-gradient-boost (XGBoost), k-nearest neighbor, and deep neural network. Noteworthy, XGBoost, with an accuracy of 76.25%, and AUROC (area-under-receiver-operating-characteristic) of 0.81 with 69% specificity and 76% sensitivity, outperform the other five classification algorithms using LDA-transformed features. Validation on an independent dataset confirmed its robustness with an AUROC of 0.71, with minimal false positives and negatives. This study is the first to classify AC and SCC subtypes using lung-tissue microbiome.
Collapse
Affiliation(s)
- Pragya Kashyap
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, India
| | - Kalbhavi Vadhi Raj
- Department of Electrical Engineering, Indian Institute of Technology, Jodhpur, Rajasthan, India
| | - Jyoti Sharma
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, India
| | - Naveen Dutt
- Department of Pulmonary Medicine, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Pankaj Yadav
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, India.
- School of Artificial Intelligence and Data Science, Indian Institute of Technology, Jodhpur, Rajasthan, India.
| |
Collapse
|
35
|
Obafemi OT, Ayeleso AO, Adewale OB, Unuofin J, Ekundayo BE, Ntwasa M, Lebelo SL. Animal models in biomedical research: Relevance of Drosophila melanogaster. Heliyon 2025; 11:e41605. [PMID: 39850441 PMCID: PMC11754520 DOI: 10.1016/j.heliyon.2024.e41605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 01/25/2025] Open
Abstract
Animal models have become veritable tools in gaining insight into the pathogenesis and progression of several human diseases. These models could range in complexity from Caenorhabditis elegans to non-human primates. With the aid of these animal models, a lot of new knowledge has been gained about several diseases which otherwise would not have been possible. Most times, the utilization of these animal models is predicated on the level of homology they share with humans, which suggests that outcomes of studies using them could be extrapolated to humans. However, this has not always been the case. Drosophila melanogaster is becoming increasingly relevant as preferred model for understanding the biochemical basis of several human diseases. Apart from its relatively short lifespan, high fecundity and ease of rearing, the simplicity of its genome and lower redundancy of its genes when compared with vertebrate models, as well as availability of genetic tool kit for easy manipulation of its genome, have all contributed to its emergence as a valid animal model of human diseases. This review aimed at highlighting the contributions of selected animal models in biomedical research with a focus on the relevance of Drosophila melanogaster in understanding the biochemical basis of some diseases that have continued to plague mankind.
Collapse
Affiliation(s)
- Olabisi Tajudeen Obafemi
- Department of Life and Consumer Sciences, School of Agriculture and Life Sciences, University of South Africa, 1710, Johannesburg, South Africa
| | - Ademola Olabode Ayeleso
- Department of Life and Consumer Sciences, School of Agriculture and Life Sciences, University of South Africa, 1710, Johannesburg, South Africa
- Biochemistry Programme, College of Agriculture, Engineering and Science, Bowen University, PMB 284, Iwo, Osun State, Nigeria
| | | | - Jeremiah Unuofin
- Department of Life and Consumer Sciences, School of Agriculture and Life Sciences, University of South Africa, 1710, Johannesburg, South Africa
| | | | - Monde Ntwasa
- Department of Life and Consumer Sciences, School of Agriculture and Life Sciences, University of South Africa, 1710, Johannesburg, South Africa
| | - Sogolo Lucky Lebelo
- Department of Life and Consumer Sciences, School of Agriculture and Life Sciences, University of South Africa, 1710, Johannesburg, South Africa
| |
Collapse
|
36
|
Jiang ML, Liu L, Wang Z, Yang X, Lin Z, Jiang R, Zhang CJ, Wang W. Kanglaite alleviates lung squamous cell carcinoma through ferroptosis. Int Immunopharmacol 2025; 144:113616. [PMID: 39579539 DOI: 10.1016/j.intimp.2024.113616] [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: 09/19/2024] [Revised: 10/27/2024] [Accepted: 11/08/2024] [Indexed: 11/25/2024]
Abstract
Kanglaite, a compound predominantly composed of polyunsaturated fatty acids (PUFAs), has been employed in the clinical treatment of adenocarcinoma non-small cell lung cancer (NSCLC) in China for decades. However, its therapeutic efficacy and specific mechanism in the treatment of squamous NSCLC remains unexplored. In this study, we demonstrate that the co-treatment with ferric ion significantly enhances the cytotoxic effects of kanglaite by inducing ferroptosis in NCL-H1703, a cell line of human lung squamous cell carcinoma. Mechanistic investigations reveal that kanglaite induces mitochondrial dysfunction resulting in reactive oxygen species (ROS) excessive production, which is critical for the induction of ferroptosis. Further analysis shows that kanglaite suppresses the PI3K/AKT signaling pathway, leading to increased IP3 generation. IP3 subsequently binds to and activates IP3R, an endoplasmic reticulum (ER) calcium channel, exacerbating the excessive calcium transfer from the ER to mitochondria. The overloaded mitochondrial calcium contributes to its dysfunction and elevates ROS production. To optimize the synergistic effects of ferric ion and kanglaite, we develop a mesoporous silica-based nanodrug delivery system co-loaded with Kanglaite and Fe3O4, which offers several notable advantages, including reduced drug dosage and a faster therapeutic onset. Finally, in an NCL-H1703 xenograft model, the DMSN/Fe3O4-Kanglaite nanodrug significantly inhibited tumor growth. In conclusion, we identified the function and mechanism of kanglaite in treatment of squamous NSCLC and have developed a DMSN/Fe3O4-Kanglaite nanodrug, providing a superior therapeutic approach for the treatment of squamous NSCLC.
Collapse
Affiliation(s)
- Mei-Ling Jiang
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Li Liu
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Zilin Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 611731, China
| | - Xue Yang
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Zhiyong Lin
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Runqiu Jiang
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210000, China.
| | - Cun-Jin Zhang
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China.
| | - Weiyan Wang
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China.
| |
Collapse
|
37
|
Burt JR, Qaqish N, Stoddard G, Jridi A, Anderson PS, Woods L, Newman A, Carter MR, Ellessy R, Chamberlin J, Kabakus I. Non-small cell lung cancer in ever-smokers vs never-smokers. BMC Med 2025; 23:3. [PMID: 39757150 PMCID: PMC11702147 DOI: 10.1186/s12916-024-03844-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 12/24/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Lung cancer is a leading cause of cancer-related mortality. Non-small cell lung cancer (NSCLC) comprises 85% of cases with rising incidence among never-smokers (NS). This study seeks to compare clinical, imaging, pathology, and outcomes between NS and ever-smokers (S) NSCLC patients to identify significant differences if any. METHODS Retrospective cohort study of 155 NSCLC patients (88 S and 67 NS). The main predictor was smoking. Clinical, imaging, and pathology findings were evaluated at initial biopsy for staging. The primary outcome was all-cause mortality, and the secondary outcome was 12-month progression-free survival. RESULTS Imaging: NS and S had similar nodule size (0.81), calcification (> 0.99), and invasion of adjacent structures (> 0.99) (p values). NS slightly trended to more commonly involve the RLL vs S the RUL (p = 0.11). NS had higher numbers of extrathoracic metastases at initial biopsy for staging (p = 0.055). PATHOLOGY NS more commonly had adenocarcinoma compared to S, who had equal numbers of adenocarcinoma and squamous cell carcinoma (p = 0.001). Rates of lymphovascular and pleural invasion were similar (p = 0.84 and 0.28). Initial staging: NS were more often initially diagnosed with stage IV disease (p = 0.046), positive nodal disease (p = 0.002), and metastatic disease (p = 0.004). OUTCOMES S had a non-significant trend toward worse 12-month progression-free survival (rate ratio = 1.31, p = 0.31; HR = 1.33, p = 0.28). NS and S had similar 1-year all-cause mortality (HR = 1.06, p = 0.90). S had nearly double the risk of all-cause mortality in 5 years (HR = 1.73, p = 0.056) and 10 years (HR = 1.77, p = 0.02). Median survival was 6.6 years for NS and 3.9 years for S, with NS surviving 2.7 years longer on average (p = 0.045). CONCLUSIONS CT nodule features were similar in NS and S. NS more often had metastatic adenopathy, distant metastases, and stage IV disease at initial biopsy. Despite similar 12-month progression-free survival and 1-year all-cause mortality, S had nearly double the risk of mortality in the first 5 and 10 years post-diagnosis. TRIAL REGISTRATION Retrospectively registered.
Collapse
Affiliation(s)
- Jeremy R Burt
- Department of Radiology, Cardiothoracic Imaging, University of Utah, Spencer Fox Eccles School of Medicine, 50 N Medical Dr, Salt Lake City, UT, 84132, USA.
- Department of Radiology, Medical University of South Carolina, 171 Ashley Ave, Charleston, SC, 29425, USA.
| | - Naim Qaqish
- Department of Radiology, Cardiothoracic Imaging, University of Utah, Spencer Fox Eccles School of Medicine, 50 N Medical Dr, Salt Lake City, UT, 84132, USA
| | - Greg Stoddard
- Department of Biomedical Informatics, University of Utah, Salt Lake City, UT, 84132, USA
| | - Amani Jridi
- Department of Biomedical Informatics, University of Utah, Salt Lake City, UT, 84132, USA
| | - Parker Sage Anderson
- Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, 84132, USA
| | - Lacey Woods
- Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, 84132, USA
| | - Anna Newman
- Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, 84132, USA
| | - Malorie R Carter
- Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, 84132, USA
| | - Reham Ellessy
- University of Utah Health, Salt Lake City, UT, 84132, USA
| | - Jordan Chamberlin
- Department of Radiology, Medical University of South Carolina, 171 Ashley Ave, Charleston, SC, 29425, USA
| | - Ismail Kabakus
- Department of Radiology, Medical University of South Carolina, 171 Ashley Ave, Charleston, SC, 29425, USA
| |
Collapse
|
38
|
Yang R, Li W, Yu S, Wu Z, Zhang H, Liu X, Tao L, Li X, Huang J, Guo X. Enhanced NSCLC subtyping and staging through attention-augmented multi-task deep learning: A novel diagnostic tool. Int J Med Inform 2025; 193:105694. [PMID: 39515045 DOI: 10.1016/j.ijmedinf.2024.105694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 09/24/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES The objective of this study is to develop a novel multi-task learning approach with attention encoders for classifying histologic subtypes and clinical stages of non-small cell lung cancer (NSCLC), with superior performance compared to currently popular deep-learning models. MATERIAL AND METHODS Data were collected from six publicly available datasets in The Cancer Imaging Archive (TCIA). Following the inclusion and exclusion criteria, a total of 4548 CT slices from 758 cases were allocated. We evaluated multiple multi-task learning models that integrate attention mechanisms to resolve challenges in NSCLC subtype classification and clinical staging. These models utilized convolution-based modules in their shared layers for feature extraction, while the task layers were dedicated to histological subtype classification and staging. Each branch sequentially processed features through convolution-based and attention-based modules prior to classification. RESULTS Our study evaluated 758 NSCLC patients (mean age, 66.2 years ± 10.3; 473 men), spanning ADC and SCC cases. In the classification of histological subtypes and clinical staging of NSCLC, the MobileNet-based multi-task learning model enhanced with attention mechanisms (MN-MTL-A) demonstrated superior performance, achieving Area Under the Curve (AUC) scores of 0.963 (95 % CI: 0.943, 0.981) and 0.966 (95 % CI: 0.945, 0.982) for each task, respectively. The model significantly surpassed its counterparts lacking attention mechanisms and those configured for single-task learning, as evidenced by P-values of 0.01 or less for both tasks, according to DeLong's test. CONCLUSIONS The integration of attention encoder blocks into our multi-task learning network significantly enhanced the accuracy of NSCLC histological subtyping and clinical staging. Given the reduced reliance on precise radiologist annotation, our proposed model shows promising potential for clinical application.
Collapse
Affiliation(s)
- Runhuang Yang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China.
| | - Weiming Li
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China.
| | - Siqi Yu
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China.
| | - Zhiyuan Wu
- Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| | - Haiping Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China.
| | - Xiangtong Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China.
| | - Lixin Tao
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China.
| | - Xia Li
- Department of Mathematics and Statistics, La Trobe University, Melbourne, Australia.
| | - Jian Huang
- School of Mathematical Sciences, University College Cork, Cork, Ireland.
| | - Xiuhua Guo
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China; Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia.
| |
Collapse
|
39
|
Kumar RMR. Exosomal microRNAs: impact on cancer detection, treatment, and monitoring. Clin Transl Oncol 2025; 27:83-94. [PMID: 38971914 DOI: 10.1007/s12094-024-03590-6] [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: 04/11/2024] [Accepted: 06/30/2024] [Indexed: 07/08/2024]
Abstract
Exosomes, measuring between 30 and 150 nm in diameter, are small vesicles enclosed by a lipid bilayer membrane. They are released by various cells in the body and carry a diverse payload of molecules, including proteins, lipids, mRNA, and different RNA species such as long non-coding RNA, circular RNA, and microRNA (miRNA). With lengths of approximately 19-22 nucleotides, miRNAs constitute the predominant cargo in exosomes and serve as crucial regulators of protein biosynthesis. In cancer detection, exosomal miRNAs show promise as non-invasive biomarkers due to their stability and presence in various bodily fluids, aiding in early detection and precise diagnosis with specific miRNA signatures linked to different cancer types. Moreover, exosomal miRNAs influence treatment outcomes by affecting cellular processes like cell growth, cell death, and drug resistance, thereby impacting response to therapy. Additionally, they serve as indicators of disease progression and treatment response, providing insights that can guide treatment decisions and improve patient care. Through longitudinal studies, changes in exosomal miRNA profiles have been observed to correlate with disease progression, metastasis, and response to therapy, highlighting their potential for real-time monitoring of tumor dynamics and treatment efficacy. Understanding the intricate roles of exosomal miRNAs in cancer biology offers opportunities for developing innovative diagnostic tools and therapeutic strategies tailored to individual patients, ultimately advancing precision medicine approaches and improving outcomes for cancer patients. This review aims to provide an understanding of the role of exosomal miRNAs in cancer detection, treatment, and monitoring, shedding light on their potential for revolutionising oncology practices and patient care.
Collapse
Affiliation(s)
- Ram Mohan Ram Kumar
- Department of Pharmaceutical Biotechnology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, Karnataka, India.
| |
Collapse
|
40
|
Chen L, Wang X, Xie N, Zhang Z, Xu X, Xue M, Yang Y, Liu L, Su L, Bjaanæs M, Karlsson A, Planck M, Staaf J, Helland Å, Esteller M, Christiani DC, Chen F, Zhang R. A two-phase epigenome-wide four-way gene-smoking interaction study of overall survival for early-stage non-small cell lung cancer. Mol Oncol 2025; 19:173-187. [PMID: 39630602 PMCID: PMC11705728 DOI: 10.1002/1878-0261.13766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/05/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
High-order interactions associated with non-small cell lung cancer (NSCLC) survival may elucidate underlying molecular mechanisms and identify potential therapeutic targets. Our previous work has identified a three-way interaction among pack-year of smoking (the number of packs of cigarettes smoked per day multiplied by the number of years the person has smoked) and two DNA methylation probes (cg05293407TRIM27 and cg00060500KIAA0226). However, whether a four-way interaction exists remains unclear. Therefore, we adopted a two-phase design to identify the four-way gene-smoking interactions by a hill-climbing strategy on the basis of the previously detected three-way interaction. One CpG probe, cg16658473SHISA9, was identified with FDR-q ≤ 0.05 in the discovery phase and P ≤ 0.05 in the validation phase. Meanwhile, the four-way interaction improved the discrimination ability for the prognostic prediction model, as indicated by the area under the receiver operating characteristic curve (AUC) for both 3- and 5-year survival. In summary, we identified a four-way interaction associated with NSCLC survival among pack-year of smoking, cg05293407TRIM27, cg00060500KIAA0226 and g16658473SHISA9, providing novel insights into the complex mechanisms underlying NSCLC progression.
Collapse
Affiliation(s)
- Leyi Chen
- Department of Biostatistics, Center for Global Health, School of Public HealthNanjing Medical UniversityChina
| | - Xiang Wang
- Department of Biostatistics, Center for Global Health, School of Public HealthNanjing Medical UniversityChina
| | - Ning Xie
- Department of Biostatistics, Center for Global Health, School of Public HealthNanjing Medical UniversityChina
| | - Zhongwen Zhang
- Department of Biostatistics, Center for Global Health, School of Public HealthNanjing Medical UniversityChina
| | - Xiaowen Xu
- Department of Biostatistics, Center for Global Health, School of Public HealthNanjing Medical UniversityChina
| | - Maojie Xue
- Department of Biostatistics, Center for Global Health, School of Public HealthNanjing Medical UniversityChina
- Department of Health Inspection and Quarantine, Center for Global Health, School of Public HealthNanjing Medical UniversityChina
| | - Yuqing Yang
- Department of Biostatistics, Center for Global Health, School of Public HealthNanjing Medical UniversityChina
| | - Liya Liu
- School of Public Health, Health Science CenterNingbo UniversityChina
| | - Li Su
- Department of Environmental HealthHarvard T.H. Chan School of Public HealthBostonMAUSA
- Pulmonary and Critical Care Division, Department of MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Maria Bjaanæs
- Department of Cancer Genetics, Institute for Cancer ResearchOslo University HospitalNorway
| | - Anna Karlsson
- Division of Oncology, Department of Clinical Sciences Lund and CREATE Health Strategic Center for Translational Cancer ResearchLund UniversitySweden
| | - Maria Planck
- Division of Oncology, Department of Clinical Sciences Lund and CREATE Health Strategic Center for Translational Cancer ResearchLund UniversitySweden
| | - Johan Staaf
- Division of Oncology, Department of Clinical Sciences Lund and CREATE Health Strategic Center for Translational Cancer ResearchLund UniversitySweden
| | - Åslaug Helland
- Department of Cancer Genetics, Institute for Cancer ResearchOslo University HospitalNorway
- Institute of Clinical MedicineUniversity of OsloNorway
| | - Manel Esteller
- Josep Carreras Leukaemia Research InstituteBarcelonaSpain
- Centro de Investigacion Biomedica en Red CancerMadridSpain
- Institucio Catalana de Recerca i Estudis AvançatsBarcelonaSpain
- Physiological Sciences Department, School of Medicine and Health SciencesUniversity of BarcelonaSpain
| | - David C. Christiani
- Department of Environmental HealthHarvard T.H. Chan School of Public HealthBostonMAUSA
- Pulmonary and Critical Care Division, Department of MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Feng Chen
- Department of Biostatistics, Center for Global Health, School of Public HealthNanjing Medical UniversityChina
| | - Ruyang Zhang
- Department of Biostatistics, Center for Global Health, School of Public HealthNanjing Medical UniversityChina
- China International Cooperation Center for Environment and Human HealthNanjing Medical UniversityChina
- Changzhou Medical CenterNanjing Medical UniversityChangzhouChina
- Information CenterThe Affiliated Changzhou Second People's Hospital of Nanjing Medical UniversityChangzhouChina
| |
Collapse
|
41
|
Liguori L, Salomone F, Viggiano A, Sabbatino F, Pepe S, Formisano L, Bianco R, Servetto A. KRAS mutations in advanced non-small cell lung cancer: From biology to novel therapeutic strategies. Crit Rev Oncol Hematol 2025; 205:104554. [PMID: 39522850 DOI: 10.1016/j.critrevonc.2024.104554] [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: 05/16/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Kristen rat sarcoma viral oncogene homolog (KRAS) mutations play a major role in the carcinogenesis of many types of solid tumors including non-small cell lung cancer (NSCLC). Among KRAS mutations, p.G12C single-nucleotide variant (KRASG12C) is the most frequently reported in NSCLC patients, with a prevalence of about 12-13 %. For many decades, KRAS mutations including KRASG12C were considered "undruggable" because of the lack of effective and well-tolerated selective therapies. Noteworthy, CodeBreaK100 and KRYSTAL-1 clinical trials have recently demonstrated that sotorasib and adagrasib, two novel selective KRASG12C inhibitors, have clinical activity with acceptable adverse-event profile for the treatment of advanced NSCLC patients with KRASG12C mutation. On the other hand, no selective therapies are approved for the treatment of advanced NSCLC patients with non-G12C KRAS mutations. As a result, these patients receive the same treatments as those without KRAS mutations. In this paper, we describe the role of KRAS mutations in NSCLC focusing on the clinical and molecular characteristics which potentially identify specific subtypes of NSCLC patients based on different KRAS mutations. We also provide an overview of the main clinical trials testing novel selective KRASG12C inhibitors as well as novel potential therapeutic strategies for NSCLC patients with non-G12C KRAS mutations.
Collapse
Affiliation(s)
- Luigi Liguori
- Department of Clinical Medicine and Surgery, University of Naples II, Naples 80131, Italy; Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi 84031, Italy.
| | - Fabio Salomone
- Department of Clinical Medicine and Surgery, University of Naples II, Naples 80131, Italy.
| | - Angela Viggiano
- Department of Clinical Medicine and Surgery, University of Naples II, Naples 80131, Italy
| | - Francesco Sabbatino
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi 84031, Italy.
| | - Stefano Pepe
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi 84031, Italy.
| | - Luigi Formisano
- Department of Clinical Medicine and Surgery, University of Naples II, Naples 80131, Italy.
| | - Roberto Bianco
- Department of Clinical Medicine and Surgery, University of Naples II, Naples 80131, Italy.
| | - Alberto Servetto
- Department of Clinical Medicine and Surgery, University of Naples II, Naples 80131, Italy.
| |
Collapse
|
42
|
Beigi A, Naghib SM, Matini A, Tajabadi M, Mozafari MR. Lipid-Based Nanocarriers for Targeted Gene Delivery in Lung Cancer Therapy: Exploring a Novel Therapeutic Paradigm. Curr Gene Ther 2025; 25:92-112. [PMID: 38778601 DOI: 10.2174/0115665232292768240503050508] [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: 11/14/2023] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 05/25/2024]
Abstract
Lung cancer is a significant cause of cancer-related death worldwide. It can be broadly categorised into small-cell lung cancer (SCLC) and Non-small cell lung cancer (NSCLC). Surgical intervention, radiation therapy, and the administration of chemotherapeutic medications are among the current treatment modalities. However, the application of chemotherapy may be limited in more advanced stages of metastasis due to the potential for adverse effects and a lack of cell selectivity. Although small-molecule anticancer treatments have demonstrated effectiveness, they still face several challenges. The challenges at hand in this context comprise insufficient solubility in water, limited bioavailability at specific sites, adverse effects, and the requirement for epidermal growth factor receptor inhibitors that are genetically tailored. Bio-macromolecular drugs, including small interfering RNA (siRNA) and messenger RNA (mRNA), are susceptible to degradation when exposed to the bodily fluids of humans, which can reduce stability and concentration. In this context, nanoscale delivery technologies are utilised. These agents offer encouraging prospects for the preservation and regulation of pharmaceutical substances, in addition to improving the solubility and stability of medications. Nanocarrier-based systems possess the notable advantage of facilitating accurate and sustained drug release, as opposed to traditional systemic methodologies. The primary focus of scientific investigation has been to augment the therapeutic efficacy of nanoparticles composed of lipids. Numerous nanoscale drug delivery techniques have been implemented to treat various respiratory ailments, such as lung cancer. These technologies have exhibited the potential to mitigate the limitations associated with conventional therapy. As an illustration, applying nanocarriers may enhance the solubility of small-molecule anticancer drugs and prevent the degradation of bio-macromolecular drugs. Furthermore, these devices can administer medications in a controlled and extended fashion, thereby augmenting the therapeutic intervention's effectiveness and reducing adverse reactions. However, despite these promising results, challenges remain that must be addressed. Multiple factors necessitate consideration when contemplating the application of nanoparticles in medical interventions. To begin with, the advancement of more efficient delivery methods is imperative. In addition, a comprehensive investigation into the potential toxicity of nanoparticles is required. Finally, additional research is needed to comprehend these treatments' enduring ramifications. Despite these challenges, the field of nanomedicine demonstrates considerable promise in enhancing the therapy of lung cancer and other respiratory diseases.
Collapse
Affiliation(s)
- Anahita Beigi
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Amir Matini
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Maryam Tajabadi
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology (IUST), Narmak, Tehran, 16844, Iran
| | - Mohammad Reza Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
43
|
Onur D, İlhan Y, Büge Ö. Association between PD-L1 expression with EGFR , ALK , and ROS1 driver oncogene mutations in non-small cell lung cancer. INDIAN J PATHOL MICR 2025; 68:36-41. [PMID: 38904451 DOI: 10.4103/ijpm.ijpm_939_23] [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: 11/27/2023] [Accepted: 04/26/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Driver mutations and immunological expressions have gained importance in recent years for targeted therapies and immunotherapies of nonsmall cell lung cancer (NSCLC). AIMS This study examined the association between PD-L1 expression and ALK , ROS1 , and EGFR driver oncogene mutations in patients with NSCLC. MATERIALS AND METHODS A total of 501 NSCLC patients were included for analysis. Immunohistochemistry was performed with a PD-L1 clone 22c3. EGFR mutations were detected by PCR. ALK and ROS1 rearrangement analysis was performed with FISH. Results: There was a highly statistically significant difference between PD-L1 expression and EGFR mutation. PD-L1 expression was higher in the EGFR wild-type than in mutated EGFR ( P = 0.0002). There was no relationship between PD-L1 expression and ALK and ROS1 mutations ( P = 0.8899, P = 0.2512, respectively). PD-L1 expression was higher in nonadenocarcinomas (non-AC) than in adenocarcinomas (AC) ( P = 0.0438). The ALK rearrangement and EGFR mutations were higher in ACs ( P = 0.0073, P = 0.0012, respectively). ALK , ROS1 rearrangements, and EGFR mutations were higher in nonsmokers ( P < 0.05). EGFR mutations were detected more frequently in females than males ( P = 0.001). There was no relationship between gender and ALK , ROS1 , and PD-L1 ( P > 0.05). The prevalence of EGFR , ALK , and ROS1 driver mutations in the Turkish population was 9.3%, 5.3%, and 2.4%, respectively. CONCLUSIONS In conclusion, PD-L1 expression and mutated EGFR status have a highly negative association. PD-L1 expression was higher in EGFR wild-type patients. Therefore, it shows that the opportunity to receive PD-L1-related treatment may be higher in these patients. We think that PD-L1 immunohistochemical evaluation will increase the clinical predictive importance in EGFR wild-type cases.
Collapse
Affiliation(s)
- Dülger Onur
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
- Department of Pathology, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Yaylım İlhan
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Öz Büge
- Department of Pathology, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| |
Collapse
|
44
|
Wang T, Hong W, Yao X, Fang C, Qian X, Yu B, Zhou B, Ye X, Wang Y, Li Y. The impact of rhG-CSF on risk of recurrence after postoperative chemotherapy in NSCLC Patients: A retrospective cohort study. Int Immunopharmacol 2024; 143:113519. [PMID: 39550841 DOI: 10.1016/j.intimp.2024.113519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/19/2024]
Abstract
PURPOSE Recombinant human granulocyte colony-stimulating factor (rhG-CSF) is widespread in the prevention and treatment of blood-related toxic effects associated with chemotherapy. This study aimed to explore the correlation between rhG-CSF and the recurrence of non-small cell lung cancer (NSCLC) in patients who have undergone postoperative chemotherapy. METHODS Our study encompassed 517 NSCLC patients at pathological stage I-III, who underwent surgical removal and subsequent chemotherapy from January 2012 to December 2019 at the First Affiliated Hospital of Nanchang University. The research focused on evaluating the separate impact of rhG-CSF on the likelihood of postoperative recurrence. The analysis employed both univariate and multivariate Cox regression models. RESULTS Of 517 NSCLC patients, 123 patients did not receive rhG-CSF, while 394 patients received rhG-CSF. Unexpectedly, it was discovered that rhG-CSF usage correlated with the emergence of distant metastasis (HR: 1.8, 95 %CI 1.2-2.7, p = 0.005), though not with local recurrence (HR: 1.4, 95 %CI 0.9-2.3, p = 0.142). By multifactorial Cox analysis, rhG-CSF was an independent risk factor for distant metastasis (adjusted HR: 1.7, 95 %CI 1.0-2.6, p = 0.033). We additionally discovered that rhG-CSF could increase the risk of brain metastasis (adjusted HR: 3.9, 95 %CI 1.5-9.8, p = 0.005) and bone metastasis (adjusted HR: 3.1, 95 %CI 1.2-8.2, p = 0.02). CONCLUSION Our findings indicate that rhG-CSF independently contributes to the risk of distant metastasis, yet it shows no correlation with local recurrence. Furthermore, employing rhG-CSF played a crucial role in predicting brain metastasis and bone metastasis after postoperative chemotherapy in NSCLC patients.
Collapse
Affiliation(s)
- Tong Wang
- The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwai Zheng Road, Nanchang, China; Medical innovation center, The 1st Affiliated Hospital of Nanchang University, 17 Yongwai Zheng Road, Nanchang, China
| | - Weiwei Hong
- The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwai Zheng Road, Nanchang, China; Medical innovation center, The 1st Affiliated Hospital of Nanchang University, 17 Yongwai Zheng Road, Nanchang, China
| | - Xinyuan Yao
- The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwai Zheng Road, Nanchang, China; Medical innovation center, The 1st Affiliated Hospital of Nanchang University, 17 Yongwai Zheng Road, Nanchang, China
| | - Chen Fang
- The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwai Zheng Road, Nanchang, China
| | - Xiaoying Qian
- The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwai Zheng Road, Nanchang, China; Medical innovation center, The 1st Affiliated Hospital of Nanchang University, 17 Yongwai Zheng Road, Nanchang, China
| | - Biao Yu
- The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwai Zheng Road, Nanchang, China; Medical innovation center, The 1st Affiliated Hospital of Nanchang University, 17 Yongwai Zheng Road, Nanchang, China
| | - Bingbiao Zhou
- The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwai Zheng Road, Nanchang, China
| | - Xin Ye
- The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwai Zheng Road, Nanchang, China; Medical innovation center, The 1st Affiliated Hospital of Nanchang University, 17 Yongwai Zheng Road, Nanchang, China
| | - Yong Wang
- The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwai Zheng Road, Nanchang, China.
| | - Yong Li
- The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwai Zheng Road, Nanchang, China.
| |
Collapse
|
45
|
Wang M, Qin Y, Ai X, Liu X. RBM15-dependent m6A modification mediates progression of non-small cell lung cancer cells. Mol Med 2024; 30:267. [PMID: 39716068 DOI: 10.1186/s10020-024-01018-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 11/28/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the predominant form of lung cancer, contributing significantly to global health and economic challenges. This study elucidated the role of RBM15 in NSCLC progression through its involvement in m6A modifications. METHODS RBM15 levels in NSCLC tissues and cells were assessed via RT-qPCR and Western blotting. The impact of RBM15 knockdown on NSCLC proliferation, invasion, and migration was evaluated using CCK-8, colony formation, and Transwell assays. Expression levels of KLF1, TRIM13, and ANXA8 were determined by RT-qPCR and Western blot. m6A methylation levels were analyzed, while RIP and MeRIP assays were employed to explore the interaction between YTHDF1/YTHDF2/m6A and KLF1/TRIM13, as well as KLF1 binding to the ANXA8 promoter. The ubiquitination of ANXA8 was examined through ubiquitination assays. Xenograft and metastasis models were utilized to assess RBM15's role in vivo. RESULTS RBM15 was found to be overexpressed in NSCLC. Silencing RBM15 led to decreased cell proliferation, invasion, and migration of NSCLC cells. RBM15 upregulated KLF1 and downregulated TRIM13 via YTHDF1/YTHDF2, resulting in the promotion of ANXA8 expression. KLF1 overexpression or TRIM13 downregulation partially reversed the suppressive effects of RBM15 knockdown on NSCLC cell proliferation. ANXA8, upregulated in NSCLC, mitigated the inhibitory effects of RBM15 silencing on malignant behaviors. In vivo, RBM15 downregulation hindered NSCLC cell proliferation and metastasis by modulating the KLF1-TRIM13/ANXA8 axis. CONCLUSION RBM15-mediated m6A methylation enhances KLF1 expression and suppresses TRIM13 via YTHDF1/YTHDF2, thereby promoting ANXA8 and facilitating NSCLC progression. These findings provide novel insights and potential therapeutic targets for NSCLC treatment.
Collapse
Affiliation(s)
- Man Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Yujiao Qin
- Department of Respiratory Medicine, The First Affiliated Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Xiaoqi Ai
- Department of Respiratory Medicine, The First Affiliated Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Xiuhua Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, Jilin, China.
| |
Collapse
|
46
|
Wei CX, Althouse SK, Mamdani H, Hanna NH, Durm GA. Association of Immune-Related Adverse Events With Efficacy in Consolidation Nivolumab Plus Ipilimumab or Nivolumab Alone After Chemoradiation in Patients With Unresectable Stage III Nonsmall Cell Lung Cancer: An Exploratory Analysis From the Big 10 Cancer Research Consortium Study BTCRC LUN 16-081. Clin Lung Cancer 2024:S1525-7304(24)00270-5. [PMID: 39824659 DOI: 10.1016/j.cllc.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/20/2025]
Abstract
BACKGROUND Immunotherapy has been widely incorporated into the treatment of patients with non-small-cell lung cancer (NSCLC). Many of these patients will experience immune-related adverse events (irAEs) without decreased efficacy. We report a retrospective analysis of the association between irAEs and efficacy outcomes from the BTCRC LUN 16-081 randomized phase 2 trial of consolidation nivolumab (N) plus ipilimumab (IPI) vs N alone following chemoradiotherapy in unresectable Stage IIIA/IIIB NSCLC. RESULTS A total of 105 patients enrolled from 9/2017 to 4/2021. In arm A (N alone), 65 % of patients developed irAEs with no difference in PFS or OS in patients with and without irAEs. In arm B (IPI+N), 84 % of patients developed irAE with no difference in OS in patients with and without irAEs, but longer PFS in those who experienced irAEs (30.9 vs. 6.8mo, P = .010). Patients in Arm A that discontinued treatment due to irAE (n = 8) had shorter PFS (8.2 vs. 31.9mo, P ≤ .0001) and OS (12.3mo vs. NE, P < .0001). Patients in Arm B that discontinued treatment due to irAEs (n = 18) had no difference in either PFS or OS. CONCLUSIONS The development of irAEs after chemoradiotherapy due to the use of Nivolumab alone or in combination with Ipilimumab did not result in reduced efficacy outcomes, with an observed improvement in PFS in the combination arm. If the irAEs resulted in discontinuation of treatment, this was associated with decreased efficacy outcomes in the N alone arm but not in the IPI + N arm.
Collapse
Affiliation(s)
| | | | | | - Nasser H Hanna
- Indiana University Simon Cancer Center, Indianapolis, IN
| | - Greg A Durm
- Indiana University Simon Cancer Center, Indianapolis, IN.
| |
Collapse
|
47
|
Wang J, Zhang H, Feng Y, Gong X, Song X, Wei M, Hu Y, Li J. Aging-Related Gene-Based Prognostic Model for Lung Adenocarcinoma: Insights into Tumor Microenvironment and Therapeutic Implications. Int J Mol Sci 2024; 25:13572. [PMID: 39769336 PMCID: PMC11678022 DOI: 10.3390/ijms252413572] [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: 10/28/2024] [Revised: 11/18/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Lung cancer remains the leading cause of cancer-related mortality globally, with a poor prognosis primarily due to late diagnosis and limited treatment options. This research highlights the critical demand for advanced prognostic tools by creating a model centered on aging-related genes (ARGs) to improve prediction and treatment strategies for lung adenocarcinoma (LUAD). By leveraging datasets from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), we developed a prognostic model that integrates 14 ARGs using the least absolute shrinkage and selection operator (LASSO) alongside Cox regression analyses. The model exhibited strong predictive performance, achieving area under the curve (AUC) values greater than 0.8 for one-year survival in both internal and external validation cohorts. The risk scores generated by our model were significantly correlated with critical features of the tumor microenvironment, including the presence of cancer-associated fibroblasts (CAFs) and markers of immune evasion, such as T-cell dysfunction and exclusion. Higher risk scores correlated with a more tumor-promoting microenvironment and increased immune suppression, highlighting the model's relevance in understanding LUAD progression. Additionally, XRCC6, a protein involved in DNA repair and cellular senescence, was found to be upregulated in LUAD. Functional assays demonstrated that the knockdown of XRCC6 led to decreased cell proliferation, whereas its overexpression alleviated DNA damage, highlighting its significance in tumor biology and its potential therapeutic applications. This study provides a novel ARG-based prognostic model for LUAD, offering valuable insights into tumor dynamics and the tumor microenvironment, which may guide the development of targeted therapies and improve patient outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jianxiang Li
- Department of Toxicology, School of Public Health, Suzhou Medicine College of Soochow University, Suzhou 215123, China; (J.W.); (H.Z.); (Y.F.); (X.G.); (X.S.); (M.W.); (Y.H.)
| |
Collapse
|
48
|
Yang D, Karanth SD, Yoon HS, Yang JJ, Lou X, Bian J, Zhang D, Guo Y, Yaghjyan L, Akinyemiju T, Rodriguez E, Mehta HJ, Braithwaite D. Disparities in Utilization of Immune Checkpoint Inhibitor Therapy Among Older Patients With Advanced Non-Small Cell Lung Cancer: A SEER-Medicare Analysis. JCO ONCOLOGY ADVANCES 2024; 1:e2400008. [PMID: 39758136 PMCID: PMC11698018 DOI: 10.1200/oa.24.00008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/30/2024] [Accepted: 10/28/2024] [Indexed: 01/07/2025]
Abstract
PURPOSE In the United States, there are disparities in access to care for patients with non-small cell lung cancer (NSCLC) on the basis of socioeconomic and racial/ethnic factors. This study investigates the association between race/ethnicity and the utilization of immune checkpoint inhibitor (ICI) therapy among older patients with advanced NSCLC (aNSCLC). METHODS This retrospective study used data from the SEER-Medicare-linked database. The cohort included patients (age 66 years or older) diagnosed with aNSCLC (stage III/IV) between March 2015 and December 2017, and they were followed through December 2019. Race/ethnicity was categorized as non-Hispanic (NH)-White, NH-Black, Hispanic, and Other. ICI therapy utilization was determined by identifying any usage of ICI agents (nivolumab, pembrolizumab, atezolizumab, durvalumab, ipilimumab, and cemiplimab-rwlc) from the Medicare database. Multivariable logistic regression models assessed the association between race/ethnicity and ICI therapy utilization (yes, no). Effect measure modification analyses were conducted by sex, socioeconomic status, and comorbidity. RESULTS The final sample included 26,836 patients; 76.2% were NH-White, 10.1% NH-Black, 5.7% Hispanic, and 8.0% Other. The overall ICI therapy utilization proportion was 17.8%, varying across ethnicities: NH-Black 14.1%, Hispanic 16.3%, NH-White 18.4%, and Other 18.5%. In comparison with NH-White patients, NH-Black patients were 15% less likely to receive ICI therapy (adjusted odds ratio, 0.85 [95% CI, 0.75 to 0.96]). Furthermore, the association between race/ethnicity and utilization of ICI therapy was modified by comorbidity status, sex, and socioeconomic status. CONCLUSION NH-Black patients with aNSCLC were less likely to receive ICI therapy than their NH-White counterparts. Our findings indicate the racial/ethnic disparities in ICI therapy utilization and call for further interventions to optimize access to care.
Collapse
Affiliation(s)
- Danting Yang
- Department of Epidemiology, University of Florida College of Public Health and Health Professions, Gainesville, FL
| | - Shama D. Karanth
- University of Florida Health Cancer Center, Gainesville, FL
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL
| | - Hyung-Suk Yoon
- University of Florida Health Cancer Center, Gainesville, FL
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL
| | - Jae Jeong Yang
- University of Florida Health Cancer Center, Gainesville, FL
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL
| | - Xiwei Lou
- Department of Health Outcomes and Biomedical Informatics, University of Florida College of Medicine, Gainesville, FL
| | - Jiang Bian
- University of Florida Health Cancer Center, Gainesville, FL
- Department of Health Outcomes and Biomedical Informatics, University of Florida College of Medicine, Gainesville, FL
| | | | - Yi Guo
- University of Florida Health Cancer Center, Gainesville, FL
- Department of Health Outcomes and Biomedical Informatics, University of Florida College of Medicine, Gainesville, FL
| | - Lusine Yaghjyan
- Department of Epidemiology, University of Florida College of Public Health and Health Professions, Gainesville, FL
| | - Tomi Akinyemiju
- Department of Population Health Sciences, School of Medicine, Duke University, Durham, NC
- Duke Cancer Institute, School of Medicine, Duke University, Durham, NC
| | | | - Hiren J. Mehta
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Florida, Gainesville, FL
| | - Dejana Braithwaite
- Department of Epidemiology, University of Florida College of Public Health and Health Professions, Gainesville, FL
- University of Florida Health Cancer Center, Gainesville, FL
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL
| |
Collapse
|
49
|
Huang C, Huang X, Qiu X, Kong X, Wu C, Jiang X, Yao M, Wang M, Su L, Lv C, Wong P. Pericytes Modulate Third-Generation Tyrosine Kinase Inhibitor Sensitivity in EGFR-Mutated Lung Cancer Cells Through IL32-β5-Integrin Paracrine Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405130. [PMID: 39435643 PMCID: PMC11633494 DOI: 10.1002/advs.202405130] [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: 05/16/2024] [Revised: 09/17/2024] [Indexed: 10/23/2024]
Abstract
EGFR-mutated lung cancer patients sometimes display restricted responses to third-generation tyrosine kinase inhibitors (TKIs), potentially attributable to undervalued input from stromal cells, notably pericytes (PCs). The study shows that PCs isolated from EGFR-mutated patients have a unique secretome profile, notably secreting IL32 and affecting signaling pathways and biological processes linked to TKI sensitivity. Clinical evidence, supported by single-cell RNA sequencing and multiplex immunostaining of tumor tissues, confirms the presence of IL32-expressing pericytes closely interacting with β5-integrin-expressing cancer cells in EGFR-mutated patients, impacting therapeutic response and prognosis. Co-culture and conditioned medium experiments demonstrate that PCs reduce TKI effectiveness in EGFR-mutated cancer cells, a reversible phenomenon through silencing IL32 expression in PCs or depleting the IL32 receptor β5-integrin on cancer cells, thereby restoring cancer cell sensitivity. Mechanistically, it is shown that YY1 signaling upregulates IL32 secretion in PCs, subsequently activating the β5-integrin-Src-Akt pathway in EGFR-mutated cancer cells, contributing to their TKI sensitivity. In animal studies, co-injection of cancer cells with PCs compromises TKI effectiveness, independently of blood vessel functions, while inhibition of β5-integrin restores tumor cell sensitivity. Overall, the findings highlight direct crosstalk between cancer cells and pericytes, impacting TKI sensitivity via IL32-β5-integrin paracrine signaling, proposing an enhanced therapeutic approach for EGFR-mutated patients.
Collapse
Affiliation(s)
- Cheng Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Guangzhou Key Laboratory of Precise Diagnosis and Treatment of Biliary Tract CancerDepartment of Biliary‐Pancreatic SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Xi Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Xiaoyi Qiu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Xiangzhan Kong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Chunmiao Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Xue Jiang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Mingkang Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Department of Respiratory MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Minghui Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Department of Thoracic SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Liangping Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Guangdong Provincial Key Laboratory of Urological DiseasesGuangzhou Medical UniversityGuangzhou510120China
| | - Cui Lv
- Clinical Biobank CenterZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Ping‐Pui Wong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
- Guangzhou Key Laboratory of Precise Diagnosis and Treatment of Biliary Tract CancerDepartment of Biliary‐Pancreatic SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| |
Collapse
|
50
|
An M, Chen C, Xiang J, Li Y, Qiu P, Tang Y, Liu X, Gu Y, Qin N, He Y, Zhu M, Jiang Y, Dai J, Jin G, Ma H, Wang C, Hu Z, Shen H. Systematic identification of pathogenic variants of non-small cell lung cancer in the promoters of DNA-damage repair genes. EBioMedicine 2024; 110:105480. [PMID: 39631147 DOI: 10.1016/j.ebiom.2024.105480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Deficiency in DNA-damage repair (DDR) genes, often due to disruptive coding variants, is linked to higher cancer risk. Our previous study has revealed the association between rare loss-of-function variants in DDR genes and the risk of lung cancer. However, it is still challenging to study the predisposing role of rare regulatory variants of these genes. METHODS Based on whole-genome sequencing data from 2984 patients with non-small cell lung cancer (NSCLC) and 3020 controls, we performed massively parallel reporter assays on 1818 rare variants located in the promoters of DDR genes. Pathway- or gene-level burden analyses were performed using Firth's logistic regression or generalized linear model. FINDINGS We identified 750 rare functional regulatory variants (frVars) that showed allelic differences in transcriptional activity within the promoter regions of DDR genes. Interestingly, the burden of frVars was significantly elevated in cases (odds ratio [OR] = 1.17, p = 0.026), whereas the burden of variants prioritized solely based on bioinformatics annotation was comparable between cases and controls (OR = 1.04, p = 0.549). Among the frVars, 297 were down-regulated transcriptional activity (dr-frVars) and 453 were up-regulated transcriptional activity (ur-frVars); especially, dr-frVars (OR = 1.30, p = 0.008) rather than ur-frVars (OR = 1.06, p = 0.495) were significantly associated with risk of NSCLC. Individuals with NSCLC carried more dr-frVars from Fanconi anemia, homologous recombination, and nucleotide excision repair pathways. In addition, we identified seven genes (i.e., BRCA2, GTF2H1, DDB2, BLM, ALKBH2, APEX1, and RAD51B) with promoter dr-frVars that were associated with lung cancer susceptibility. INTERPRETATION Our findings indicate that functional promoter variants in DDR genes, in addition to protein-truncating variants, can be pathogenic and contribute to lung cancer susceptibility. FUNDING National Natural Science Foundation of China, Youth Foundation of Jiangsu Province, Research Unit of Prospective Cohort of Cardiovascular Diseases and Cancer of Chinese Academy of Medical Sciences, and Natural Science Foundation of Jiangsu Province.
Collapse
Affiliation(s)
- Mingxing An
- Department of Epidemiology, School of Public Health, Southeast University, Nanjing, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Congcong Chen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou 213003, China
| | - Jun Xiang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yang Li
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Pinyu Qiu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yiru Tang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xinyue Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yayun Gu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Na Qin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yuanlin He
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Meng Zhu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yue Jiang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Juncheng Dai
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Guangfu Jin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Hongxia Ma
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Research Units of Cohort Study on Cardiovascular Diseases and Cancers, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Cheng Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China; The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou 213003, China.
| | - Zhibin Hu
- Department of Epidemiology, School of Public Health, Southeast University, Nanjing, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China.
| | - Hongbing Shen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Research Units of Cohort Study on Cardiovascular Diseases and Cancers, Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|