1
|
Xu J, Ji Q, Deng J, Yu F. Concurrent vs. sequential chemoradiotherapy: a survival boost for lung cancer patients. Biomed Eng Online 2025; 24:60. [PMID: 40380294 PMCID: PMC12083122 DOI: 10.1186/s12938-025-01390-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 04/26/2025] [Indexed: 05/19/2025] Open
Abstract
OBJECTIVE To investigate the clinical efficacy, incidence of radiation pneumonitis, and impact on lung function of sequential chemoradiotherapy (SCRT) and concurrent chemoradiotherapy (CCRT) in the treatment of lung cancer. METHOD From January 2020 to December 2022, 158 patients with non-small cell lung cancer (NSCLC) were admitted to our hospital and chosen as the study subjects. Their clinical data were analyzed retrospectively and organized into a control group (n = 78, received SCRT) and an observation group (n = 80, received CCRT). Lesion sizes measured through CT scans were used to compare the clinical efficacy between the two groups. The study also compared the rates of adverse reactions, radiation pneumonitis, and lung function pre- and post-treatment, including forced expiratory volume in one second (FEV1), forced vital capacity (FVC), and FEV1/FVC ratio. The comparison of serum tumor marker levels was conducted between two groups, with patients being observed over a 36-month period. Kaplan-Meier survival curves were used to analyze the changes in overall survival rate (OSR), progression-free survival (PFS), and overall survival (OS) between two groups of patients. RESULTS For the observation group, the remission rate was 90.00%, and for the control group, it was 74.36%. The control rates were 96.25% for the observation group and 89.74% for the control group. Significantly higher remission and control rates were observed in the observation group than in the control group (P < 0.05). The hemoglobin reduction grade 0 was 81.2% in the observation group, compared to 58.9% in the control group. In terms of leukopenia reduction (grades 0-III) and hemoglobin reduction (grades 0-II), the observation group outperformed the control group (P < 0.05). In the observation group, 25.00% of patients experienced radiation pneumonitis, a higher rate compared to the 8.97% in the control group (P < 0.05). Overall, the control group experienced more severe radiation-induced lung injury compared to the observation group, with 6.41% of cases reaching grade IV, unlike the 0.00% in the observation group. Grade II accounted for 1.28% in the control group, a figure significantly lower than the 21.25% in the observation group (P < 0.05). Post-treatment, the FEV1, FVC, and FEV1/FVC values rose in both groups, with the observation group displaying significantly greater increases than the control group (P < 0.05). Also, after treatment, there was a decrease in CA125, SCC Ag, and CYFRA21-1 levels in both groups, with the observation group having significantly lower levels than the control group (P < 0.05). According to the Kaplan-Meier survival curve analysis, the observation group achieved an OSR of 90.00%, which exceeded the 83.33% of the control group (P > 0.05). Furthermore, PFS and OS levels were elevated in the observation group relative to the control group (P < 0.05). CONCLUSION CCRT could optimize the treatment effect for NSCLC patients by improving lung function, reducing serum tumor marker levels, and prolonging survival without increasing toxicity. Nonetheless, the occurrence of radiation pneumonitis was somewhat above expectations, and the treatment plan should be tailored to the patient's specific circumstances in clinical practice.
Collapse
Affiliation(s)
- Jinbiao Xu
- Departerment of Oncology, Gaoxin Branch of the First Affiliated Hospital of Nanchang University, No. 7889 Changdong Avenue, High-tech Zone, Nanchang, 330000, Jiangxi, China
| | - Qiao Ji
- Department of Radiation Oncology, Nanchang People's Hospital, Nanchang, 330000, Jiangxi, China
| | - Jianxiong Deng
- Departerment of Oncology, Gaoxin Branch of the First Affiliated Hospital of Nanchang University, No. 7889 Changdong Avenue, High-tech Zone, Nanchang, 330000, Jiangxi, China
| | - Feng Yu
- Departerment of Oncology, Gaoxin Branch of the First Affiliated Hospital of Nanchang University, No. 7889 Changdong Avenue, High-tech Zone, Nanchang, 330000, Jiangxi, China.
| |
Collapse
|
2
|
Rong D, Gao L, Chen Y, Gao XZ, Tang M, Tang H, Gao Y, Lu G, Ling ZQ, Shen HM. Suppression of the LKB1-AMPK-SLC7A11-GSH signaling pathway sensitizes NSCLC to albumin-bound paclitaxel via oxidative stress. Redox Biol 2025; 81:103567. [PMID: 40023979 PMCID: PMC11915006 DOI: 10.1016/j.redox.2025.103567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
Albumin-bound paclitaxel (nab-PTX) is an important chemotherapeutic drug used for the treatment of advanced and metastatic non-small cell lung cancer (NSCLC). One critical issue in its clinical application is the development of resistance; thus, a deeper understanding of the mechanisms underlying the primary resistance to nab-PTX is expected to help to develop effective therapeutic strategies to overcome resistance. In this study, we made an unexpected discovery that NSCLC with wild-type (WT) Liver kinase B1 (LKB1), an important tumor suppressor and upstream kinase of AMP-activated protein kinase (AMPK), is more resistant to nab-PTX than NSCLC with mutant LKB1. Mechanistically, LKB1 status does not alter the intracellular concentration of nab-PTX or affect its canonical pharmacological action in promoting microtubule polymerization. Instead, we found that LKB1 mediates AMPK activation, leading to increased expression of SLC7A11, a key amino acid transporter and intracellular level of glutathione (GSH), which then attenuates the production of reactive oxygen species (ROS) and apoptotic cell death induced by nab-PTX. On the other hand, genetic or pharmacological inhibition of AMPK in LKB1-WT NSCLC reduces the expression of SLC7A11 and intracellular GSH, increases ROS level, and eventually promotes the apoptotic cell death induced by nab-PTX in vitro. Consistently, the combination of nab-PTX with an AMPK inhibitor exhibits a greater therapeutic efficacy in LKB1-WT NSCLC using xenograft models in vivo. Taken together, our data reveal a novel role of LKB1-AMPK-SLC7A11-GSH signaling pathway in the primary resistance to nab-PTX, and provide a therapeutic strategy for the treatment of LKB1-WT NSCLC by targeting the LKB1-AMPK-SLC7A11-GSH pathway.
Collapse
Affiliation(s)
- Dade Rong
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Liangliang Gao
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Yiguan Chen
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Xiang-Zheng Gao
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Mingzhu Tang
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Haimei Tang
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China; Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Yuan Gao
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Guang Lu
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Qiang Ling
- Experimental Research Centre, The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Han-Ming Shen
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China.
| |
Collapse
|
3
|
Tao JH, Ruan PL, Zhang J, Zhou Y, Guan CX. Identification of the potential Pan-CDK antagonists: tracing the path of virtual screening and inhibitory activity on lung cancer cells. Mol Divers 2025; 29:1641-1653. [PMID: 39069541 DOI: 10.1007/s11030-024-10939-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024]
Abstract
Cyclin-dependent kinases (CDKs) are overexpressed in tumor cells, and their aberrant activation can promote the progression of non-small-cell lung cancer (NSCLC). We utilized structure-based virtual screening and experimental validation to screen for potential CDKs antagonists among TargetMol natural products. Molecular docking and molecular dynamics simulation results indicate that Dolastatin 10 exhibits strong interactions with multiple subtypes of CDKs (CDK1, CDK2, CDK3, CDK4, and CDK6), forming stable CDKs-Dolastatin 10 complex compounds. Furthermore, in vitro experiments demonstrate that Dolastatin 10 significantly inhibits the viability, migration, and invasion of H1299 cells in a concentration-dependent manner, arresting the cell cycle at the G2/M phase by inducing cell senescence. These findings suggest that Dolastatin 10 may serve as a potential CDKs antagonist deserving further investigation.
Collapse
Affiliation(s)
- Jia-Hao Tao
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China
| | - Ping-Lang Ruan
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, 410078, Hunan, China
| | - Jun Zhang
- Ascle Therapeutics, Suzhou, 215000, Jiangsu, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China.
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China.
| |
Collapse
|
4
|
Li K, Gui S, Wang N, Li X, Zhao C, Liu M, Zhang Z. Sequential pH/GSH-responsive stealth nanoparticles for co-delivery of anti-PD-1 antibody and paclitaxel to enhance chemoimmunotherapy of lung cancer. Eur J Med Chem 2025; 285:117273. [PMID: 39813775 DOI: 10.1016/j.ejmech.2025.117273] [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: 11/26/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 01/18/2025]
Abstract
Intravenously administered nanoparticles (NPs) often bind with plasma proteins, forming the protein corona that promotes rapid systemic clearance, a primary challenge in nanomedicine. In this study, we developed a pH- and GSH-sensitive "stealth" nanodelivery system, PTX@NPs-aPD1-IL, for sequential drug release. By using a biocompatible choline-based ionic liquid (IL) as the coating for NPs, the interaction and adsorption of NPs with serum proteins were reduced, achieving targeted delivery to the lung organ and increasing drug accumulation. In the weakly acidic extracellular tumor microenvironment (pH 6.5), the anti-PD-1 antibody (aPD-1) was first released to block the PD-1/PD-L1 pathway and restore the immunocidal function of T cells. In the highly reductive intracellular environment of tumor cells, the disulfide bonds were cleaved, causing NPs to rupture and release paclitaxel (PTX). It induced tumor cell apoptosis and triggered immunogenic cell death (ICD), promoted dendritic cells (DCs) maturation and activated T cells for chemo-immunotherapy. In the mouse orthotopic lung cancer model, PTX@NPs-aPD1-IL exhibited superior efficacy to other treatment groups at the same dose. This was due to the significantly increase in the release of immune factors, including TNF-α and IFN-γ, and the promotion of CD8+ T cells recruitment, which induced a stronger immune response, and thus enhanced the anti-lung cancer effect. In summary, PTX@NPs-aPD1-IL provided a promising strategy for effective chemo-immunotherapy for lung cancer through sequential release profile.
Collapse
Affiliation(s)
- Kan Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Shengbin Gui
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Ningning Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xiao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Chunqin Zhao
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Mingyu Liu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Zhen Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
5
|
Chen Y, Qi F, Sun C, Jiang P, Xue X, Yang X, Li X, He X, Wang Y, Zhang T. Navigating the landscape of neoadjuvant immunotherapy for NSCLC: progress and controversies. Ther Adv Med Oncol 2025; 17:17588359241312501. [PMID: 39781239 PMCID: PMC11707791 DOI: 10.1177/17588359241312501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
Recently, attention has increasingly centered on non-small-cell lung cancer (NSCLC) with immune checkpoint inhibitors application. Numerous clinical studies have underscored the potential of immunotherapy in treating resectable NSCLC, highlighting its role in improving patient outcomes. However, despite these promising results, there is ongoing debate regarding the efficacy of immunological combination therapy strategies, the prevalence of treatment-related side effects, the identification of predictive biomarkers, and various other challenges within the neoadjuvant context. Careful consideration is essential to maximize the benefits of immunotherapy for patients with resectable NSCLC. This article offers a detailed overview of recent advancements in neoadjuvant immunotherapy for resectable NSCLC. By examining these developments, we aim to provide new perspectives and valuable insights into the benefits and challenges of applying neoadjuvant immunotherapy in clinical settings.
Collapse
Affiliation(s)
- Yuzhu Chen
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Fei Qi
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Chenhao Sun
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Peng Jiang
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xiangyu Xue
- Department of Biochemistry and Molecular Biology, Heilongjiang Provincial Science and Technology Innovation Team in Higher Education Institutes for Infection and Immunity, Harbin Medical University, Harbin, China
| | - Xiaomei Yang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
- Joint Laboratory for Precision Diagnosis and Treatment Translational Research in Malignant Tumors, Gynecologic Oncology Basic and Clinical Research Laboratory, Capital Medical University, Beijing, China
| | - Xiaomi Li
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xin He
- Department of Nephrology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yishuo Wang
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Tongmei Zhang
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, No. 9 Beiguan Street, Tongzhou District, Beijing 101149, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
He G, Yang K, Gao Z, Zhang X, Han A. Drug-Eluting Beads Bronchial Arterial Chemoembolization Combined with Immunotherapy Resulted in Pathological Complete Response of Squamous Cell Lung Cancer: A Case Report. Int Med Case Rep J 2024; 17:1041-1047. [PMID: 39723431 PMCID: PMC11668917 DOI: 10.2147/imcrj.s491862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/08/2024] [Indexed: 12/28/2024] Open
Abstract
The incidence of lung cancer is the highest among all tumors, and treatment has become an urgent problem to be solved. The Drug-eluting bead-based bronchial arterial chemoembolization (DEB-BACE) combination immunotherapy is a rare neoadjuvant therapy for lung cancer surgery, which can significantly reduce the time it takes for lung cancer patients to undergo surgery.We report a male patient, aged 59-year-old, with Stage-III b squamous cell lung cancer accompanied by hemoptysis underwent surgical resection after DEB-BACE combination immunotherapy treatment 21 days later without obvious adverse events. A pathologic complete response (pCR) was observed postoperatively, and the patient has made a successful recovery. The DEB-BACE combined with immunotherapy might be a new neoadjuvant therapy option for locally advanced non-small cell lung.
Collapse
Affiliation(s)
- Guanghui He
- Interventional Department, Weifang No. 2 People’s Hospital, Weifang, People’s Republic of China
| | - Kunning Yang
- Respiratory Medicine Department, Weifang No. 2 People’s Hospital, Weifang, People’s Republic of China
| | - Zhi Gao
- Interventional Department, Weifang No. 2 People’s Hospital, Weifang, People’s Republic of China
| | - Xiaofa Zhang
- Interventional Department, Anqiu People’s Hospital of Weifang, Weifang, People’s Republic of China
| | - Aiqiang Han
- Department of Gerontology, Weifang No. 2 People’s Hospital, Weifang, People’s Republic of China
| |
Collapse
|
7
|
Avancini A, Giannarielli D, Belluomini L, Schena F, Milella M, Pilotto S. Physical Exercise During Neoadjuvant Treatments for Non-Small Cell Lung Cancer: The Time is Coming. Clin Lung Cancer 2024; 25:e431-e435. [PMID: 39127606 DOI: 10.1016/j.cllc.2024.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/10/2024] [Accepted: 07/21/2024] [Indexed: 08/12/2024]
Affiliation(s)
- Alice Avancini
- Department of Engineering for Innovation Medicine (DIMI), Section of Innovation Biomedicine - Oncology Area, University of Verona and University and Hospital Trust (AOUI) of Verona, Verona, Italy; Department of Neuroscience, Biomedicine, and Movement Sciences, University of Verona, Verona, Italy.
| | - Diana Giannarielli
- Department of Epidemiology & Biostatistic, Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Lorenzo Belluomini
- Department of Engineering for Innovation Medicine (DIMI), Section of Innovation Biomedicine - Oncology Area, University of Verona and University and Hospital Trust (AOUI) of Verona, Verona, Italy
| | - Federico Schena
- Department of Neuroscience, Biomedicine, and Movement Sciences, University of Verona, Verona, Italy
| | - Michele Milella
- Department of Engineering for Innovation Medicine (DIMI), Section of Innovation Biomedicine - Oncology Area, University of Verona and University and Hospital Trust (AOUI) of Verona, Verona, Italy
| | - Sara Pilotto
- Department of Engineering for Innovation Medicine (DIMI), Section of Innovation Biomedicine - Oncology Area, University of Verona and University and Hospital Trust (AOUI) of Verona, Verona, Italy
| |
Collapse
|
8
|
Sha C, Lee PC. EGFR-Targeted Therapies: A Literature Review. J Clin Med 2024; 13:6391. [PMID: 39518531 PMCID: PMC11546688 DOI: 10.3390/jcm13216391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Lung cancer remains the leading cause of cancer death in the United States, underscoring the critical need to optimize treatment strategies. Compared to conventional treatments such as surgical resection, radiotherapy, chemotherapy, and immunotherapy, targeted therapy stands out for its higher selectivity and minimal adverse effects. Among these, epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) are the most widely used in targeted therapy for non-small-cell lung cancer (NSCLC). In our paper, we will conduct a comprehensive review of current literature on EGFR TKIs to contribute to advancements in molecular genomics and the treatment of lung cancer.
Collapse
Affiliation(s)
| | - Paul C. Lee
- Department of Cardiothoracic Surgery, Long Island Jewish Medical Center, New Hyde Park, NY 11040, USA;
| |
Collapse
|
9
|
Shi H, Wang B, Ma H, Li Y, Du J, Zhang B, Gao Y, Liu Y, Wu C. Preparation of Biomimetic Selenium-Baicalein Nanoparticles and Their Targeted Therapeutic Application in Nonsmall Cell Lung Cancer. Mol Pharm 2024; 21:4476-4489. [PMID: 39106303 DOI: 10.1021/acs.molpharmaceut.4c00390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2024]
Abstract
In this study, we prepared bionic selenium-baicalein nanoparticles (ACM-SSe-BE) for the targeted treatment of nonsmall cell lung cancer. Due to the coating of the A549 membrane, the system has homologous targeting capabilities, allowing for the preparation of target tumor cells. The borate ester bond between selenium nanoparticles (SSe) and baicalein (BE) is pH-sensitive and can break under acidic conditions in the tumor microenvironment to achieve the targeted release of BE at the tumor site. Moreover, SSe further enhances the antitumor effect of BE by increasing the production of ROS in tumor cells. Transmission electron microscopy (TEM) images and dynamic light scattering (DLS) showed that the ACM-SSe-BE had a particle size of approximately 155 ± 2 nm. FTIR verified the successful coupling of SSe and BE. In vitro release experiments indicated that the cumulative release of ACM-SSe-BE at pH 5.5 after 24 h was 69.39 ± 1.07%, which was less than the 20% release at pH 7.4, confirming the pH-sensitive release of BE in ACM-SSe-BE. Cell uptake experiments and in vivo imaging showed that ACM-SSe-BE had good targeting ability. The results of MTT, flow cytometry, Western blot, and cell immunofluorescence staining demonstrated that ACM-SSe-BE promoted A549 cell apoptosis and inhibited cell proliferation. The in vivo antitumor results were consistent with those of the cell experiments. These results clearly suggested that ACM-SSe-BE will be a promising bionic nanosystem for the treatment of nonsmall cell lung cancer.
Collapse
Affiliation(s)
- Huan Shi
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe District, Jinzhou, Liaoning 121001, China
| | - Biaobiao Wang
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe District, Jinzhou, Liaoning 121001, China
| | - Huilin Ma
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe District, Jinzhou, Liaoning 121001, China
| | - Yunmei Li
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe District, Jinzhou, Liaoning 121001, China
| | - Jiaqun Du
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe District, Jinzhou, Liaoning 121001, China
| | - Bo Zhang
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe District, Jinzhou, Liaoning 121001, China
| | - Yu Gao
- Department of Medical Oncology, the First Affiliated Hospital of Jinzhou Medical University, the Fifth Section of Renmin Street, Guta District, Jinzhou, Liaoning 121001, China
| | - Ying Liu
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe District, Jinzhou, Liaoning 121001, China
| | - Chao Wu
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe District, Jinzhou, Liaoning 121001, China
| |
Collapse
|
10
|
Guo W, Qiao T, Li H, Zhao Y, Qin J, Zhang C, Shi C. Peripheral CD8 +PD-1 + T cells as novel biomarker for neoadjuvant chemoimmunotherapy in humanized mice of non-small cell lung cancer. Cancer Lett 2024; 597:217073. [PMID: 38906523 DOI: 10.1016/j.canlet.2024.217073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
Neoadjuvant immunotherapy has shown promising clinical activity in the treatment of early non-small cell lung cancer (NSCLC); however, further clarification of the specific mechanism and identification of biomarkers are imperative prior to implementing it as a daily practice. The study investigated the reprogramming of T cells in both tumor and peripheral blood following neoadjuvant chemoimmunotherapy in a preclinical NSCLC mouse model engrafted with a human immune system. Samples were also collected from 21 NSCLC patients (Stage IA-IIIB) who received neoadjuvant chemoimmunotherapy, and the dynamics of potential biomarkers within these samples were measured and further subjected to correlation analysis with prognosis. Further, we initially investigated the sources of the potential biomarkers. We observed in the humanized mouse model, neoadjuvant chemoimmunotherapy could prevent postoperative recurrence and metastasis by increasing the frequency and cytotoxicity of CD8+ T cells in both peripheral blood (p < 0.001) and tumor immune microenvironment (TIME) (p < 0.001). The kinetics of peripheral CD8+PD-1+ T cells reflected the changes in the TIME and pathological responses, ultimately predicting survival outcome of mice. In the clinical cohort, patients exhibiting an increase in these T cells post-treatment had a higher rate of complete or major pathological response (p < 0.05) and increased immune infiltration (p = 0.0012, r = 0.792). We identified these T cells originating from tumor draining lymph nodes and subsequently entering the TIME. In conclusion, the kinetics of peripheral CD8+PD-1+ T cells can serve as a predictor for changes in TIME and optimal timing for surgery, ultimately reflecting the outcomes of neoadjuvant chemoimmunotherapy in both preclinical and clinical setting.
Collapse
Affiliation(s)
- Wenwen Guo
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Clinical Research Center, Xianyang Central Hospital, Xianyang, Shaanxi, 712099, China
| | - Tianyun Qiao
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Hui Li
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yong Zhao
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jing Qin
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Caiqin Zhang
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
11
|
Grant C, Nagasaka M. Neoadjuvant EGFR-TKI therapy in Non-Small cell lung cancer. Cancer Treat Rev 2024; 126:102724. [PMID: 38636443 DOI: 10.1016/j.ctrv.2024.102724] [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: 01/02/2024] [Revised: 02/27/2024] [Accepted: 03/26/2024] [Indexed: 04/20/2024]
Abstract
Non-small cell lung cancer (NSCLC) stages I-III are predominantly treated with surgery and combination immunotherapy and chemotherapy. A majority of these studies excluded patients with EGFR and ALK alterations. There are several completed and ongoing trials evaluating neoadjuvant treatment with EGFR-TKI monotherapy, combination therapy with chemotherapy, and combination therapy with immunotherapy. Here, we review completed clinical trials and discuss current ongoing trials' potential benefits, challenges, and future directions in the field.
Collapse
Affiliation(s)
- Christopher Grant
- Department of Medicine, University of California Irvine Medical Center, Orange CA, USA
| | - Misako Nagasaka
- Department of Medicine, University of California Irvine Medical Center, Orange CA, USA; Division of Hematology and Oncology, Department of Medicine, University of California Irvine Medical Center, Orange CA, USA.
| |
Collapse
|