1
|
Zhong W, Xu Y, Wang Z, Wang X, Li Y, Liu J, Zhao C, Shi X, He Z, Sun B, Tian C. Dual role of triglyceride structures facilitates anti-tumor drug delivery: Both as a self-assembling module and a responsive module. J Colloid Interface Sci 2025; 678:24-34. [PMID: 39277950 DOI: 10.1016/j.jcis.2024.09.096] [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: 04/22/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
Small molecule prodrugs self-assembled nano-delivery systems with tumor responsive linkages are emerging as an effective platform. However, the heterogeneity of tumor microenvironment may limit the anti-tumor effect of prodrug nanomedicines with a single response module. Here, we chose disulfide bond as the response module and branched chain alcohol as the self-assembly modification module to construct a single-responsive prodrug. We also constructed a double-responsive paclitaxel prodrug combining triglyceride and disulfide bond, taking into account of the highly expressed lipase and glutathione levels in tumor cells. The results showed that the anti-tumor effect of single-responsive branched chain alcohol modified prodrug nanoparticles was inferior to triglyceride prodrug nanoparticles with dual response modules. The triglyceride structure can not only serve as a self-assembly modification module, but also serve as a response module for intelligent drug release in tumor. Such dual roles will facilitate the efficient delivery of small molecule self-assembled prodrugs to tumor sites.
Collapse
Affiliation(s)
- Wenxin Zhong
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang, Liaoning 110016, China
| | - Yalin Xu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Zixuan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xiyan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yaqi Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Jinrui Liu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Can Zhao
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang, Liaoning 110016, China
| | - Bingjun Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang, Liaoning 110016, China.
| | - Chutong Tian
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang, Liaoning 110016, China.
| |
Collapse
|
2
|
Fang S, Cao H, Liu J, Cao G, Li T. Antitumor effects of IOX1 combined with bevacizumab-induced apoptosis and immunity on colorectal cancer cells. Int Immunopharmacol 2024; 141:112896. [PMID: 39146782 DOI: 10.1016/j.intimp.2024.112896] [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: 05/24/2024] [Revised: 07/11/2024] [Accepted: 08/05/2024] [Indexed: 08/17/2024]
Abstract
Colorectal cancer (CRC), as a fatal cancer, is one of the most common cancers worldwide. Although the standard treatment for colorectal cancer is well researched and established, long-term patient survival remains poor, and mortality remains high. Therefore, more and more effective treatment options are needed. To evaluate the efficacy of bevacizumab, the histone demethylase inhibitor IOX1, or their combination for the treatment of colorectal cancer, we examined the effects of IOX1, bevacizumab, and IOX1 combined with bevacizumab on cell activity, proliferation, and migration of colorectal cancer cell lines HCT116, RKO, and CT26 by CCK8, colony formation assay, wound healing assay, and transwell assay. The effects of the drugs alone as well as in combination on apoptosis in colorectal cancer cell lines were examined by flow cytometry and further validated by Western blotting for apoptosis-related proteins. The antitumor effects of treatment alone or in combination on colorectal cancer cells were examined in animal models. Mice were injected subcutaneously with CT26 cells and the growth and immune infiltration in tumor tissues were detected by IHC after drug treatment. We found that IOX1 could effectively inhibit the activity of CRC cells and had a significant inhibitory effect on the proliferation and migration of CRC cells. The apoptosis rate increased in a dose-dependent manner after IOX1 treatment on colorectal cancer cells, and the expression of apoptosis-related proteins changed accordingly. Further combination with bevacizumab revealed that the combination had a more significant effect on the proliferation, migration, and apoptosis of CRC cells than either IOX1 or bevacizumab alone. In vivo experiments have found that both alone and combination drugs can inhibit the growth of mouse tumors, but the effect of combination inhibition is the most obvious. Combination therapy significantly inhibited the expression of proliferative marker (Ki67) in tumor xenograft models, and increased content of antigen-specific CD4+, CD8+T cell growth, and granzymeB (GZMB), which is associated with T cell cytotoxicity, was detected in combination therapy. Immunoassays suppressed the expression of relevant PD-1 and decreased. The anticancer drug bevacizumab and the histone demethylase inhibitor IOX1 may inhibit colon cancer cell growth by regulating apoptosis. The inhibitory effect of combination therapy on tumor growth may be achieved, in part, through upregulation of infiltration-mediated tumor immunity by T lymphocytes. The combination of IOX1 and bevacizumab produced significant synergistic effects. This study aims to provide a new direction for CRC combination therapy.
Collapse
Affiliation(s)
- Shuilong Fang
- Zhengzhou University People's Hospital, Zhengzhou, Henan, China; Department of Comprehensive Intervention, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Huicun Cao
- Department of Comprehensive Intervention, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Jian Liu
- Department of Comprehensive Intervention, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Guangshao Cao
- Department of Comprehensive Intervention, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Tianxiao Li
- Zhengzhou University People's Hospital, Zhengzhou, Henan, China; Interventional Center, Henan Provincial People's Hospital, Zhengzhou, Henan, China.
| |
Collapse
|
3
|
Chen D, Gu X, Nurzat Y, Xu L, Li X, Wu L, Jiao H, Gao P, Zhu X, Yan D, Li S, Xue C. Writers, readers, and erasers RNA modifications and drug resistance in cancer. Mol Cancer 2024; 23:178. [PMID: 39215288 PMCID: PMC11363509 DOI: 10.1186/s12943-024-02089-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Drug resistance in cancer cells significantly diminishes treatment efficacy, leading to recurrence and metastasis. A critical factor contributing to this resistance is the epigenetic alteration of gene expression via RNA modifications, such as N6-methyladenosine (m6A), N1-methyladenosine (m1A), 5-methylcytosine (m5C), 7-methylguanosine (m7G), pseudouridine (Ψ), and adenosine-to-inosine (A-to-I) editing. These modifications are pivotal in regulating RNA splicing, translation, transport, degradation, and stability. Governed by "writers," "readers," and "erasers," RNA modifications impact numerous biological processes and cancer progression, including cell proliferation, stemness, autophagy, invasion, and apoptosis. Aberrant RNA modifications can lead to drug resistance and adverse outcomes in various cancers. Thus, targeting RNA modification regulators offers a promising strategy for overcoming drug resistance and enhancing treatment efficacy. This review consolidates recent research on the role of prevalent RNA modifications in cancer drug resistance, with a focus on m6A, m1A, m5C, m7G, Ψ, and A-to-I editing. Additionally, it examines the regulatory mechanisms of RNA modifications linked to drug resistance in cancer and underscores the existing limitations in this field.
Collapse
Affiliation(s)
- Di Chen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Yeltai Nurzat
- State Key Laboratory of Respiratory Disease, Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lixia Xu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Xueyuan Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Lixin Wu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Henan Jiao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Peng Gao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Xuqiang Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Dongming Yan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Shaohua Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Chen Xue
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
4
|
Fang Z, Bai J. Integrated bioinformatics analysis reveals the bidirectional effects of TSPAN6 for cisplatin resistance in lung cancer. Chem Biol Drug Des 2024; 103:e14570. [PMID: 38887156 DOI: 10.1111/cbdd.14570] [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: 04/05/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 06/20/2024]
Abstract
Cisplatin-based chemotherapy is frequently employed as the primary therapeutic approach for advanced lung cancer. Nevertheless, a significant proportion of patients may develop resistance to cisplatin, leading to diminished efficacy of chemotherapy. Through analysis of Gene Expression Omnibus databases, TSPAN6 has been identified as a key factor in conferring resistance to cisplatin, attributed to its activation of the NF-κB signaling pathway. Knockdown of TSPAN6 using siRNA resulted in decreased expression levels of NF-κB in A549 cells. This indicates that TSPAN6 may have dual effects on lung cancer cisplatin resistance and could serve as a promising therapeutic target for individuals with cisplatin resistance.
Collapse
Affiliation(s)
- Zhihong Fang
- Department of General Surgery, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, Jiangsu, China
| | - Jinmei Bai
- Department of Respiratory, Affiliated Wuxi Fifth Hospital of Jiangnan University (The Fifth People's Hospital of Wuxi), Wuxi, Jiangsu, China
| |
Collapse
|
5
|
Wang Y, Yu B, Qu M, Liu F, Wu X. Britannin inhibits cell proliferation, migration and glycolysis by downregulating KLF5 in lung cancer. Exp Ther Med 2024; 27:109. [PMID: 38361511 PMCID: PMC10867720 DOI: 10.3892/etm.2024.12397] [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: 06/30/2023] [Accepted: 09/12/2023] [Indexed: 02/17/2024] Open
Abstract
Lung cancer is a harmful type of malignancy and the leading cause of cancer-associated mortality. It is therefore imperative to develop novel drugs effective for treating this cancer. The Traditional Chinese Medicine compound Britannin has been previously reported to inhibit the development of certain cancers, such as pancreatic, breast and liver cancer. Moreover, Kruppel-like factor 5 (KLF5) has been identified an on oncogene in lung cancer. In the present study, the possible regulatory effects and underlying mechanism of Britannin in lung cancer were investigated. A549 and 16HBE cells were treated with different concentrations of Britannin. Subsequently, Cell counting kit-8, EdU staining and colony formation assays were used to detect the proliferative ability of these cells. Cell migration was detected by wound healing and Transwell assays, respectively. XF96 extracellular flux analyzer was used to analyze the extent of extracellular acidification and oxygen consumption rate in cells, whereas assay kits were used to detect glucose and lactic acid levels in the cell supernatant. The targeting effect between Britannin and the KLF5 protein was investigated using molecular docking technology. The protein expression levels of KLF5 in cells challenged with Britannin was detected by western blotting. Finally, overexpression of KLF5 in A549 cells was performed before cell proliferation, migration and the glycolysis rate were measured to explore the regulatory effects of Britannin. Britannin was found to inhibit the proliferation, migration and glycolysis of lung cancer cells, during which the protein expression levels of KLF5 were decreased. This suggests that Britannin regulated the expression of KLF5 in A549 cells. Overexpression of KLF5 reversed the inhibitory effects of Britannin on the proliferation, migration and glycolysis in lung cancer cells. In conclusion, these results suggest that Britannin can inhibit cell proliferation, migration and glycolysis by downregulating KLF5 expression in lung cancer cells.
Collapse
Affiliation(s)
- Ying Wang
- Department of Nosocomial Infection, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, Shandong 266042, P.R. China
| | - Botao Yu
- Department of Emergency Medicine, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, Shandong 266042, P.R. China
| | - Mengyuan Qu
- Department of Radiophysics, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, Shandong 266042, P.R. China
| | - Fengjuan Liu
- Ward for Phase I Clinical Trial, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, Shandong 266042, P.R. China
| | - Xiao Wu
- Department of Pulmonary and Critical Care Medicine, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, Shandong 266042, P.R. China
| |
Collapse
|
6
|
Faeq MH, Al-Haideri M, Mohammad TAM, Gharebakhshi F, Marofi F, Tahmasebi S, Modaresahmadi S. CAR-modified immune cells as a rapidly evolving approach in the context of cancer immunotherapies. Med Oncol 2023; 40:155. [PMID: 37083979 PMCID: PMC10119530 DOI: 10.1007/s12032-023-02019-4] [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: 03/06/2023] [Accepted: 03/28/2023] [Indexed: 04/22/2023]
Abstract
Nowadays, one of the main challenges clinicians face is malignancies. Through the progression of technology in recent years, tumor nature and tumor microenvironment (TME) can be better understood. Because of immune system involvement in tumorigenesis and immune cell dysfunction in the tumor microenvironment, clinicians encounter significant challenges in patient treatment and normal function recovery. The tumor microenvironment can stop the development of tumor antigen-specific helper and cytotoxic T cells in the tumor invasion process. Tumors stimulate the production of proinflammatory and immunosuppressive factors and cells that inhibit immune responses. Despite the more successful outcomes, the current cancer therapeutic approaches, including surgery, chemotherapy, and radiotherapy, have not been effective enough for tumor eradication. Hence, developing new treatment strategies such as monoclonal antibodies, adaptive cell therapies, cancer vaccines, checkpoint inhibitors, and cytokines helps improve cancer treatment. Among adoptive cell therapies, the interaction between the immune system and malignancies and using molecular biology led to the development of chimeric antigen receptor (CAR) T cell therapy. CAR-modified immune cells are one of the modern cancer therapeutic methods with encouraging outcomes in most hematological and solid cancers. The current study aimed to discuss the structure, formation, subtypes, and application of CAR immune cells in hematologic malignancies and solid tumors.
Collapse
Affiliation(s)
- Mohammed Hikmat Faeq
- Student of General Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maysoon Al-Haideri
- Department of Physiotherapy, Cihan University-Erbil, Kurdistan Region, Erbil, Iraq
| | - Talar Ahmad Merza Mohammad
- Department of Pharmacology, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
| | - Farshad Gharebakhshi
- Department of Radiology, School of Medicine, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Safa Tahmasebi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Shadan Modaresahmadi
- Department of Immunology and Biotechnology, Texas Tech University Health Siences Center, Abilene, TX, USA
| |
Collapse
|
7
|
Wang G, Zhao M, Li J, Li G, Zheng F, Xu G, Hong X. m7G-Associated subtypes, tumor microenvironment, and validation of prognostic signature in lung adenocarcinoma. Front Genet 2022; 13:954840. [PMID: 36046251 PMCID: PMC9422053 DOI: 10.3389/fgene.2022.954840] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/27/2022] [Indexed: 12/14/2022] Open
Abstract
Background: 7-Methylguanosine (m7G) is an important posttranscriptional modification that regulates gene expression and is involved in tumorigenesis and development. Tumor microenvironment has been proven to be highly involved in tumor progression and prognosis. However, how m7G-associated genes affect the tumor microenvironment of patients with lung adenocarcinoma (LUAD) remains to be further clarified. Methods: The genetic alterations of m7G-associated genes and their associations with the prognosis and tumor microenvironment in LUAD patients were systemically analyzed. An m7G-Riskscore was established and analyzed for its performance in disease prognosis and association with patient response to immunotherapy. Expression of the model genes at the protein level was investigated through ex vivo experiments. A nomogram was finally obtained based on the m7G-Riskscore and several significant clinical pathological features. Results: m7G-Associated genes were obtained from five LUAD datasets from The Cancer Genome Atlas and Gene Expression Omnibus databases, and their expression pattern was determined. Based on the m7G-associated genes, three LUAD clusters were defined. The differentially expressed genes from the three clusters were screened and used to further divide the LUAD patients into two gene clusters. It was demonstrated that the alterations of m7G-associated genes were associated with the clinical pathological features, prognosis, and tumor immune infiltration in LUAD patients. An m7G-Riskscore including CAND1, RRM2, and SLC2A1 was obtained with robust and accurate prognostic performance. WB and cell immunofluorescence also showed significant dysregulation of CAND1, RRM2, and SLC2A1 in LUAD. In addition, a nomogram was established to improve the clinical feasibility of the m7G-Riskscore. Correlation analysis revealed that patients with a lower m7G-Riskscore had higher immune and stromal scores, responded well to chemotherapeutics and multiple targeted drugs, and survived longer. Patients with a higher m7G-Riskscore tended to suffer from a higher tumor mutation burden. Furthermore, the m7G-Riskscore exhibited significant associations with immune cell infiltration and cancer stemness. Conclusion: This study systemically analyzed m7G-associated genes and identified their potential role in tumor microenvironment and prognosis in patients with LUAD. The findings of the present study may help better understand LUAD from the m7G perspective and also provide a new thought toward the prognosis and treatment of LUAD.
Collapse
Affiliation(s)
- Guangyao Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Mei Zhao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Jiao Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Guosheng Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Fukui Zheng
- Department of Intensive Care Unit, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Guanglan Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
- *Correspondence: Guanglan Xu, ; Xiaohua Hong,
| | - Xiaohua Hong
- Graduate School, Guangxi University of Chinese Medicine, Nanning, China
- *Correspondence: Guanglan Xu, ; Xiaohua Hong,
| |
Collapse
|
8
|
Xiao BF, Zhang JT, Zhu YG, Cui XR, Lu ZM, Yu BT, Wu N. Chimeric Antigen Receptor T-Cell Therapy in Lung Cancer: Potential and Challenges. Front Immunol 2021; 12:782775. [PMID: 34790207 PMCID: PMC8591168 DOI: 10.3389/fimmu.2021.782775] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/13/2021] [Indexed: 12/21/2022] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has exhibited a substantial clinical response in hematological malignancies, including B-cell leukemia, lymphoma, and multiple myeloma. Therefore, the feasibility of using CAR-T cells to treat solid tumors is actively evaluated. Currently, multiple basic research projects and clinical trials are being conducted to treat lung cancer with CAR-T cell therapy. Although numerous advances in CAR-T cell therapy have been made in hematological tumors, the technology still entails considerable challenges in treating lung cancer, such as on−target, of−tumor toxicity, paucity of tumor-specific antigen targets, T cell exhaustion in the tumor microenvironment, and low infiltration level of immune cells into solid tumor niches, which are even more complicated than their application in hematological tumors. Thus, progress in the scientific understanding of tumor immunology and improvements in the manufacture of cell products are advancing the clinical translation of these important cellular immunotherapies. This review focused on the latest research progress of CAR-T cell therapy in lung cancer treatment and for the first time, demonstrated the underlying challenges and future engineering strategies for the clinical application of CAR-T cell therapy against lung cancer.
Collapse
Affiliation(s)
- Bu-Fan Xiao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jing-Tao Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yu-Ge Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xin-Run Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhe-Ming Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ben-Tong Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Nan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
9
|
Bora-Singhal N, Mohankumar D, Saha B, Colin CM, Lee JY, Martin MW, Zheng X, Coppola D, Chellappan S. Novel HDAC11 inhibitors suppress lung adenocarcinoma stem cell self-renewal and overcome drug resistance by suppressing Sox2. Sci Rep 2020; 10:4722. [PMID: 32170113 PMCID: PMC7069992 DOI: 10.1038/s41598-020-61295-6] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 02/20/2020] [Indexed: 01/06/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is known to have poor patient outcomes due to development of resistance to chemotherapy agents and the EGFR inhibitors, which results in recurrence of highly aggressive lung tumors. Even with recent success in immunotherapy using the checkpoint inhibitors, additional investigations are essential to identify novel therapeutic strategies for efficacious treatment for NSCLC. Our finding that high levels of histone deacetylase 11 (HDAC11) in human lung tumor tissues correlate with poor patient outcome and that depletion or inhibition of HDAC11 not only significantly reduces self-renewal of cancer stem cells (CSCs) from NSCLC but also decreases Sox2 expression that is essential for maintenance of CSCs, indicates that HDAC11 is a potential target to combat NSCLC. We find that HDAC11 suppresses Sox2 expression through the mediation of Gli1, the Hedgehog pathway transcription factor. In addition, we have used highly selective HDAC11 inhibitors that not only target stemness and adherence independent growth of lung cancer cells but these inhibitors could also efficiently ablate the growth of drug-insensitive stem-like cells as well as therapy resistant lung cancer cells. These inhibitors were found to be efficacious even in presence of cancer associated fibroblasts which have been shown to contribute in therapy resistance. Our study presents a novel role of HDAC11 in lung adenocarcinoma progression and the potential use of highly selective inhibitors of HDAC11 in combating lung cancers.
Collapse
Affiliation(s)
- Namrata Bora-Singhal
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Durairaj Mohankumar
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Biswarup Saha
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Christelle M Colin
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Jennifer Y Lee
- FORMA Therapeutics, 500 Arsenal St, Suite 100, Watertown, MA, 02472, USA
| | - Matthew W Martin
- FORMA Therapeutics, 500 Arsenal St, Suite 100, Watertown, MA, 02472, USA
| | - Xiaozhang Zheng
- FORMA Therapeutics, 500 Arsenal St, Suite 100, Watertown, MA, 02472, USA
| | - Domenico Coppola
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Srikumar Chellappan
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA.
| |
Collapse
|
10
|
Omballi M, Fernandez-Bussy S, Patel PP, Jantz MA, Becnel D, Patel NM, Mehta HJ. Surveillance Imaging After Curative Intent Therapy for Lung Cancer. Semin Roentgenol 2019; 55:60-69. [PMID: 31964482 DOI: 10.1053/j.ro.2019.10.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Mohamed Omballi
- Division of Pulmonary and Critical Care Medicine, University of Florida, Gainesville, FL
| | | | - Priya P Patel
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN
| | - Michael A Jantz
- Division of Pulmonary and Critical Care Medicine, University of Florida, Gainesville, FL
| | - David Becnel
- Division of Pulmonary and Critical Care Medicine, University of Florida, Gainesville, FL
| | - Neal M Patel
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Jacksonville, FL
| | - Hiren J Mehta
- Division of Pulmonary and Critical Care Medicine, University of Florida, Gainesville, FL.
| |
Collapse
|
11
|
Li H, Ji Y, Zhang S, Gao Z, Hu C, Jiang R, Chen M, Li G, Zhang X. Kangai Injection Combined with Platinum-based Chemotherapy for the Treatment of Stage III/IV Non-Small Cell Lung Cancer: A Meta-analysis and Systematic Review of 35 Randomized Controlled Trials. J Cancer 2019; 10:5283-5298. [PMID: 31602279 PMCID: PMC6775612 DOI: 10.7150/jca.31928] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 06/25/2019] [Indexed: 12/15/2022] Open
Abstract
Objective: In an effort to inform evidence-based guidelines for clinical practice, we performed a meta-analysis to systematically evaluate the safety and efficacy of Kangai injection (KAI) plus platinum-based chemotherapy for stage III/IV non-small cell lung cancer (NSCLC). Methods: Randomized controlled trials (RCTs) comparing KAI plus platinum-based chemotherapy (experimental group) to chemotherapy alone (control group) were electronically retrieved from the Cochrane Library, PubMed, EMbase, Web of Science, Chinese National Knowledge Infrastructure (CNKI), Chinese Biological Medicine (CBM) Database, Wanfang Database, and the VIP Database for Chinese Technical Periodicals. RCTs published from the date of inception to July 5, 2018, were included. All trials were assessed for methodological quality in accordance with the Cochrane Reviewer's Handbook for Systematic Reviews of Intervention. Meta-analysis was performed using RevMan5.3 Software and Comprehensive Meta-Analysis (CMA) 2.0. Results: The final analysis included 35 RCTs involving 2,618 patients. Our meta-analysis revealed that KAI combined with platinum-based chemotherapy was associated with significantly greater objective response rate (ORR) (RR=1.36, 95% CI: 1.25-1.49, P<0.00001) and disease control rate (DCR) (RR=1.14, 95% CI: 1.09-1.18, P<0.00001), improvements in quality of life (QOL) (RR=1.75, 95% CI: 1.59-1.93, P<0.00001), and decreases in the incidence of gastrointestinal reactions (RR=0.64, 95% CI: 0.54-0.77, P<0.00001), leukocytopenia (RR=0.54, 95% CI: 0.46-0.63, P<0.00001) and thrombocytopenia (RR=0.52, 95% CI: 0.36-0.76, P=0.0007) when compared with chemotherapy alone. In addition, combined treatment was associated with greater regulation of tumor immune function, as indicated by increases in the proportion of NK, CD3+ , and CD4+ cells (MD=2.27, 95% CI: 1.18-3.36, P<0.0001; MD=12.86, 95% CI: 11.64-14.08, P<0.00001; and MD=5.48, 95% CI: 2.68-8.28, P=0.0001) and decreases in the percentage of CD8+ cells (MD= -2.37, 95% CI from -4.51 to -0.23, P=0.03). Conclusions: From the available evidence, our results indicate that KAI plus platinum-based chemotherapy could be more effective in improving clinical efficacy, decreasing the incidence of adverse reactions and regulating the tumor immune function than chemotherapy alone in the treatment of stage III/IV NSCLC. Nevertheless, considering the limitations of the included studies, rigorous designed, high-quality, multicenter clinical trials are still need to further confirm the results.
Collapse
Affiliation(s)
- Hongxiao Li
- School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing 210023, China.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuejin Ji
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Shiping Zhang
- School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing 210023, China.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zishan Gao
- Clinical Acupuncture and Moxibustion Department, Second School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Cheng Hu
- School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing 210023, China.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Rilei Jiang
- School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing 210023, China.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Meijuan Chen
- School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing 210023, China.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Guochun Li
- School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing 210023, China.,Department of epidemiology and biostatistics, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xu Zhang
- School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing 210023, China.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
12
|
A phase II study of carboplatin, pemetrexed, and bevacizumab followed by erlotinib and bevacizumab maintenance for non-squamous non-small cell lung cancer with wild-type EGFR (HOT1101). Int J Clin Oncol 2018; 23:1060-1069. [PMID: 30027464 DOI: 10.1007/s10147-018-1318-z] [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: 02/09/2018] [Accepted: 07/16/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND This study evaluated the efficacy and safety of switch maintenance erlotinib and bevacizumab after induction therapy with carboplatin/pemetrexed/bevacizumab for non-squamous non-small cell lung cancer (NSCLC) with wild-type EGFR. METHODS Enrolled patients had treatment-naïve, advanced non-squamous NSCLC with wild-type EGFR. Carboplatin [area under the curve (AUC) 5.0], pemetrexed (500 mg/m2) and bevacizumab (15 mg/kg) were administered on day 1 every 3 weeks for 4-6 cycles. Maintenance therapy with erlotinib (150 mg/body) on day 1 through 21 plus bevacizumab on day 1 every 3 weeks was continued until disease progression or unacceptable toxicity. The primary endpoint was 6-month progression-free survival (PFS); secondary endpoints included overall survival (OS), overall response rate (ORR), toxicity, and quality of life (QOL). RESULTS Fifty-one patients were enrolled between September 2011 and June 2014. The median number of cycles for induction and maintenance therapy was 4 (range 1-6) and 4 (range 1-20). Twenty-nine patients (58%) received maintenance therapy. The 6-month PFS rate was 59.5% [95% confidence interval (CI) 45.0-72.6%]. The ORR was 48.0% (95% CI 34.8-61.5%), and disease control rate was 86.0% (95% CI 73.8-93.0%). The median PFS and OS were 6.5 months (95% CI 5.8-7.2 months) and 21.4 months (95% CI 15.9-26.9 months), respectively. Although grades ≥ 3 adverse events were observed in 33 patients (66.0%), most were hematologic; there was no febrile neutropenia. QOL was maintained throughout treatment. CONCLUSIONS Carboplatin/pemetrexed/bevacizumab followed by erlotinib and bevacizumab maintenance showed modest efficacy and was well tolerated in non-squamous NSCLC patients with wild-type EGFR. TRIAL REGISTRATION UMIN000005872.
Collapse
|
13
|
Wang Y, Wang H, Jiang Y, Zhang Y, Wang X. A randomized phase III study of combining erlotinib with bevacizumab and panitumumab versus erlotinib alone as second-line therapy for Chinese patients with non-small-cell lung cancer. Biomed Pharmacother 2017; 89:875-879. [PMID: 28282789 DOI: 10.1016/j.biopha.2017.02.097] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 02/25/2017] [Accepted: 02/26/2017] [Indexed: 01/22/2023] Open
Abstract
PURPOSE In this phase III clinical study, we assessed the clinical outcomes of combining erlotinib with bevacizumab and panitumumab as second-line chemotherapy for patients with non-small-cell lung cancer (NSCLC). METHODS Chinese NSCLC patients, who received first-line platinum-based chemotherapy but still experienced disease progression, were assigned to receive second-line treatment of erlotinib plus bevacizumab and panitumumab (arm I), or erlotinib plus placebo (arm II). The primary endpoint was progression-free survival (PFS). The secondary endpoints were overall survival (OS) and response rates. RESULTS 150 patients were enrolled in arm I, and 147 in arm II. Median PFS of arm I was 4.6 months (95% CI, 2.3-9.4 months), much longer than the median PFS in arm II (1.9 months, 95% CI 0.8-5.2 months) (P=0.003). The median OS of arm I was 10.4 months (95% CI, 7.5-13.1 months), also significantly longer than the median OS in arm II (8.9 months, 95% CI 3.3-10.9 months) (P=0.031). Partial response in arm I was 38%, significantly higher than the partial response rate of 15% in arm II (P=0.014). The occurrence rates of adverse events, including diarrhea, fatigue and rash, were higher in arm I than in arm II. CONCLUSIONS Erlotinib plus bevacizumab and panitumumab is an efficient second-line treatment option for patients with NSCLC.
Collapse
Affiliation(s)
- Ying Wang
- Department of Integrative Oncology, Cancer Center and Department of Clinical Pharmacy, Xiaoshan Hospital, Hangzhou, 311202, Zhejiang Province, China
| | - Hui Wang
- Department of Integrative Oncology, Cancer Center and Department of Clinical Pharmacy, Xiaoshan Hospital, Hangzhou, 311202, Zhejiang Province, China
| | - Yiling Jiang
- Department of Integrative Oncology, Cancer Center and Department of Clinical Pharmacy, Xiaoshan Hospital, Hangzhou, 311202, Zhejiang Province, China
| | - Yaping Zhang
- Department of Integrative Oncology, Cancer Center and Department of Clinical Pharmacy, Xiaoshan Hospital, Hangzhou, 311202, Zhejiang Province, China
| | - Xiaoyan Wang
- Department of Integrative Oncology, Cancer Center and Department of Clinical Pharmacy, Xiaoshan Hospital, Hangzhou, 311202, Zhejiang Province, China.
| |
Collapse
|
14
|
Franks SE, Jones RA, Briah R, Murray P, Moorehead RA. BMS-754807 is cytotoxic to non-small cell lung cancer cells and enhances the effects of platinum chemotherapeutics in the human lung cancer cell line A549. BMC Res Notes 2016; 9:134. [PMID: 26928578 PMCID: PMC4772483 DOI: 10.1186/s13104-016-1919-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/08/2016] [Indexed: 12/11/2022] Open
Abstract
Background Despite advances in targeted therapy for lung cancer, survival for patients remains poor and lung cancer remains the leading cause of cancer-related deaths worldwide. The type I insulin-like growth factor receptor (IGF-IR) has emerged as a potential target for lung cancer treatment, however, clinical trials to date have provided disappointing results. Further research is needed to identify if certain patients would benefit from IGF-IR targeted therapies and the ideal approach to incorporate IGF-IR targeted agents with current therapies. Methods The dual IGF-IR/insulin receptor inhibitor, BMS-754807, was evaluated alone and in combination with platinum-based chemotherapeutics in two human non-small cell lung cancer (NSCLC) cell lines. Cell survival was determined using WST-1 assays and drug interaction was evaluated using Calcusyn software. Proliferation and apoptosis were determined using immunofluorescence for phospho-histone H3 and cleaved caspase 3, respectively. Results Treatment with BMS-754807 alone reduced cell survival and wound closure while enhancing apoptosis in both human lung cancer cell lines. These effects appear to be mediated through IGF-IR/IR signaling and, at least in part, through the PI3K/AKT pathway as administration of BMS-754807 to A549 or NCI-H358 cells significantly suppressed IGF-IR/IR and AKT phosphorylation. In addition of BMS-754807 enhanced the cytotoxic effects of carboplatin or cisplatin in a synergistic manner when given simultaneously to A549 cells. Conclusions BMS-754807 may be an effective therapeutic agent for the treatment of NSCLC, particularly in lung cancer cells expressing high levels of IGF-IR.
Collapse
Affiliation(s)
- S Elizabeth Franks
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G2W1, Canada.
| | - Robert A Jones
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G2W1, Canada.
| | - Ritesh Briah
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G2W1, Canada.
| | - Payton Murray
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G2W1, Canada.
| | - Roger A Moorehead
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G2W1, Canada.
| |
Collapse
|
15
|
Zhang Y, Gao C, Qu W, Gao Y, Zhu S, Zhang S, He W, Yu Y. A randomized phase II study of erlotinib plus nab-paclitaxel versus erlotinib alone as second-line therapy for Chinese patients with advanced EGFR wild-type non-small-cell lung cancer. Cancer Invest 2015; 33:241-5. [PMID: 25951232 DOI: 10.3109/07357907.2015.1024318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Erlotinib is a standard second-line therapy for patients with advanced non-small-cell lung cancer (NSCLC). However, its efficacy for those patients with epidermal growth factor receptor (EGFR) wild-type (WT) tumors is undecided. In this randomized phase II study, NSCLC patients with EGFR-WT tumors, who had been treated with platinum-based chemotherapy but still developed disease progression, were assigned to receive second-line treatment of erlotinib plus nab-paclitaxel or erlotinib alone. We found PFS and OS were significantly improved by erlotinib plus nab-paclitaxel. The adverse events were also well tolerable.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Radiation Oncology, Shandong Tumor Hospital and Institute , Jinan , China
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Qiao H, Huang X, Guo H, Liu Y, Yue C. ERCC1, RRM1 and TUBB3 mRNA expression on the tumor response and overall survival of non-small cell lung cancer treated with platinum-based chemotherapy. Pak J Med Sci 2015; 30:1403-8. [PMID: 25674147 PMCID: PMC4320739 DOI: 10.12669/pjms.306.5768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/02/2014] [Accepted: 08/04/2014] [Indexed: 11/15/2022] Open
Abstract
Objective: We aimed to analyze the expression of ERCC1, RRM1 and TUBB3 in 305 patients with advanced non-small cell lung cancer (NSCLC) and investigate whether these genes can be used as biomarkers for predicting tumor response and clinical outcome. Methods: Total 305 patients with unresectable and locally advanced NSCLC were collected between January 2007 and December 2008. cDNA of ERCC1, RRM1 and TUBB3 was isolated by a fluorescence-based real-time detection method. Results: All the patients were followed up until December 2012. One hundred seventy five patients showed good response and 130 patients showed poor response to chemotherapy. 126 patients died and 166 patients showed progressive disease during the follow-up period. The median levels of ERCC1, RRM1 and TUBB3 mRNA were 0.53±0.13, 0.31±0.15 and 0.18±0.16, respectively. We found that patients with low ERCC1 expression showed a significantly higher rate of good tumor response, and the adjusted OR (95% CI) was 2.16(1.32-3.45). By Cox regression analysis. We also found that low ERCC1 expression level were correlated with longer overall survival of NSCLC patients, with the adjusted HR (95% CI) was 2.15 (1.26–3.35). Conclusion: This study showed that ERCC1 mRNA expression can not affect the response to chemotherapy and clinical outcome of advanced non-small cell lung cancer (NSCLC) patients.
Collapse
Affiliation(s)
- Hongwei Qiao
- Hongwei Qiao, The Second Respiratory Medicine Department, Xinxiang Central Hospital, Xinxiang, China
| | - Xiaoping Huang
- Xiaoping Huang, The Second Respiratory Medicine Department, Xinxiang Central Hospital, Xinxiang, China
| | - Hua Guo
- Hua Guo, The Second Oncology Department, Xinxiang Central Hospital, Xinxiang, China
| | - Yan Liu
- Yan Liu, Department of Tumor Surgery, Xinxiang Central Hospital, Xinxiang, China
| | - Chunyan Yue
- Chunyan Yue, The Second Oncology Department, Xinxiang Central Hospital, Xinxiang, China
| |
Collapse
|