1
|
Dong W, He B, Cao Y, Yang R, Zhang S, Kong Y, Lu D, Zheng X, Hou Y, Zhu M, Wang C, Yu S, Cui D, Wang H, Wang B. Low-dose SAHA enhances CD8 + T cell-mediated antitumor immunity by boosting MHC I expression in non-small cell lung cancer. Cell Oncol (Dordr) 2025; 48:249-264. [PMID: 39283477 PMCID: PMC11850570 DOI: 10.1007/s13402-024-00989-9] [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] [Accepted: 08/30/2024] [Indexed: 02/25/2025] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is a highly aggressive type of lung cancer with poor responses to traditional therapies such as surgery, radiotherapy, and chemotherapy. While immunotherapy has become an effective approach for treating multiple types of cancer, solid tumors frequently exhibit immune escape through various mechanisms, including downregulation of MHC I expression. However, whether the upregulation of MHC I expression can improve the immunotherapeutic effect on NSCLC remains unexplored. Suberoylanilide hydroxamic acid (SAHA) is a potent histone deacetylase (HDAC) inhibitor that has been applied clinically to treat lymphoma, but a high dose of SAHA kills tumor cells and normal cells without preference. Here, we report that low-dose SAHA enhances CD8+ T cell-mediated antitumor immunity by upregulating MHC I expression in NSCLC cells. METHODS Flow cytometric analysis, quantitative real-time PCR and western blot were used to analyze the expression of MHC I, STAT1 and Smad2/3 in both human and mouse NSCLC cell lines after SAHA treatment. The nuclear translocation of phosphorylated STAT1 and Smad2/3 was investigated by western blot and immunofluorescence staining. The mechanisms underlying STAT1 and Smad2/3 upregulation were analyzed through database searches and chromatin immunoprecipitation-qPCR. Finally, we assessed the antitumor effect of specific CD8+ T cells with SAHA treatment in vivo and in vitro. RESULTS We showed that low-dose SAHA upregulated the expression of MHC I in NSCLC cell lines without affecting cell viability. We also provided evidence that high levels of MHC I induced by SAHA promoted the activation, proliferation, and cytotoxicity of specific CD8+ T cells in mouse models. Mechanistically, low-dose SAHA increased the levels of H3K9ac and H3K27ac in the promoters of the STAT1, Smad2 and Smad3 genes in NSCLC cells by inhibiting HDAC activity, resulting in elevated expression levels of STAT1, Smad2 and Smad3. The nuclear translocation of phosphorylated STAT1 and Smad2/3 markedly upregulated the expression of MHC I in NSCLC cells. CONCLUSIONS Low-dose SAHA enhances CD8+ T cell-mediated antitumor immunity by boosting MHC I expression in NSCLC cells. Thus, we revealed a key mechanism of SAHA-mediated enhanced antitumor immunity, providing insights into a novel immunotherapy strategy for NSCLC.
Collapse
Affiliation(s)
- Wenqian Dong
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, Anhui, China
| | - Bing He
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, Anhui, China
| | - Yanhong Cao
- Department of Genetic Laboratory, Affiliated Maternity and Child Health Hospital of Anhui Medical University, Maternity and Child Health Hospital of Anhui Province, Hefei, Anhui, China
| | - Rui Yang
- Department of Clinical Laboratory, Anhui No. 2 Provincial People's Hospital, Hefei, Anhui, China
| | - Shuang Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, Anhui, China
| | - Yujie Kong
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, Anhui, China
| | - Dapeng Lu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, Anhui, China
| | - Xu Zheng
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, Anhui, China
| | - Yanjiao Hou
- Department of Clinical Laboratory, Qilu Hospital of Shandong University Dezhou Hospital (Dezhou People's Hospital), 1166 Dongfanghong Road, Decheng District, Dezhou, Shandong, China
| | - Maoxin Zhu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, Anhui, China
| | - Chen Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, Anhui, China
| | - Shihao Yu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, Anhui, China
| | - Dechun Cui
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China
| | - Hao Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, Anhui, China.
| | - Baolong Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, Anhui, China.
| |
Collapse
|
2
|
Wang HL, Zhou SX, Kuang J, Xiao S, Li M. Feasibility and Tolerability of Anlotinib Plus PD-1 Inhibitors for Previously-Treated Advanced Non-Small Cell Lung Cancer: A Retrospective Exploratory Study. Biologics 2024; 18:313-326. [PMID: 39524378 PMCID: PMC11549916 DOI: 10.2147/btt.s489363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024]
Abstract
Objective Anlotinib demonstrated encouraging therapeutic activity as third-line treatment for patients with advanced non-small cell lung cancer (NSCLC). Programmed cell death protein 1 (PD-1) inhibitors also exhibited promising and durable response against previously-treated advanced NSCLC. Therefore, the present study aimed to determine the feasibility and safety of anlotinib plus PD-1 inhibitors for previously-treated NSCLC in clinical practice. Methods This retrospective study included 56 patients with advanced NSCLC treated with systemic treatment previously. Patients included were treated with anlotinib plus PD-1 inhibitors in clinical practice. Therapeutic outcomes of the patients were evaluated radiologically using target lesions, and the prognostic outcomes were generated by follow-up. Adverse reactions experienced throughout the treatment were documented and analyzed. Results Between August 2018 and November 2022, 56 patients with advanced NSCLC were eligible to participate in this study consecutively. Therapeutic outcomes resulted in an overall response rate of 28.6% [95% confidence interval (CI): 17.3%-42.2%] and a disease control rate of 91.1% (95% CI: 80.4%-97.0%). Furthermore, this combination regimen among the 56 patients yielded a median progression-free survival (PFS) of 6.5 months (95% CI: 4.81-8.19) and a median overall survival (OS) of 15.8 months (95% CI: 10.23-21.37), respectively. And the median duration of response (DoR) among patients who responded was 8.3 months (95% CI: 4.38-12.22). Additionally, adverse reactions of all grades throughout the treatment were observed in 50 patients (89.3%), and adverse reactions of grade ≥3 were detected in 23 patients (41.1%). Fatigue, hypertension, diarrhea, nausea, and vomiting were the most common adverse reactions. Association analysis between PFS and baseline characteristic subgroups indicated that ECOG score and number of metastatic lesions might be potential predictors of PFS in the exploratory analysis. Conclusion Anlotinib plus PD-1 inhibitors demonstrated a tolerable safety profile and encouraging therapeutic activity as subsequent-line therapy in patients with advanced NSCLC. This conclusion should be confirmed in prospective large-scale clinical trials subsequently.
Collapse
Affiliation(s)
- Hai-Li Wang
- Department of Oncology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450014, People’s Republic of China
| | - Shi-Xia Zhou
- Department of Oncology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450014, People’s Republic of China
| | - Jing Kuang
- Department of Oncology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450014, People’s Republic of China
| | - Sa Xiao
- Department of Oncology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450014, People’s Republic of China
| | - Min Li
- Department of Oncology, Zhengzhou People’s Hospital, Zhengzhou, 450053, People’s Republic of China
| |
Collapse
|
3
|
Restrepo JC, Martínez Guevara D, Pareja López A, Montenegro Palacios JF, Liscano Y. Identification and Application of Emerging Biomarkers in Treatment of Non-Small-Cell Lung Cancer: Systematic Review. Cancers (Basel) 2024; 16:2338. [PMID: 39001401 PMCID: PMC11240412 DOI: 10.3390/cancers16132338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 07/16/2024] Open
Abstract
Non-small-cell lung cancer (NSCLC) comprises approximately 85% of all lung cancer cases, often diagnosed at advanced stages, which diminishes the effective treatment options and survival rates. This systematic review assesses the utility of emerging biomarkers-circulating tumor DNA (ctDNA), microRNAs (miRNAs), and the blood tumor mutational burden (bTMB)-enhanced by next-generation sequencing (NGS) to improve the diagnostic accuracy, prognostic evaluation, and treatment strategies in NSCLC. Analyzing data from 37 studies involving 10,332 patients from 2020 to 2024, the review highlights how biomarkers like ctDNA and PD-L1 expression critically inform the selection of personalized therapies, particularly beneficial in the advanced stages of NSCLC. These biomarkers are critical for prognostic assessments and in dynamically adapting treatment plans, where high PD-L1 expression and specific genetic mutations (e.g., ALK fusions, EGFR mutations) significantly guide the use of targeted therapies and immunotherapies. The findings recommend integrating these biomarkers into standardized clinical pathways to maximize their potential in enhancing the treatment precision, ultimately fostering significant advancements in oncology and improving patient outcomes and quality of life. This review substantiates the prognostic and predictive value of these biomarkers and emphasizes the need for ongoing innovation in biomarker research.
Collapse
Affiliation(s)
- Juan Carlos Restrepo
- Grupo de Investigación en Salud Integral (GISI), Departamento Facultad de Salud, Universidad Santiago de Cali, Cali 760035, Colombia
| | - Darly Martínez Guevara
- Grupo de Investigación en Salud Integral (GISI), Departamento Facultad de Salud, Universidad Santiago de Cali, Cali 760035, Colombia
| | - Andrés Pareja López
- Grupo de Investigación Unidad de Toxicidad In Vitro-UTi, Facultad de Ciencias, Universidad CES, Medellin 050021, Colombia
| | | | - Yamil Liscano
- Grupo de Investigación en Salud Integral (GISI), Departamento Facultad de Salud, Universidad Santiago de Cali, Cali 760035, Colombia
| |
Collapse
|
4
|
Ma Y, Han B, Yu Q, Zha N, Deng Z, Liang J, Yu R. Single-cell and bulk RNA sequencing data jointly reveals VDAC2's impacts on prognosis and immune landscape of NSCLC. Aging (Albany NY) 2024; 16:3160-3184. [PMID: 38382091 PMCID: PMC10929798 DOI: 10.18632/aging.205517] [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: 06/08/2023] [Accepted: 11/20/2023] [Indexed: 02/23/2024]
Abstract
Non-small cell lung cancer (NSCLC) is characterized by stronger metastatic ability and worse prognosis. In NSCLC, hypoxia is a major cause of invasion and metastasis through promoting angiogenesis. In present study, NSCLC cell clusters were extracted from single cell-sequencing dataset GSE131907, which were combined with hypoxia-related genes to group clusters. qRT-PCR and western blot were used to validate the expression of target gene. Nine NSCLC clusters were extracted, which were divided into two hypoxia-related subgroups, C1 and C2. Totally 101 differentially expressed prognostic genes were identified between subgroups. Of which, VDAC2 showed excellent prognostic value for NSCLC and was selected for further analysis. VDAC2 was upregulated in tumor samples in TCGA and was correlated with advanced stages. In vitro experiments validated this trend. Five crucial immune cells showed differential infiltration proportions between high and low VDAC2 expression groups. VDAC2 knockdown significantly inhibited the proliferation and invasion ability of NSCLC cells. Integrating single cell and bulk sequencing data as well as wet lab experiments, hypoxia-related VDAC2 exhibited important prognostic value and showed the promise of becoming immune-therapy target in NSCLC.
Collapse
Affiliation(s)
- Ying Ma
- Department of Thoracic Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010059, Inner Mongolia Autonomous Region, China
| | - Bateer Han
- Department of Thoracic Surgery, Peking University Cancer Hospital (Inner Mongolia Campus) and Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot 010020, Inner Mongolia Autonomous Region, China
| | - Qin Yu
- Department of Radiation Oncology, Peking University Cancer Hospital (Inner Mongolia Campus) and Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot 010020, Inner Mongolia Autonomous Region, China
| | - Nashunbayaer Zha
- Department of Thoracic Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010059, Inner Mongolia Autonomous Region, China
| | - Zhiyuan Deng
- Department of Radiation Oncology, Peking University Cancer Hospital (Inner Mongolia Campus) and Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot 010020, Inner Mongolia Autonomous Region, China
| | - Junguo Liang
- Department of Thoracic Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010059, Inner Mongolia Autonomous Region, China
| | - Rong Yu
- Department of Radiation Oncology, Peking University Cancer Hospital (Inner Mongolia Campus) and Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot 010020, Inner Mongolia Autonomous Region, China
| |
Collapse
|
5
|
Liu L, Zhang L, Zhang X, Dong X, Jiang X, Huang X, Li W, Xie X, Qiu X. Analysis of cellular response to drugs with a microfluidic single-cell platform based on hyperspectral imaging. Anal Chim Acta 2024; 1288:342158. [PMID: 38220290 DOI: 10.1016/j.aca.2023.342158] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/07/2023] [Accepted: 12/16/2023] [Indexed: 01/16/2024]
Abstract
BACKGROUND Cellular response to pharmacological action of drugs is significant for drug development. Traditional detection method for cellular response to drugs normally rely on cell proliferation assay and metabolomics examination. In principle, these analytical methods often required cell labeling, invasion analysis, and hours of co-culture with drugs, which are relatively complex and time-consuming. Moreover, these methods can only indicate the drug effectiveness on cell colony rather than single cells. Thus, to meet the requirements of personal precision medicine, the development of drug response analysis on the high resolution of single cell is demanded. RESULTS To provide precise result for drug response on single-cell level, a microfluidic platform coupled with the label-free hyperspectral imaging was developed. With the help of horizontal single-cell trapping sieves, hundreds of single cells were trapped independently in microfluidic channels for the purposes of real-time drug delivery and single-cell hyperspectral image recording. To significantly identify the cellular hyperspectral change after drug stimulation, the differenced single-cell spectrum was proposed. Compared with the deep learning classification method based on hyperspectral images, an optimal performance can be achieved by the classification strategy based on differenced spectra. And the cellular response to different reagents, for example, K+, Epidermal Growth Factor (EGF), and Gefitinib at different concentrations can be accurately characterized by the differenced single-cell spectra analysis. SIGNIFICANCE AND NOVELTY The high-throughput, rapid analysis of cellular response to drugs at the single-cell level can be accurately performed by our platform. After systematically analyzing the materials and the structures of the single-cell microfluidic chip, the optimal single-cell trapping method was proposed to contribute to the further application of hyperspectral imaging on microfluidic single-cell analysis. And the hyperspectral characterization of single-cell with cancer drug stimulation proved the application potential of our method in personal cancer medication.
Collapse
Affiliation(s)
- Luyao Liu
- Institute of Microfluidic Chip Development in Biomedical Engineering, School of Information Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Lulu Zhang
- Institute of Microfluidic Chip Development in Biomedical Engineering, School of Information Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xueyu Zhang
- School of Information and Electronics, Beijing Institute of Technology, Beijing, 100081, China
| | - Xiaobin Dong
- Institute of Microfluidic Chip Development in Biomedical Engineering, School of Information Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xiaodan Jiang
- Institute of Microfluidic Chip Development in Biomedical Engineering, School of Information Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xiaoqi Huang
- School of Information and Electronics, Beijing Institute of Technology, Beijing, 100081, China
| | - Wei Li
- School of Information and Electronics, Beijing Institute of Technology, Beijing, 100081, China
| | - Xiaoming Xie
- School of Information Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xianbo Qiu
- Institute of Microfluidic Chip Development in Biomedical Engineering, School of Information Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China.
| |
Collapse
|
6
|
Zhang Y, Chen Q, Huang T, Zhu D, Lu Y. Bioinformatics-based screening of key genes for transformation of tyrosine kinase inhibitor-resistant lung adenocarcinoma to small cell lung cancer. Front Med (Lausanne) 2023; 10:1203461. [PMID: 37583423 PMCID: PMC10424445 DOI: 10.3389/fmed.2023.1203461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 07/17/2023] [Indexed: 08/17/2023] Open
Abstract
Purpose Lung adenocarcinoma (LUAD) is a common type of lung cancer. Cancer in a small number of patients with EGFR mutations will transform from LUAD to small cell lung cancer (SCLC) during epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) therapiesr. The purpose of the present study was to identify the core genes related to the transformation of LUAD into SCLC and to explore the associated molecular mechanisms. Methods GSE29016, GSE1037, GSE6044 and GSE40275 mRNA microarray datasets from Gene Expression Omnibus (GEO) were analyzed to obtain differentially expressed genes (DEGs) between LUAD and SCLC tissues, and the results were used for network analysis of protein-protein interactions (PPIs). After identifying the hub gene by STRING and Cytoscape platform, we explored the relationship between hub genes and the occurrence and development of SCLC. Finally, the obtained hub genes were validated in treated LUAD cells. Results A total of 41 DEGs were obtained, four hub genes (EZH2, NUSAP1, TTK and UBE2C) were identified, and related prognostic information was obtained. The coexpressed genes of the hub gene set were further screened, and the analysis identified many genes related to the cell cycle. Subsequently, LUAD cell models with TP53 and RB1 inactivation and overexpression of ASCL1 were constructed, and then the expression of hub genes was detected, the results showed that the four hub genes were all elevated in the established cell model. Conclusion EZH2, NUSAP1, TTK and UBE2C may affect the transformation of LUAD to SCLC and represent new candidate molecular markers for the occurrence and development of SCLC.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Oncology, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Qiang Chen
- Department of Oncology, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ting Huang
- Department of Clinical Pathology, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Di Zhu
- Department of Clinical Pathology, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yuanzhi Lu
- Department of Clinical Pathology, First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
7
|
Kashefizadeh A, Kazemizadeh H. Immunogenic cell death (ICD)-inducers in non-small-cell lung carcinoma (NSCLC): current knowledge and future perspective. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:316-322. [PMID: 36180811 DOI: 10.1007/s12094-022-02949-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 09/05/2022] [Indexed: 01/27/2023]
Abstract
The prevalence of non-small-cell lung cancer (NSCLC) is rising every year all around the world. The interaction between cancer cells and the tumor microenvironment (TME) is a crucial factor in determining the development of human neoplasms. Organellar and cellular stress are induced during immunogenic cell death (ICD), a particularly functional response pattern. ICD is a separate but poorly characterized entity caused by various cancer treatments. The induction of ICD has the potential to change TME and the recruitment of tumor-infiltrating lymphocytes (TILs), and the coupling of ICD-inducers and other therapeutic approaches can have a synergistic role in boosting anticancer impacts. The purpose of this study is to review the studies in the field of NSCLC using ICD-inducers as a treatment strategy or as a combination therapy. This review provide for researches a better view of what has been done so far and the challenges they face in the future.
Collapse
Affiliation(s)
- Alireza Kashefizadeh
- Department of Pulmonology, Shahid Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Kazemizadeh
- Advanced Thoracic Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Hu X, Li J, Chen Y, Long Q, Bai Y, Li R, Wang K, Jiang M, Chen C, Mao J, Zheng Y, Gao Z. A Self-Assembly ICG Nanoparticle Potentiating Targeted Photothermal and Photodynamic Therapy in NSCLC. ACS Biomater Sci Eng 2022; 8:4535-4546. [PMID: 36070516 DOI: 10.1021/acsbiomaterials.2c00620] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In nonsmall cell lung cancers (NSCLC), near-infrared (NIR) fluorescence imaging using indocyanine green (ICG) has proven to be an efficient approach for locating pulmonary nodules and pulmonary sentinel lymph nodes. However, due to a lack of tumor selectivity, ICG's use as a photosensitizer for photothermal therapy (PTT) and photodynamic therapy (PDT) is restricted. In the current study, we aimed to develop a type of high-performance NIR nanoparticle formulated with ICG to enhance its targeted efficacy and tumor specificity on NSCLC. An ICG-osimertinib nanoparticle (ICG-Osi) was self-assembled through π-π stacking, with a size of 276 nm and a surface charge of -7.4 mV. The NIR visibility and epidermal growth factor receptor (EGFR) targetability of the ICG-Osi was confirmed by its binding efficiency to EGFR-expressing NSCLC cells in vitro and in vivo, regardless of EGFR mutation status. The targeted effect was further confirmed in mouse xenograft models and showed an extended tumor retention time (>96 h). We demonstrated a significantly enhanced hyperthermia effect and a retained reactive oxygen species (ROS) generating ability of ICG-Osi, resulting in a 2-fold higher cell death rate than ICG alone. The ICG-Osi down-regulated GPX4 and p62 expression while up-regulating caspase-3 and beclin1 expression in NSCLC cells, indicating a complex network of cell death-related proteins. Considering the merits of simple assembly, EGFR binding efficacy, improved hyperthermia effect, and efficient cancer cell suppression, the ICG-Osi exhibits great potential for clinical application in EGFR-expressing NSCLC therapy.
Collapse
Affiliation(s)
- Xiaoyi Hu
- Department of Respiratory and Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China.,Institute of Chest and Lung Diseases, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
| | - Jiwei Li
- Department of Respiratory and Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China.,Institute of Chest and Lung Diseases, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
| | - Yulun Chen
- Department of Radiology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Qiuyue Long
- Department of Respiratory and Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China.,Institute of Chest and Lung Diseases, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
| | - Yangyuyan Bai
- Department of Respiratory and Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China.,Institute of Chest and Lung Diseases, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
| | - Ran Li
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing 100044, China
| | - Keqiang Wang
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing 100044, China
| | - Mingzheng Jiang
- Department of Respiratory and Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China.,Institute of Chest and Lung Diseases, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
| | - Chaoyang Chen
- Institute of Chest and Lung Diseases, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
| | - Jingsong Mao
- Department of Radiology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China.,Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen 361101, China
| | - Yali Zheng
- Department of Respiratory and Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China.,Institute of Chest and Lung Diseases, Xiang'an Hospital of Xiamen University, Xiamen 361101, China
| | - Zhancheng Gao
- Department of Respiratory and Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China.,Institute of Chest and Lung Diseases, Xiang'an Hospital of Xiamen University, Xiamen 361101, China.,Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
9
|
Occurrence of hyperprogressive disease following administration of immune checkpoint inhibitors in lung squamous cell carcinoma: A case report. Exp Ther Med 2022; 24:617. [PMID: 36160895 PMCID: PMC9468829 DOI: 10.3892/etm.2022.11554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/15/2022] [Indexed: 11/16/2022] Open
Abstract
Immunotherapy through blocking programmed cell death 1, programmed death-ligand 1 and cytotoxic T lymphocyte antigen 4 is developing rapidly and has gained increasing attention as a treatment for malignant tumors. However, some patients experience varying degrees of immune-related side effects after undergoing immunotherapy, with hyperprogressive disease (HPD) occurring in severe cases which increases the risk of mortality. The present study discussed the risk factors for HPD following immunotherapy in a case of lung squamous cell carcinoma, after treatment with a combination of anti-angiogenic drugs and biological cytotoxic drugs, the mass was found to have become smaller than before, along with follow-up treatment options, to provide a reference for clinical treatment decisions.
Collapse
|