1
|
Mehra A, Sangwan R. A Promising Paradigm Shift in Cancer Treatment with FGFR Inhibitors. Anticancer Agents Med Chem 2025; 25:2-23. [PMID: 39192641 DOI: 10.2174/0118715206318833240819031953] [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: 04/18/2024] [Revised: 07/04/2024] [Accepted: 07/12/2024] [Indexed: 08/29/2024]
Abstract
FGFR have been demonstrated to perform a crucial role in biological processes but their overexpression has been perceived as the operator component in the occurrence and progression of different types of carcinoma. Out of all the interest around cancer, FGFR inhibitors have assembled pace over the past few years. Therefore, FGFR inhibitors are one of the main fundamental tools to reverse drug resistance, tumor growth, and angiogenesis. Currently, many FGFR inhibitors are under the development stage or have been developed. Due to great demand and hotspots, different pharmacophores were approached to access structurally diverse FGFR inhibitors. Here, we have selected to present several representative examples such as Naphthyl, Pyrimidine, Pyridazine, Indole, and Quinoline derivatives that illustrate the diversity and advances of FGFR inhibitors in medicinal chemistry. This review focuses on the SAR study of FGFR inhibitors last five years which will be a great future scope that influences the medicinal chemist to work towards more achievements in this area.
Collapse
Affiliation(s)
- Anuradha Mehra
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road (NH-1), Phagwara (Punjab), 144411, India
| | - Rekha Sangwan
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road (NH-1), Phagwara (Punjab), 144411, India
| |
Collapse
|
2
|
Yan F, Teng Y, Li X, Zhong Y, Li C, Yan F, He X. Hypoxia promotes non-small cell lung cancer cell stemness, migration, and invasion via promoting glycolysis by lactylation of SOX9. Cancer Biol Ther 2024; 25:2304161. [PMID: 38226837 PMCID: PMC10793688 DOI: 10.1080/15384047.2024.2304161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 01/08/2024] [Indexed: 01/17/2024] Open
Abstract
BACKGROUND Lung cancer is the deadliest form of malignancy and the most common subtype is non-small cell lung cancer (NSCLC). Hypoxia is a typical feature of solid tumor microenvironment. In the current study, we clarified the effects of hypoxia on stemness and metastasis and the molecular mechanism. METHODS The biological functions were assessed using the sphere formation assay, Transwell assay, and XF96 extracellular flux analyzer. The protein levels were detected by western blot. The lactylation modification was assessed by western blot and immunoprecipitation. The role of SOX9 in vivo was explored using a xenografted tumor model. RESULTS We observed that hypoxia promoted sphere formation, migration, invasion, glucose consumption, lactate production, glycolysis, and global lactylation. Inhibition of glycolysis suppressed cell stemness, migration, invasion, and lactylation. Moreover, hypoxia increased the levels of SOX9 and lactylation of SOX9, whereas inhibition of glycolysis reversed the increase. Additionally, knockdown of SOX9 abrogated the promotion of cell stemness, migration, and invasion. In tumor-bearing mice, overexpression of SOX9 promoted tumor growth, and inhibition of glycolysis suppressed tumor growth. CONCLUSION Hypoxia induced the lactylation of SOX9 to promote stemness, migration, and invasion via promoting glycolysis. The findings suggested that targeting hypoxia may be an effective way for NSCLC treatment and reveal a new mechanism of hypoxia in NSCLC.
Collapse
Affiliation(s)
- Fei Yan
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Yue Teng
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Xiaoyou Li
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Yuejiao Zhong
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Chunyi Li
- Department of Medical Oncology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Feng Yan
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Xia He
- Department of Radiotherapy, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| |
Collapse
|
3
|
Zhang Q, Liang XY, Wang ZS, Sun A, Cao TB, Zhang YP, Li N, Yi TY, Qu KP. Efficacy of immune checkpoint inhibitors for NSCLC in patients with different age: A systematic review and meta-analysis. Asian J Surg 2024; 47:4691-4698. [PMID: 38641539 DOI: 10.1016/j.asjsur.2024.03.145] [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: 11/29/2023] [Revised: 03/11/2024] [Accepted: 03/22/2024] [Indexed: 04/21/2024] Open
Abstract
OBJECTIVE This article is a Meta-analysis aiming to systematically evaluate the difference in efficacy of immune checkpoint inhibitor in patients with non-small cell lung cancer (NSCLC) by age. METHODS We performed a Meta-analysis of published randomized controlled trials concerning for patients with NSCLC by age. We compared overall survival among three groups (age <65 years, age 65-75 years, age ≥75 years). Hazard ratios (HRs) and 95% confidence intervals (CIs) were collected and pooled. RESULTS A total of 10,291 patients from 17 RCTs were included. In the group under age 65 years, immune checkpoint inhibitor can significantly prolong the overall survival of patients with NSCLC (HR = 0.73, 95% CI: 0.66∼0.81, P < 0.00001). In the age 65-75 years group, immune checkpoint inhibitors prolonged overall survival in patients with NSCLC (HR = 0.78, 95% CI:0.71∼0.84, P < 0.00001). However, it has no significant effect on the overall survival of NSCLC patients (HR = 0.88, 95% CI:0.72∼1.08, P > 0.05) in the group older than 75 years. CONCLUSIONS Immune checkpoint inhibitors prolonged the overall survival of NSCLC patients in the age <65 years group and the age 65-75 years group, but in the age ≥75 years group, there was no significant effect on overall survival. This may be related to innate immune and adaptive immune dysregulation due to "immunosenescence" in older patients.
Collapse
Affiliation(s)
- Qi Zhang
- Gansu Provincial Central Hospital, Lanzhou, China
| | | | | | - An Sun
- Gansu Provincial Central Hospital, Lanzhou, China
| | - Tin-Bao Cao
- Gansu Provincial Central Hospital, Lanzhou, China
| | | | - Nan Li
- Gansu Provincial Central Hospital, Lanzhou, China
| | - Tong-Ying Yi
- Gansu Provincial Central Hospital, Lanzhou, China
| | - Kun-Peng Qu
- Gansu Provincial Central Hospital, Lanzhou, China.
| |
Collapse
|
4
|
Huang T, Ren K, Ling X, Li Z, Chen L. Transcription factor Yin Yang 1 enhances epithelial-mesenchymal transition, migration, and stemness of non-small cell lung cancer cells by targeting sonic hedgehog. Mol Cell Biochem 2024:10.1007/s11010-024-05104-y. [PMID: 39261409 DOI: 10.1007/s11010-024-05104-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/21/2024] [Indexed: 09/13/2024]
Abstract
Non-small cell lung cancer (NSCLC) is a frequent type of lung cancer. Transcription factor Yin Yang 1 (YY1), an endogenous transcription factor containing zinc finger structure, can accelerate NSCLC progression. However, the impact of YY1 on the stemness of NSCLC cells and the mechanism of promoting NSCLC cell progression is unclear. YY1 and Sonic hedgehog (Shh) expressions were monitored by RT-qPCR, western blot, and immunohistochemistry. Overall survival was tested through Kaplan-Meier analysis. The interaction between YY1 and Shh was confirmed. Then, cell migration, stemness, and epithelial-mesenchymal transition (EMT) were assessed with functional experiments in vitro and in vivo. YY1 and Shh were highly expressed in NSCLC tissues and positively correlated with the poor OS of NSCLC patients. Functional experiments denoted that YY1 or Shh overexpression could accelerate EMT, migration, and stemness of NSCLC cells, and YY1 or Shh knockdown played the opposite role to its overexpression. Mechanism analysis disclosed that Shh, as a target gene of YY1, was positively related to YY1. The rescued experiment manifested that Shh silencing could reverse the induction effect of YY1 overexpression on EMT, migration, and stemness of NSCLC cells. In vivo experiments also confirmed that YY1 could accelerate tumor growth and EMT and weaken apoptosis. YY1 promotes NSCLC EMT, migration, and stemness by Shh, which might be novel diagnostic markers and therapeutic targets for NSCLC therapy.
Collapse
Affiliation(s)
- Tonghai Huang
- Department of Thoracic Surgery, Shenzhen People's Hospital, 1st Affiliated Hospital of Southern University of Science and Technology, 2, Clinical Medical College of Jinan University, No.1017, East Gate Rd, Shenzhen, 518020, Guangdong, China.
| | - Kangqi Ren
- Department of Thoracic Surgery, Shenzhen People's Hospital, 1st Affiliated Hospital of Southern University of Science and Technology, 2, Clinical Medical College of Jinan University, No.1017, East Gate Rd, Shenzhen, 518020, Guangdong, China
| | - Xiean Ling
- Department of Thoracic Surgery, Shenzhen People's Hospital, 1st Affiliated Hospital of Southern University of Science and Technology, 2, Clinical Medical College of Jinan University, No.1017, East Gate Rd, Shenzhen, 518020, Guangdong, China
| | - Zeyao Li
- Department of Thoracic Surgery, Shenzhen People's Hospital, 1st Affiliated Hospital of Southern University of Science and Technology, 2, Clinical Medical College of Jinan University, No.1017, East Gate Rd, Shenzhen, 518020, Guangdong, China
| | - Lin Chen
- Department of Thoracic Surgery, Shenzhen People's Hospital, 1st Affiliated Hospital of Southern University of Science and Technology, 2, Clinical Medical College of Jinan University, No.1017, East Gate Rd, Shenzhen, 518020, Guangdong, China
| |
Collapse
|
5
|
Luo J, Ren Q, Liu X, Zheng Q, Yang L, Meng M, Ma H, He S. LncRNA MALAT-1 modulates EGFR-TKI resistance in lung adenocarcinoma cells by downregulating miR-125. Discov Oncol 2024; 15:379. [PMID: 39196297 PMCID: PMC11358566 DOI: 10.1007/s12672-024-01133-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 06/28/2024] [Indexed: 08/29/2024] Open
Abstract
Molecular targeted therapy resistance remains a major challenge in treating lung adenocarcinoma (LUAD). The resistance of Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs, epidermal growth factor receptor-tyrosine kinase inhibitor) plays a dominant role in molecular targeted therapy. Our previous research demonstrated the role of MALAT-1 (Metastasis-associated lung adenocarcinoma transcript 1) in the formation of Erlotinib-resistant LUAD cells. This study aims to uncover the mechanism of MALAT-1 overexpression in Erlotinib-resistant LUAD cells. The RT2 LncRNA PCR array system was used to explore MALAT-1 regulation in Erlotinib-resistant LUAD cells through patient serum analysis. Dual luciferase reporter experiments confirmed the binding between MALAT-1 and miR-125, leading to regulation of miR-125 expression. Functional assays were performed to elucidate the impact of MALAT1 on modulating drug resistance, growth, and Epithelial-mesenchymal transition (EMT, Epithelial-mesenchymal transition) in both parental and Erlotinib-resistant LUAD cells. The investigation unveiled the mechanism underlying the competing endogenous RNA (ceRNA, competing endogenouse RNA) pathway. MALAT1 exerted its regulatory effect on miR-125 as a competing endogenous RNA (ceRNA). Moreover, MALAT1 played a role in modulating the sensitivity of LUAD cells to Erlotinib. Rab25 was identified as the direct target of miR-125 and mediated the functional effects of MALAT1 in Erlotinib-resistant LUAD cells. In conclusion, our study reveals overexpress MALAT-1 cause the drug resistance of EGFR-TKIs in non-small cell lung cancer (NSCLC) through the MALAT-1/miR-125/Rab25 axis. These findings present a potential novel therapeutic target and perspective for the treatment of LUAD.
Collapse
Affiliation(s)
- Jie Luo
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Zunyi Medical University, Zunyi, China
| | - Qiaoya Ren
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Zunyi Medical University, Zunyi, China
| | | | - Qian Zheng
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Zunyi Medical University, Zunyi, China
| | - Ling Yang
- Department of Pathology, Suining Central Hospital, Suining, Sichuan, China
| | - Mi Meng
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hu Ma
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China.
- Zunyi Medical University, Zunyi, China.
| | - Sisi He
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Zunyi Medical University, Zunyi, China
| |
Collapse
|
6
|
Wang Y, Qiu Q, Deng X, Wan M. EGFR-TKIs - induced cardiotoxicity in NSCLC: incidence, evaluation, and monitoring. Front Oncol 2024; 14:1426796. [PMID: 38983928 PMCID: PMC11232364 DOI: 10.3389/fonc.2024.1426796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/11/2024] [Indexed: 07/11/2024] Open
Abstract
The advent of targeted drug therapy has greatly changed the treatment landscape of advanced non-small cell lung cancer(NSCLC), but the cardioxic side effects of targeted drug anti-cancer therapy seriously affect the prognosis of NSCLC, and it has become the second leading cause of death in cancer patients. Therefore, early identification of the cardiotoxic side effects of targeted drugs is crucial for the prevention and treatment of cardiovascular diseases. The cardiotoxic side effects that may be caused by novel targeted drugs epidermal growth factor receptor inhibitors, including thromboembolic events, heart failure, cardiomyopathy, arrhythmia and hypertension, are discussed, and the mechanisms of their respective adverse cardiovascular reactions are summarized, to provide useful recommendations for cardiac management of patients with advanced lung cancer to maximize treatment outcomes for lung cancer survivors. Clinicians need to balance the risk-benefit ratio between targeted therapy for malignant tumors and drug-induced cardiotoxicity, and evaluate and monitor TKIs-induced cardiotoxicity through electrocardiogram, cardiac imaging, biomarkers, etc., so as to remove the susceptibility risk factors as soon as possible and provide a reference for the clinical use of such drugs in the treatment of malignant tumors.
Collapse
Affiliation(s)
- Yunlong Wang
- Department of Oncology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Qinggui Qiu
- Department of Oncology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Xuan Deng
- Department of Oncology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Mengchao Wan
- Department of Outpatient, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| |
Collapse
|
7
|
Hua C, Guo Z, Dai M, Zhou J, Ge H, Xue G, Xu F, Ru L, Lv K, Zhang G, Zheng L, Wang M, Teng Y, Yu W, Guo W. Lumbrokinase Extracted from Earthworms Synergizes with Bevacizumab and Chemotherapeutics in Treating Non-Small Cell Lung Cancer by Targeted Inactivation of BPTF/VEGF and NF-κB/COX-2 Signaling. Biomolecules 2024; 14:741. [PMID: 39062456 PMCID: PMC11274885 DOI: 10.3390/biom14070741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/11/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
As a kind of proteolytic enzyme extracted from earthworms, lumbrokinase has been used as an antithrombotic drug clinically. Nevertheless, its potential in anti-cancer, especially in anti-non-small cell lung cancer (NSCLC), as a single form of treatment or in combination with other therapies, is still poorly understood. In this study, we explored the anti-tumor role and the responsive molecular mechanisms of lumbrokinase in suppressing tumor angiogenesis and chemoresistance development in NSCLC and its clinical potential in combination with bevacizumab and chemotherapeutics. Lumbrokinase was found to inhibit cell proliferation in a concentration-dependent manner and caused metastasis suppression and apoptosis induction to varying degrees in NSCLC cells. Lumbrokinase enhanced the anti-angiogenesis efficiency of bevacizumab by down-regulating BPTF expression, decreasing its anchoring at the VEGF promoter region and subsequent VEGF expression and secretion. Furthermore, lumbrokinase treatment reduced IC50 values of chemotherapeutics and improved their cytotoxicity in parental and chemo-resistant NSCLC cells via inactivating the NF-κB pathway, inhibiting the expression of COX-2 and subsequent secretion of PGE2. LPS-induced NF-κB activation reversed its inhibition on NSCLC cell proliferation and its synergy with chemotherapeutic cytotoxicity, while COX-2 inhibitor celecoxib treatment boosted such effects. Lumbrokinase combined with bevacizumab, paclitaxel, or vincristine inhibited the xenograft growth of NSCLC cells in mice more significantly than a single treatment. In conclusion, lumbrokinase inhibited NSCLC survival and sensitized NSCLC cells to bevacizumab or chemotherapeutics treatment by targeted down-regulation of BPTF/VEGF signaling and inactivation of NF-κB/COX-2 signaling, respectively. The combinational applications of lumbrokinase with bevacizumab or chemotherapeutics are expected to be developed as promising candidate therapeutic strategies to improve the efficacy of the original monotherapy in anti-NSCLC.
Collapse
Affiliation(s)
- Chunyu Hua
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Ziyue Guo
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Meng Dai
- Dalian Municipal Central Hospital, Dalian University of Technology, Dalian 116044, China;
| | - Jie Zhou
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Hanxiao Ge
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Guoqing Xue
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Fahui Xu
- The Second Clinical College, Dalian Medical University, Dalian 116044, China;
| | - Liyuan Ru
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Kuan Lv
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Guohui Zhang
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Lina Zheng
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Meiyi Wang
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Yun Teng
- The Second Affiliated Hospital, Dalian Medical University, Dalian 116044, China;
| | - Wendan Yu
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Wei Guo
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| |
Collapse
|
8
|
Zhang Z, Westover D, Tang Z, Liu Y, Sun J, Sun Y, Zhang R, Wang X, Zhou S, Hesilaiti N, Xia Q, Du Z. Wnt/β-catenin signaling in the development and therapeutic resistance of non-small cell lung cancer. J Transl Med 2024; 22:565. [PMID: 38872189 PMCID: PMC11170811 DOI: 10.1186/s12967-024-05380-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024] Open
Abstract
Wnt/β-catenin signaling is a critical pathway that influences development and therapeutic response of non-small cell lung cancer (NSCLC). In recent years, many Wnt regulators, including proteins, miRNAs, lncRNAs, and circRNAs, have been found to promote or inhibit signaling by acting on Wnt proteins, receptors, signal transducers and transcriptional effectors. The identification of these regulators and their underlying molecular mechanisms provides important implications for how to target this pathway therapeutically. In this review, we summarize recent studies of Wnt regulators in the development and therapeutic response of NSCLC.
Collapse
Affiliation(s)
- Zixu Zhang
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - David Westover
- High-Throughput Analytics, Analytical Research and Development, Merck & Co. Inc., Rahway, NJ, USA
| | - Zhantong Tang
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - Yue Liu
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - Jinghan Sun
- School of Life Science and Technology, Southeast University, Nanjing, 210018, China
| | - Yunxi Sun
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - Runqing Zhang
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - Xingyue Wang
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - Shihui Zhou
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - Nigaerayi Hesilaiti
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - Qi Xia
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - Zhenfang Du
- Department of Genetic and Developmental Biology, School of Medicine, Southeast University, Nanjing, 210003, China.
| |
Collapse
|
9
|
Gou Q, Gou Q, Gan X, Xie Y. Novel therapeutic strategies for rare mutations in non-small cell lung cancer. Sci Rep 2024; 14:10317. [PMID: 38705930 PMCID: PMC11070427 DOI: 10.1038/s41598-024-61087-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/30/2024] [Indexed: 05/07/2024] Open
Abstract
Lung cancer is still the leading cause of cancer-related mortality. Over the past two decades, the management of non-small cell lung cancer (NSCLC) has undergone a significant revolution. Since the first identification of activating mutations in the epidermal growth factor receptor (EGFR) gene in 2004, several genetic aberrations, such as anaplastic lymphoma kinase rearrangements (ALK), neurotrophic tropomyosin receptor kinase (NTRK) and hepatocyte growth factor receptor (MET), have been found. With the development of gene sequencing technology, the development of targeted drugs for rare mutations, such as multikinase inhibitors, has provided new strategies for treating lung cancer patients with rare mutations. Patients who harbor this type of oncologic driver might acquire a greater survival benefit from the use of targeted therapy than from the use of chemotherapy and immunotherapy. To date, more new agents and regimens can achieve satisfactory results in patients with NSCLC. In this review, we focus on recent advances and highlight the new approval of molecular targeted therapy for NSCLC patients with rare oncologic drivers.
Collapse
Affiliation(s)
- Qitao Gou
- Department of Radiation Oncology and Department of Head & Neck Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qiheng Gou
- Department of Radiation Oncology and Department of Head & Neck Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Xiaochuan Gan
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuxin Xie
- Department of Medical Oncology of Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Yang X, Liu Q, Li G. Anti-NSCLC role of SCN4B by negative regulation of the cGMP-PKG pathway: Integrated utilization of bioinformatics analysis and in vitro assay validation. Drug Dev Res 2024; 85:e22192. [PMID: 38678552 DOI: 10.1002/ddr.22192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 04/04/2024] [Accepted: 04/12/2024] [Indexed: 05/01/2024]
Abstract
Non-small cell lung cancer (NSCLC) is a malignant tumor with low overall cure and survival rates. Uncovering abnormally expressed genes is significantly important for developing novel targeted therapies in NSCLC. This study aimed to discover new differentially expressed genes (DEGs) of NSCLC. The DEGs of NSCLC were identified in eight data sets from Gene Expression Omnibus (GEO) database. The expression profiles and the prognostic significance of SCN4B in LUAD and LUSC were analyzed using GEPIA database. LinkedOmics was used to identify co-expressed genes with SCN4B, which were further subjected to KEGG pathway enrichment analysis. SCN4B-overexpressing plasmid (pcDNA/SCN4B) was transfected into A549 and NCI-H2170 cells to elevate the expression of SCN4B. MTT and TUNEL assays were performed to evaluate cell viability and apoptosis. Relying on the screened DEGs from GEO database, we identified that SCN4B was significantly downregulated in LUAD and LUSC. We confirmed the downregulation of SCN4B in NSCLC tissues using GEPIA database. SCN4B has a prognostic value in LUAD, but not LUSC. KEGG pathway enrichment analysis of SCN4B-related genes showed that cGMP-PKG signaling pathway might be involved in the role of SCN4B in NSCLC. Overexpression of SCN4B in A549 and NCI-H2170 cells inhibited the cell viability. Besides, SCN4B overexpression induced apoptosis of A549 and NCI-H2170 cells. SCN4B inhibited the expression of PKG1 and p-CREB in NSCLC cells. Moreover, the inhibitory effects of SCN4B on tumor malignancy were attenuated by the activator of PKG. In conclusion, integrated bioinformatical analysis proved that SCN4B was downregulated and had a prognostic significance in NSCLC. In vitro experimental studies demonstrated that SCN4B regulated NSCLC cells viability and apoptosis via inhibiting cGMP-PKG signaling pathway.
Collapse
Affiliation(s)
- Xiujun Yang
- Department of Respiratory and Critical Care Medicine, Huai'an People's Hospital of Hongze District, Huai'an, China
| | - Qun Liu
- Medical Ward 20, Lianshui County People's Hospital, Huai'an, China
| | - Gang Li
- Department of Respiratory and Critical Care Medicine, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| |
Collapse
|
11
|
Ji X, Liu M, Zhang T, Zhang W, Xue F, Wan Q, Liu Y. KRAS/PI3K axis driven GTF3C6 expression and promotes LUAD via FAK pathway. J Adv Res 2024:S2090-1232(24)00171-1. [PMID: 38685529 DOI: 10.1016/j.jare.2024.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024] Open
Abstract
INTRODUCTION Effective targeting drugs for KRAS mutation-mediated Lung Adenocarcinoma (LUAD) are currently are limited. OBJECTIVES Investigating and intervening in the downstream key target genes of KRAS is crucial for clinically managing KRAS mutant-driven LUAD. GTF3C6, a newly identified member of the general transcription factor III (GTF3) family, plays a role in the transcription of RNA polymerase III (pol III)-dependent genes. However, its involvement in cancer remains unexplored. METHODS This study examined the expression, roles, and potential molecular mechanisms of GTF3C6 in LUAD tissues, LSL-KrasG12D/+;LSL-p53-/- LUAD mouse models, and LUAD patients-derived organoid using Western blot, qRT-PCR, immunofluorescence, immunohistochemistry, and gene manipulation assays. RESULTS We present the first evidence that GTF3C6 is highly expressed in LUAD tissues, LSL-KrasG12D/+;LSL-p53-/- LUAD mouse models, and LUAD organoids, correlating with poor clinical prognosis. Furthermore, GTF3C6 was found to promote anchorage-independent proliferation, migration, and invasion of LUAD cells. Mechanistically, KRAS mutation drives GTF3C6 expression through the PI3K pathway, and GTF3C6 knockdown reverses the malignant phenotype of KRAS mutation-driven LUAD cells. Additionally, the FAK pathway emerged as a crucial downstream signaling pathway through which GTF3C6 mediates the malignant phenotype of LUAD. Finally, GTF3C6 knockdown suppresses LUAD organoid formation and inhibits tumor growth in vivo. CONCLUSION Our findings demonstrate that GTF3C6, driven by KRAS mutation, promotes LUAD development by regulating FAK phosphorylation, suggesting its potential as a biomarker and therapeutic target in KRAS mutant-driven LUAD.
Collapse
Affiliation(s)
- Xingzhao Ji
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Infections Respiratory Disease, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mingqiang Liu
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Department of Pharmacy, Pingdu People's Hospital, Qingdao, Shandong 266799, China
| | - Tianyi Zhang
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Weiying Zhang
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Fuyuan Xue
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Qiang Wan
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| | - Yi Liu
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Infections Respiratory Disease, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
12
|
Ramos R, Moura CS, Costa M, Lamas NJ, Correia R, Garcez D, Pereira JM, Sousa C, Vale N. Enhancing Lung Cancer Care in Portugal: Bridging Gaps for Improved Patient Outcomes. J Pers Med 2024; 14:446. [PMID: 38793028 PMCID: PMC11121920 DOI: 10.3390/jpm14050446] [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: 03/20/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Lung cancer has the highest incidence and cancer-related mortality worldwide. In Portugal, it ranks as the fourth most common cancer, with nearly 6000 new cases being diagnosed every year. Lung cancer is the main cause of cancer-related death among males and the third cause of cancer-related death in females. Despite the globally accepted guidelines and recommendations for what would be the ideal path for a lung cancer patient, several challenges occur in real clinical management across the world. The recommendations emphasize the importance of adequate screening of high-risk individuals, a precise tumour biopsy, and an accurate final diagnosis to confirm the neoplastic nature of the nodule. A detailed histological classification of the lung tumour type and a comprehensive molecular characterization are of utmost importance for the selection of an efficacious and patient-directed therapeutic approach. However, in the context of the Portuguese clinical organization and the national healthcare system, there are still several gaps in the ideal pathway for a lung cancer patient, involving aspects ranging from the absence of a national lung cancer screening programme through difficulties in histological diagnosis and molecular characterization to challenges in therapeutic approaches. In this manuscript, we address the most relevant weaknesses, presenting several proposals for potential solutions to improve the management of lung cancer patients, helping to decisively improve their overall survival and quality of life.
Collapse
Affiliation(s)
- Raquel Ramos
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal; (R.R.); (C.S.)
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Molecular Diagnostics Laboratory, Unilabs Portugal, Centro Empresarial Lionesa Porto, Rua Lionesa, 4465-671 Leça do Balio, Portugal; (M.C.); (N.J.L.)
| | - Conceição Souto Moura
- Pathology Laboratory, Unilabs Portugal, Rua Manuel Pinto de Azevedo 173, 4100-321 Porto, Portugal;
| | - Mariana Costa
- Molecular Diagnostics Laboratory, Unilabs Portugal, Centro Empresarial Lionesa Porto, Rua Lionesa, 4465-671 Leça do Balio, Portugal; (M.C.); (N.J.L.)
| | - Nuno Jorge Lamas
- Molecular Diagnostics Laboratory, Unilabs Portugal, Centro Empresarial Lionesa Porto, Rua Lionesa, 4465-671 Leça do Balio, Portugal; (M.C.); (N.J.L.)
- Anatomic Pathology Service, Pathology Department, Centro Hospitalar Universitário de Santo António (CHUdSA), Largo Professor Abel Salazar, 4099-001 Porto, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, Rua da Universidade, 4710-057 Braga, Portugal
| | - Renato Correia
- Technology & Innovation Department, Unilabs Portugal, Rua Manuel Pinto de Azevedo 173, 4100-321 Porto, Portugal; (R.C.); (D.G.)
| | - Diogo Garcez
- Technology & Innovation Department, Unilabs Portugal, Rua Manuel Pinto de Azevedo 173, 4100-321 Porto, Portugal; (R.C.); (D.G.)
| | - José Miguel Pereira
- Radiology Department, Unilabs Portugal, Rua de Diogo Botelho 485, 4150-255 Porto, Portugal;
| | - Carlos Sousa
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal; (R.R.); (C.S.)
- Molecular Diagnostics Laboratory, Unilabs Portugal, Centro Empresarial Lionesa Porto, Rua Lionesa, 4465-671 Leça do Balio, Portugal; (M.C.); (N.J.L.)
| | - Nuno Vale
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal; (R.R.); (C.S.)
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
13
|
Xiu-Ying H, Yue-Xiang Z, Hui-Si Y, Hong-Zhou Y, Qing-Jie X, Ting-Hua W. PDGFBB facilitates tumorigenesis and malignancy of lung adenocarcinoma associated with PI3K-AKT/MAPK signaling. Sci Rep 2024; 14:4191. [PMID: 38378786 PMCID: PMC10879171 DOI: 10.1038/s41598-024-54801-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 02/16/2024] [Indexed: 02/22/2024] Open
Abstract
Lung adenocarcinoma (LUAD) remains one of the most aggressive tumors and the efficacy of conventional treatment has been bleak. Nowadays, gene-targeted therapy has become a new favorite in tumor therapy. Herein, we investigated the effect of platelet derived growth factor BB (PDGFBB) on LUAD. Firstly, PDGFBB was upregulated in LUAD patients and closely linked with poor survival. Furthermore, the expression of PDGFBB and PDGFRα/β in LUAD cells was higher than that in normal lung cells. By loss-of-function with herpes simplex virus (HSV)-PDGFi-shRNA, we found that PDGFBB knockdown caused a significant decrease in proliferation and migration, but evoked apoptosis of LUAD cells in vitro. Conversely, exogenous PDGFBB held adverse effect. Additionally, A549 cells with PDGFBB knockdown had a low probability of tumorigenesis in vivo. Moreover, PDGFBB knockdown restrained the growth of xenografts derived from normal A549 cells. Mechanistically, PDGFBB knockdown suppressed PI3K/AKT and Ras/MAPK signaling, while PDGFBB was the opposite. Therefore, we concluded that PDGFBB might facilitate the tumorigenesis and malignancy of LUAD through its functional downstream nodes-PI3K/AKT and Ras/MAPK signaling, which supported that PDGFBB could serve as a rational therapeutic target for LUAD.
Collapse
Affiliation(s)
- He Xiu-Ying
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zheng Yue-Xiang
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Yang Hui-Si
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Yu Hong-Zhou
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xia Qing-Jie
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Wang Ting-Hua
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China.
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China.
| |
Collapse
|
14
|
Wang Y, Qi H, Wang T, Zhang W, Shi X, Zhan Q, Li Q, Zhong M. STAT3 and STAT6 polymorphisms predict the severity of adverse reactions in Chinese NSCLC patients receiving EGFR-TKIs therapy. J Chemother 2024; 36:61-71. [PMID: 37151185 DOI: 10.1080/1120009x.2023.2203610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 04/12/2023] [Indexed: 05/09/2023]
Abstract
A total of 162 non-small cell lung cancer (NSCLC) patients were divided into discovery (N = 68) and validation (N = 94) groups. Nine Janus kinase/Signal transducer and activator of transcription (JAK/STAT) pathway-related single nucleotide polymorphisms were selected to explore the potential associations between genetic polymorphisms and adverse drug reactions (ADRs). The TT genotype of STAT6 rs324011 was significantly associated with severe ADRs in the recessive genetic model (TT vs. CC + CT, OR = 13.5, 95% CI = 2.12-86.09, p = 0.006 in the discovery group; OR = 8.41, 95% CI = 1.95-36.19, p = 0.004 in the validation group). The T allele was associated with a higher incidence of severe ADRs than was the C allele of rs324011 (OR = 3.67, 95% CI = 1.46-9.19, p = 0.006 in the discovery group; OR = 3.17, 95% CI = 1.44-6.99, p = 0.004 in the validation group). Patients with the CC genotype in STAT3 rs1053023 (and rs1053005) or the TT genotype of STAT6 rs324011 were likely to experience severe epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) related ADRs.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Huijie Qi
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Tianxiao Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenxin Zhang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaojin Shi
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiong Zhan
- Department of Oncology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qunyi Li
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Mingkang Zhong
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Ge Y, Li J, Gong W, Wang J, Wei X, Liu J, Wang S, Wang L, Sun H, Cheng Q, Sun Y, Dang Q, Sun Y, Gao A. Efficacy of first-line treatment options beyond RET-TKIs in advanced RET-rearranged non-small cell lung cancer: A multi-center real-world study. Cancer Med 2024; 13:e6960. [PMID: 38349001 PMCID: PMC10832335 DOI: 10.1002/cam4.6960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/21/2023] [Accepted: 01/12/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Although RET-tyrosine kinase inhibitors (RET-TKIs) are the preferred first-line therapy for advanced RET-arranged NSCLC, most patients cannot afford them. In this population, bevacizumab, immunotherapy, and chemotherapy are the most commonly used regimens. However, the optimal scheme beyond RET-TKIs has not been defined in the first-line setting. METHODS This retrospective study included 86 stage IV NSCLC patients harboring RET rearrangement from six cancer centers between May 2017 and October 2022. RET-TKIs, chemotherapy, or one of the combination therapies (including immune checkpoint inhibitor (ICI) combined with chemotherapy (I + C), bevacizumab combined with chemotherapy (B + C), ICI and bevacizumab combined with chemotherapy (I + B + C)), were used as the first-line therapeutics. The clinical outcomes and safety were evaluated. RESULTS Fourteen of the 86 patients received RET-TKIs, 57 received combination therapies, and 15 received chemotherapy alone. Their medium PFS (mPFS) were 16.92 months (95% CI: 5.9-27.9 months), 8.7 months (95% CI: 6.5-11.0 months), and 5.55 months (95% CI: 2.4-8.7 months) respectively. Among all the combination schemes, B + C (p = 0.007) or I + B + C (p = 0.025) gave beneficial PFS compared with chemotherapy, while I + C treatment (p = 0.169) generated comparable PFS with chemotherapy. In addition, I + B + C treatment had a numerically longer mPFS (12.21 months) compared with B + C (8.74 months) or I + C (7.89 months) schemes. In terms of safety, I + B + C treatment led to the highest frequency of hematological toxicity (50%) and vomiting (75%), but no ≥G3 adverse effect was observed. CONCLUSIONS I + B + C might be a preferred option beyond RET-TKIs in the first-line therapy of RET-arranged NSCLC. Combination with Bevacizumab rather than with ICIs offered favorable survival compared with chemotherapy alone.
Collapse
Affiliation(s)
- Yihui Ge
- Phase I Clinical Research CenterShandong University Cancer CenterJinanChina
| | - Juan Li
- Phase I Clinical Research CenterShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Wenjing Gong
- Medical DepartmentThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiChina
| | - Jian Wang
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanChina
| | - Xiaojuan Wei
- Department of OncologyThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Jing Liu
- Department of OncologyAffiliated Hospital of Weifang Medical UniversityWeifangP. R. China
| | - Shuyun Wang
- Phase I Clinical Research CenterShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Leirong Wang
- Phase I Clinical Research CenterShandong University Cancer CenterJinanChina
| | | | - Qinglei Cheng
- Phase I Clinical Research CenterShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | | | - Qi Dang
- Phase I Clinical Research CenterShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Yuping Sun
- Phase I Clinical Research CenterShandong University Cancer CenterJinanChina
| | - Aiqin Gao
- Department of Thoracic Radiation OncologyShandong University Cancer CenterJinanChina
| |
Collapse
|
16
|
Wang J, Mu HJ, Sun YL, Yuan B, Wang Y. Use of honokiol in lung cancer therapy: a mini review of its pharmacological mechanism. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2023; 25:1029-1037. [PMID: 37010929 DOI: 10.1080/10286020.2023.2193695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 03/16/2023] [Accepted: 03/16/2023] [Indexed: 06/19/2023]
Abstract
Honokiol (3',5-di-(2-propenyl)-1,1'-biphenyl-2,2'-diol) is a biologically active natural product derived from Magnolia and has been shown to have excellent biological activities. This paper discusses research progress on the use of honokiol in the treatment of lung cancer, as studies have confirmed that honokiol can exert anti-lung-cancer effects through multiple pathways and multiple signaling pathways, such as inhibiting angiogenesis, affecting mitochondrial function and apoptosis, regulating of autophagy and epithelial-mesenchymal transition (EMT). In addition, honokiol combined with other chemotherapy drugs is also a way in which it can be applied.
Collapse
Affiliation(s)
- Jing Wang
- Department of Biology Science and Technology, Baotou Teacher's College, Baotou 014030, China
| | - Hui-Juan Mu
- Department of Drug Clinical Trials, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Yu-Li Sun
- Department of Hepatobiliary Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Bo Yuan
- Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Ying Wang
- Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| |
Collapse
|
17
|
Meira DD, de Castro e Caetano MC, Casotti MC, Zetum ASS, Gonçalves AFM, Moreira AR, de Oliveira AH, Pesente F, Santana GM, de Almeida Duque D, Pereira GSC, de Castro GDSC, Pavan IP, Chagas JPS, Bourguignon JHB, de Oliveira JR, Barbosa KRM, Altoé LSC, Louro LS, Merigueti LP, Alves LNR, Machado MRR, Roque MLRO, Prates PS, de Paula Segáua SH, dos Santos Uchiya T, Louro TES, Daleprane VE, Guaitolini YM, Vicente CR, dos Reis Trabach RS, de Araújo BC, dos Santos EDVW, de Paula F, Lopes TJS, de Carvalho EF, Louro ID. Prognostic Factors and Markers in Non-Small Cell Lung Cancer: Recent Progress and Future Challenges. Genes (Basel) 2023; 14:1906. [PMID: 37895255 PMCID: PMC10606762 DOI: 10.3390/genes14101906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/29/2023] [Accepted: 10/01/2023] [Indexed: 10/29/2023] Open
Abstract
Lung cancer is a highly aggressive neoplasm and, despite the development of recent therapies, tumor progression and recurrence following the initial response remains unsolved. Several questions remain unanswered about non-small cell lung cancer (NSCLC): (1) Which patients will actually benefit from therapy? (2) What are the predictive factors of response to MAbs and TKIs? (3) What are the best combination strategies with conventional treatments or new antineoplastic drugs? To answer these questions, an integrative literature review was carried out, searching articles in PUBMED, NCBI-PMC, Google Academic, and others. Here, we will examine the molecular genetics of lung cancer, emphasizing NSCLC, and delineate the primary categories of inhibitors based on their molecular targets, alongside the main treatment alternatives depending on the type of acquired resistance. We highlighted new therapies based on epigenetic information and a single-cell approach as a potential source of new biomarkers. The current and future of NSCLC management hinges upon genotyping correct prognostic markers, as well as on the evolution of precision medicine, which guarantees a tailored drug combination with precise targeting.
Collapse
Affiliation(s)
- Débora Dummer Meira
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Maria Clara de Castro e Caetano
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Matheus Correia Casotti
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Aléxia Stefani Siqueira Zetum
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - André Felipe Monteiro Gonçalves
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - André Rodrigues Moreira
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Augusto Henrique de Oliveira
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Fellipe Pesente
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Gabriel Mendonça Santana
- Centro de Ciências da Saúde, Curso de Medicina, Universidade Federal do Espírito Santo (UFES), Vitória 29090-040, Brazil
| | - Daniel de Almeida Duque
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Gierleson Santos Cangussu Pereira
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Giulia de Souza Cupertino de Castro
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Isabele Pagani Pavan
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - João Pedro Sarcinelli Chagas
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - José Henrique Borges Bourguignon
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Juliana Ribeiro de Oliveira
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Karen Ruth Michio Barbosa
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Lorena Souza Castro Altoé
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Luana Santos Louro
- Centro de Ciências da Saúde, Curso de Medicina, Universidade Federal do Espírito Santo (UFES), Vitória 29090-040, Brazil
| | - Luiza Poppe Merigueti
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Lyvia Neves Rebello Alves
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Marlon Ramos Rosado Machado
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Maria Luísa Rodrigues Oliveira Roque
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Pedro Santana Prates
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Sayuri Honorio de Paula Segáua
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Taissa dos Santos Uchiya
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Thomas Erik Santos Louro
- Escola Superior de Ciências da Santa Casa de Misericórdia de Vitória (EMESCAM), Curso de Medicina, Vitória 29027-502, Brazil
| | - Vinicius Eduardo Daleprane
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Yasmin Moreto Guaitolini
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Creuza Rachel Vicente
- Departamento de Medicina Social, Universidade Federal do Espírito Santo, Vitória 29090-040, Brazil
| | - Raquel Silva dos Reis Trabach
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Bruno Cancian de Araújo
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Eldamária de Vargas Wolfgramm dos Santos
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Flávia de Paula
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Tiago José S. Lopes
- Department of Reproductive Biology, National Center for Child Health and Development Research Institute, Tokyo 157-8535, Japan
| | - Elizeu Fagundes de Carvalho
- Instituto de Biologia Roberto Alcântara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20551-030, Brazil
| | - Iúri Drumond Louro
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| |
Collapse
|
18
|
Pan M, Wei X, Xiang X, Liu Y, Zhou Q, Yang W. Targeting CXCL9/10/11-CXCR3 axis: an important component of tumor-promoting and antitumor immunity. Clin Transl Oncol 2023; 25:2306-2320. [PMID: 37076663 DOI: 10.1007/s12094-023-03126-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 02/13/2023] [Indexed: 04/21/2023]
Abstract
Chemokines are chemotactic-competent molecules composed of a family of small cytokines, playing a key role in regulating tumor progression. The roles of chemokines in antitumor immune responses are of great interest. CXCL9, CXCL10, and CXCL11 are important members of chemokines. It has been widely investigated that these three chemokines can bind to their common receptor CXCR3 and regulate the differentiation, migration, and tumor infiltration of immune cells, directly or indirectly affecting tumor growth and metastasis. Here, we summarize the mechanism of how the CXCL9/10/11-CXCR3 axis affects the tumor microenvironment, and list the latest researches to find out how this axis predicts the prognosis of different cancers. In addition, immunotherapy improves the survival of tumor patients, but some patients show drug resistance. Studies have found that the regulation of CXCL9/10/11-CXCR3 on the tumor microenvironment is involved in the process of changing immunotherapy resistance. Here we also describe new approaches to restoring sensitivity to immune checkpoint inhibitors through the CXCL9/10/11-CXCR3 axis.
Collapse
Affiliation(s)
- Minjie Pan
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Xiaoshan Wei
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Xuan Xiang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Yanhong Liu
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Qiong Zhou
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Weibing Yang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China.
| |
Collapse
|
19
|
Héluain V, Prévot G, Cabarrou B, Calvayrac O, Taranchon- Clermont E, Didier A, Tabourier-Gouin S, Milia J, Mazières J. Clinical and molecular analysis of lung cancers associated with fibrosing interstitial lung disease. Respir Med Res 2023; 83:100946. [DOI: 10.1016/j.resmer.2022.100946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/02/2022] [Accepted: 07/10/2022] [Indexed: 12/03/2022]
|
20
|
Laface C, Maselli FM, Santoro AN, Iaia ML, Ambrogio F, Laterza M, Guarini C, De Santis P, Perrone M, Fedele P. The Resistance to EGFR-TKIs in Non-Small Cell Lung Cancer: From Molecular Mechanisms to Clinical Application of New Therapeutic Strategies. Pharmaceutics 2023; 15:1604. [PMID: 37376053 DOI: 10.3390/pharmaceutics15061604] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/13/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Almost 17% of Western patients affected by non-small cell lung cancer (NSCLC) have an activating epidermal growth factor receptor (EGFR) gene mutation. Del19 and L858R are the most-common ones; they are positive predictive factors for EGFR tyrosine kinase inhibitors (TKIs). Currently, osimertinib, a third-generation TKI, is the standard first-line therapy for advanced NSCLC patients with common EGFR mutations. This drug is also administered as a second-line treatment for those patients with the T790M EGFR mutation and previously treated with first- (erlotinib, gefitinib) or second- (afatinib) generation TKIs. However, despite the high clinical efficacy, the prognosis remains severe due to intrinsic or acquired resistance to EGRF-TKIs. Various mechanisms of resistance have been reported including the activation of other signalling pathways, the development of secondary mutations, the alteration of the downstream pathways, and phenotypic transformation. However, further data are needed to achieve the goal of overcoming resistance to EGFR-TKIs, hence the necessity of discovering novel genetic targets and developing new-generation drugs. This review aimed to deepen the knowledge of intrinsic and acquired molecular mechanisms of resistance to EGFR-TKIs and the development of new therapeutic strategies to overcome TKIs' resistance.
Collapse
Affiliation(s)
- Carmelo Laface
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| | | | | | - Maria Laura Iaia
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| | - Francesca Ambrogio
- Section of Dermatology, Department of Biomedical Science and Human Oncology, University of Bari, 70124 Bari, Italy
| | - Marigia Laterza
- Division of Cardiac Surgery, University of Bari, 70124 Bari, Italy
| | - Chiara Guarini
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| | - Pierluigi De Santis
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| | - Martina Perrone
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| | - Palma Fedele
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| |
Collapse
|
21
|
Zeng X, Tang X, Chen X, Wen H. RNF182 induces p65 ubiquitination to affect PDL1 transcription and suppress immune evasion in lung adenocarcinoma. Immun Inflamm Dis 2023; 11:e864. [PMID: 37249301 PMCID: PMC10201958 DOI: 10.1002/iid3.864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/06/2023] [Accepted: 04/21/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND The RING finger (RNF) proteins are a large group of ubiquitin ligases whose aberrant expression is often associated with disease progression. This study examines the function of RNF protein 182 (RNF182) in lung adenocarcinoma (LUAD) cells and its impact on p65 and programmed death ligand 1 (PDL1) regulation. METHODS Expression of RNF182, p65, and PDL1 in LUAD tissues and cells was measured using immunohistochemistry, reverse transcription quantitative polymerase chain reaction (RT-qPCR), and/or western blot (WB) assays. LUAD cells were induced to overexpress RNF182 and p65, followed by cell counting kit-8, colony formation, Transwell, and flow cytometry assays to evaluate the cells' malignant phenotype. Coimmunoprecipitation and WB assays were used to verify RNF182's effect on p65 ubiquitination. Chromatin immunoprecipitation-qPCR and luciferase assays were used to analyze p65's transcriptional regulation of PDL1. Coculture of LUAD with CD8+ cytotoxic T cells was performed to detect lactate dehydrogenase release and interferon-γ and interleukin-2 concentrations. LUAD cells were implanted in mice to analyze tumorigenicity. RESULTS RNF182 was poorly expressed, while p65 and PDL1 were highly expressed in LUAD tissues and cells. RNF182 overexpression suppressed the malignant properties of LUAD cells, and it promoted p65 ubiquitination and protein degradation. p65 activated PDL1 transcription. Overexpression of RNF182 suppressed the PDL1 expression, increased the cytotoxicity in LUAD cells cocultured with CD8+ T cells, and suppressed the tumorigenesis of cancer cells in vivo. However, these tumor-suppressive effects of RNF182 on LUAD cells were blocked by p65 restoration. CONCLUSION This research demonstrates that RNF182 induces p65 ubiquitination to suppress PDL1 transcription and immunosuppression in LUAD.
Collapse
Affiliation(s)
- Xingdu Zeng
- Department of Respiratory MedicineThe First Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiPeople's Republic of China
| | - Xiaoyuan Tang
- Department of Respiratory MedicineThe First Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiPeople's Republic of China
| | - Xingxiang Chen
- Department of Respiratory MedicineThe First Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiPeople's Republic of China
| | - Huilan Wen
- Department of Respiratory MedicineThe First Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiPeople's Republic of China
| |
Collapse
|
22
|
Shen M, Lu C, Gao J. Prognostic influence of PD-1/PD-L1 suppressors in combination with chemotherapeutic agents for non-small cell pulmonary carcinoma: system review and meta-analysis. Front Oncol 2023; 13:1137913. [PMID: 37152014 PMCID: PMC10154692 DOI: 10.3389/fonc.2023.1137913] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/20/2023] [Indexed: 05/09/2023] Open
Abstract
Background Lung cancer is a common malignant tumor, which is seriously harmful to human life and health. Nowadays, it has gradually become one of the best treatments for non-small cell lung cancer (NSCLC) to combine immunotherapy and chemotherapy, and its clinical efficacy is preliminary. Nevertheless, substantial differences exist between various studies and various indicators. Despite their unconvincing results, high-quality research evidence is needed to support them. In this case, further correlative studies are necessary to investigate the prognostic outcomes of PD-1/PD-L1 suppressors in combination with chemotherapeutic drugs in NSCLC. Methods The online public databases were searchable for the clinical trials that consisted of NSCLC patients who had concluded their chemotherapy and who had accepted PD-1/PD-L1 suppressors. The time-span of the search spanned from the beginning to the end of the database. Two investigators retrieved the data independently. RevMan 5.3 statistical software was utilized for the assessment of bias risk. The software followed the Cochrane Handbook 5.3 guidelines. Results There were seven clinically controlled studies with 2781 NSCLC samples finally included in this study. A meta-analysis of the post-treatment overall response rate (ORR) was undertaken. A remarkably higher ORR rate was observed in the study group (p<0.05). Study participants had a noticeably longer PFS (HR=0.61, 95% CI=0.54-0.70, P<0.00001). Study participants had markedly longer overall survival (OS) (HR=0.651, 95% CI=0.52-0.82, P<0.05). The incidence of adverse events (AEs) of Grade 3 or above was not clinically clearly different (P>0.05), as demonstrated by the incidence of AEs. The funnel plots were separately charted in accordance with ORR rate, PFE, OS, and Grade 3 AEs. The majority of the funnel plots were symmetrical and a minority of funnel plots were asymmetrical, indicating the heterogeneity of research and the limited evidence available may lead to some publication bias in the contained literature. Conclusion The combined PD-1/PD-L1 inhibitors with conventional chemotherapy can dramatically elevate the prognosis of NSCLC patients, obviously enhancing the ORR rate and prolonging their PFS and OS. Furthermore, it was found that adding PD-1/PD-L1 inhibitors to conventional chemotherapy did not result in any additional adverse effects.
Collapse
Affiliation(s)
| | - Chunxia Lu
- Department of Respiratory and critical care, Qidong People’s Hospital/Qidong Liver Cancer Institute/Affiliated Qidong Hospital of Nantong University, Qidong, China
| | | |
Collapse
|
23
|
Sun S, Zou Y, Xu N, Wang K, Rong S, Lv J, Hu B, Mai Y, Zhu D, Ding L. Long non-coding RNA ATB expedites non-small cell lung cancer progression by the miR-200b/fibronectin 1 axis. J Clin Lab Anal 2023; 37:e24822. [PMID: 36806318 PMCID: PMC10020841 DOI: 10.1002/jcla.24822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Long non-coding RNA (lncRNA) ATB belongs to an active modulator in multiple cancers, but its expression along with potential underlying non-small cell lung cancer (NSCLC) is obscure. Our study aimed to investigate the role and potential mechanism of LncRNA ATB in NSCLC. METHODS LncRNA ATB expression in NSCLC tissues and cell lines was detected by qRT-PCR. Effects of LncRNA ATB on NSCLC cell proliferation, migration and invasion were assessed by MTS, colony formation and transwell assays. The connection among LncRNA ATB, miR-200b and fibronectin 1 (FN1) was determined by bioformatics prediction and luciferase reporter assay. RESULTS In this research, upregulation of LncRNA ATB was discovered in NSCLC tissue samples and cell lines. LncRNA ATB was positively related to advanced tumor phase as well as lymph node metastasis. Cell function assays reflected LncRNA ATB expedited NSCLC cells proliferation, migration and invasion. LncRNA ATB promoted fibronectin 1 (FN1) expression via inhibiting miR-200b. Furthermore, LncRNA ATB depletion suppressed NSCLC cells proliferation, migration and invasion, while miR-200b inhibitor or pcDNA-FN1 rescued these effects. CONCLUSION In summary, our outcomes elucidated that LncRNA ATB/miR-200b axis expedited NSCLC cells proliferation, migration and invasion by up-regulating FN1.
Collapse
Affiliation(s)
- Shifang Sun
- Department of Geriatrics MedicineThe Affiliated Hospital of Medical School of Ningbo UniversityNingboChina
| | - Yifan Zou
- Department of Geriatrics MedicineThe Affiliated Hospital of Medical School of Ningbo UniversityNingboChina
| | - Ningjie Xu
- School of MedicineNingbo UniversityNingboChina
| | - Kaiyue Wang
- Department of Geriatrics MedicineThe Affiliated Hospital of Medical School of Ningbo UniversityNingboChina
| | - Shanshan Rong
- Department of Geriatrics MedicineThe Affiliated Hospital of Medical School of Ningbo UniversityNingboChina
| | - Jiarong Lv
- Department of Geriatrics MedicineThe Affiliated Hospital of Medical School of Ningbo UniversityNingboChina
| | - Bin Hu
- School of MedicineNingbo UniversityNingboChina
| | - Yifeng Mai
- Department of Geriatrics MedicineThe Affiliated Hospital of Medical School of Ningbo UniversityNingboChina
| | - Decai Zhu
- Department of Geriatrics MedicineThe Affiliated Hospital of Medical School of Ningbo UniversityNingboChina
| | - Liren Ding
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Zhejiang University Medical CollegeHangzhouChina
| |
Collapse
|
24
|
Mei Y, Wu D, Berg J, Tolksdorf B, Roehrs V, Kurreck A, Hiller T, Kurreck J. Generation of a Perfusable 3D Lung Cancer Model by Digital Light Processing. Int J Mol Sci 2023; 24:ijms24076071. [PMID: 37047045 PMCID: PMC10094257 DOI: 10.3390/ijms24076071] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/10/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Lung cancer still has one of the highest morbidity and mortality rates among all types of cancer. Its incidence continues to increase, especially in developing countries. Although the medical field has witnessed the development of targeted therapies, new treatment options need to be developed urgently. For the discovery of new drugs, human cancer models are required to study drug efficiency in a relevant setting. Here, we report the generation of a non-small cell lung cancer model with a perfusion system. The bioprinted model was produced by digital light processing (DLP). This technique has the advantage of including simulated human blood vessels, and its simple assembly and maintenance allow for easy testing of drug candidates. In a proof-of-concept study, we applied gemcitabine and determined the IC50 values in the 3D models and 2D monolayer cultures and compared the response of the model under static and dynamic cultivation by perfusion. As the drug must penetrate the hydrogel to reach the cells, the IC50 value was three orders of magnitude higher for bioprinted constructs than for 2D cell cultures. Compared to static cultivation, the viability of cells in the bioprinted 3D model was significantly increased by approximately 60% in the perfusion system. Dynamic cultivation also enhanced the cytotoxicity of the tested drug, and the drug-mediated apoptosis was increased with a fourfold higher fraction of cells with a signal for the apoptosis marker caspase-3 and a sixfold higher fraction of cells positive for PARP-1. Altogether, this easily reproducible cancer model can be used for initial testing of the cytotoxicity of new anticancer substances. For subsequent in-depth characterization of candidate drugs, further improvements will be necessary, such as the generation of a multi-cell type lung cancer model and the lining of vascular structures with endothelial cells.
Collapse
Affiliation(s)
- Yikun Mei
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, TIB 4/3-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Dongwei Wu
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, TIB 4/3-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Johanna Berg
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, TIB 4/3-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Beatrice Tolksdorf
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, TIB 4/3-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Viola Roehrs
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, TIB 4/3-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Anke Kurreck
- BioNukleo GmbH, Ackerstr. 76, 13355 Berlin, Germany
| | - Thomas Hiller
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, TIB 4/3-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
- PRAMOMOLECULAR GmbH, Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Jens Kurreck
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, TIB 4/3-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| |
Collapse
|
25
|
Dong B, Zhang F, Zhang W, Gao Y. IncRNA EPB41L4A-AS1 Mitigates the Proliferation of Non-Small-Cell Lung Cancer Cells through the miR-105-5p/GIMAP6 Axis. Crit Rev Eukaryot Gene Expr 2023; 33:27-40. [PMID: 36734855 DOI: 10.1615/critreveukaryotgeneexpr.2022044323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Non-small-cell lung cancer (NSCLC) is the major subtype of lung cancer, with a series of long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and proteins involved in its pathogenesis. This study sought to investigate the functionality of lncRNA EPB41L4A antisense RNA 1 (lncRNA EPB41L4A-AS1) in the proliferation of NSCLC cells and provide a novel theoretical reference for NSCLC treatment. Levels of lncRNA EPB41L4A-AS1, miR-105-5p, and GTPase, IMAP family member 6 (GIMAP6) in tissues and cells were measured by RT-qPCR and the correlation between lncRNA EPB41L4A-AS1 and clinicopathological characteristics was analyzed. Cell proliferation was evaluated by cell counting kit-8 and colony formation assays. The subcellular localization of lncRNA EPB41L4A-AS1 was analyzed by the subcellular fractionation assay and the binding of miR-105-5p to lncRNA EPB41L4A-AS1 or GIMAP6 was analyzed by dual-luciferase and RNA pull-down assays. Functional rescue experiments were performed to analyze the role of miR-105-5p/GIMAP6 in NSCLC cell proliferation. lncRNA EPB41L4A-AS1 and GIMAP6 were downregulated while miR-105-5p was upregulated in NSCLC tissues and cells. lncRNA EPB41L4A-AS1 was correlated with tumor size and clinical staging and its overexpression reduced NSCLC cell proliferation. lncRNA EPB41L4A-AS1 was negatively correlated with miR-105-5p and positively correlated with GIMAP6 in NSCLC tissues, and lncRNA EPB41L4A-AS1 sponged miR-105-5p to promote GIMAP6 transcription in NSCLC cells. Overexpression of miR-105-5p or knockdown of GIMAP6 reversed the inhibition of lncRNA EPB41L4A-AS1 overexpression on NSCLC cell proliferation. lncRNA EPB41L4A-AS1 was downregulated in NSCLC and mitigated NSCLC cell proliferation through the miR-105-5p/GI-MAP6 axis.
Collapse
Affiliation(s)
- Bingwei Dong
- Department of Pathology, Xianyang Central Hospital, Xianyang City, 712000 Shaanxi Province, China
| | - Fenjuan Zhang
- Department of Pathology, Xianyang Central Hospital, Xianyang City, 712000 Shaanxi Province, China
| | - Weibo Zhang
- Department of Pathology, Xianyang Central Hospital, Xianyang City, 712000 Shaanxi Province, China
| | - Yingfang Gao
- Department of Pathology, Xianyang Central Hospital, Xianyang City, 712000 Shaanxi Province, China
| |
Collapse
|
26
|
Ni Y, Liu J, Zeng L, Yang Y, Liu L, Yao M, Chai L, Zhang L, Li Y, Zhang L, Li W. Natural product manoalide promotes EGFR-TKI sensitivity of lung cancer cells by KRAS-ERK pathway and mitochondrial Ca 2+ overload-induced ferroptosis. Front Pharmacol 2023; 13:1109822. [PMID: 36712673 PMCID: PMC9873971 DOI: 10.3389/fphar.2022.1109822] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 12/30/2022] [Indexed: 01/12/2023] Open
Abstract
Background: Manoalide (MA), a proven natural inhibitor of PLA2 has anticancer effects, but its potential application and mechanism as an anticancer drug to promote EGFR-TKI sensitivity in lung cancer cells have not been studied. Methods: KRAS-mutated lung cancer cells and organoids, acquired osimertinib-resistant lung cancer cell lines HCC827OR, were used as EGFR-TKI-resistant models. CCK-8, clone formation, apoptosis assays, and calcein-AM staining were performed to investigate the inhibitory effects of MA in lung cancer cells and organoids. The flow cytometry or confocal microscope was used to detect lipid droplets, ROS, lipid peroxidation, mitochondria Ca2+, and iron content. The oxygen consumption rate (OCR) and fatty acid oxidation (FAO) were used to estimate the effect of MA on mitochondrial function. Results: MA inhibits the proliferation of KRAS-mutated lung cancer cells and organoids. In addition, MA induces ER stress in a ROS-dependent mechanism. The ROS induced by MA is mainly in mitochondrial and causes lipid peroxidation, thereby inhibiting mitochondrial FAO metabolism and promoting the accumulation of lipid droplets. MA also suppresses the KRAS-ERK pathway through ROS and promotes the sensitivity of KRAS-mutated lung cancer cells and organoids to osimertinib. Furthermore, MA induces ferroptosis by suppressing the NRF2-SLC7A11 axis and mitochondrial Ca2+ overload induced-FTH1 pathways to promote the sensitivity of osimertinib-resistant lung cancer cells to osimertinib. Conclusions: MA is a candidate EGFR-TKI sensitizer in KRAS-mutated and osimertinib-resistant lung cancer cells.
Collapse
Affiliation(s)
- Yinyun Ni
- Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network (NHC Key Laboratory of Transplant Engineering and Immunology), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiaye Liu
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lingyan Zeng
- Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network (NHC Key Laboratory of Transplant Engineering and Immunology), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ying Yang
- Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network (NHC Key Laboratory of Transplant Engineering and Immunology), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lei Liu
- Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network (NHC Key Laboratory of Transplant Engineering and Immunology), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Menglin Yao
- Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network (NHC Key Laboratory of Transplant Engineering and Immunology), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Chai
- Institute of Core facility, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lu Zhang
- Institute of Core facility, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Li
- Institute of Core facility, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Zhang
- Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network (NHC Key Laboratory of Transplant Engineering and Immunology), West China Hospital, Sichuan University, Chengdu, Sichuan, China,*Correspondence: Li Zhang, ; Weimin Li,
| | - Weimin Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network (NHC Key Laboratory of Transplant Engineering and Immunology), West China Hospital, Sichuan University, Chengdu, Sichuan, China,Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China,*Correspondence: Li Zhang, ; Weimin Li,
| |
Collapse
|
27
|
Abstract
The therapeutic landscape of lung cancer treatment is changing rapidly, and new data was presented at the recently concluded American Society of Clinical Oncology 2022 (ASCO22) meeting. We highlight studies of clinical relevance that represent significant updates in the current management of non-small cell lung cancer (SCLC) and small cell lung cancer (NSCLC). We summarize the updates in early-stage NSCLC, mutated and non-mutated advanced NSCLC as well as small cell lung cancer (SCLC), and discuss these advances in the context of the current clinical standard of care.
Collapse
Affiliation(s)
- Fawzi Abu Rous
- Department of Internal Medicine, Division of Hematology and Oncology, Henry Ford Health System, Detroit, Michigan, USA
| | - Eric K Singhi
- Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Arthi Sridhar
- Division of Hematology and Oncology, University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| | - Muhammad Salman Faisal
- Fellow Department of Hematology and Medical Oncology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Aakash Desai
- Division of Medical Oncology, Mayo Clinic, Rochester, New York, USA
| |
Collapse
|
28
|
CircPIM3 regulates taxol resistance in non-small cell lung cancer via miR-338-3p/TNFAIP8 axis. Anticancer Drugs 2023; 34:115-125. [PMID: 36539365 DOI: 10.1097/cad.0000000000001347] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Numerous work has revealed the involvement of circular RNA (circRNA) in regulating chemotherapy resistance. Here, we investigate circPIM3 role in taxol (Tax) resistance in non-small cell lung cancer (NSCLC). CircPIM3, microRNA (miR)-338-3p and tumor necrosis factor-alpha-induced protein-8 (TNFAIP8) expression were detected via quantitative real-time PCR, western blot or immunohistochemistry assay. Tax resistance was evaluated using cell counting kit-8, cell proliferation was measured by colony formation assay, cell cycle and apoptosis were examined via flow cytometry. The interplay between miR-338-3p and circPIM3 or TNFAIP8 was confirmed by dual-luciferase reporter assay. Finally, the effect of circPIM3 on Tax resistance in NSCLC in vivo was investigated by xenograft models. CircPIM3 and TNFAIP8 were upregulated in Tax-resistant NSCLC tissue and cell samples. Reducing circPIM3 expression inhibited Tax resistance, proliferation and induced cycle arrest and apoptosis in Tax-resistant NSCLC cells. Mechanically, circPIM3 absence led to downregulation of TNFAIP8 via absorbing miR-338-3p. Additionally, circPIM3 depletion increased Tax sensitivity of NSCLC in vivo. Silencing of circPIM3 suppressed Tax resistance in Tax-resistant NSCLC cells through regulation of the miR-338-3p/TNFAIP8 axis.
Collapse
|
29
|
Xiao Y, Liu P, Wei J, Zhang X, Guo J, Lin Y. Recent progress in targeted therapy for non-small cell lung cancer. Front Pharmacol 2023; 14:1125547. [PMID: 36909198 PMCID: PMC9994183 DOI: 10.3389/fphar.2023.1125547] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/10/2023] [Indexed: 02/24/2023] Open
Abstract
The high morbidity and mortality of non-small cell lung cancer (NSCLC) have always been major threats to people's health. With the identification of carcinogenic drivers in non-small cell lung cancer and the clinical application of targeted drugs, the prognosis of non-small cell lung cancer patients has greatly improved. However, in a large number of non-small cell lung cancer cases, the carcinogenic driver is unknown. Identifying genetic alterations is critical for effective individualized therapy in NSCLC. Moreover, targeted drugs are difficult to apply in the clinic. Cancer drug resistance is an unavoidable obstacle limiting the efficacy and application of targeted drugs. This review describes the mechanisms of targeted-drug resistance and newly identified non-small cell lung cancer targets (e.g., KRAS G12C, NGRs, DDRs, CLIP1-LTK, PELP1, STK11/LKB1, NFE2L2/KEAP1, RICTOR, PTEN, RASGRF1, LINE-1, and SphK1). Research into these mechanisms and targets will drive individualized treatment of non-small cell lung cancer to generate better outcomes.
Collapse
Affiliation(s)
- Yanxia Xiao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China
| | - Pu Liu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China
| | - Jie Wei
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China
| | - Xin Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China
| | - Jun Guo
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China
| | - Yajun Lin
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China.,Peking University Fifth School of Clinical Medicine, Beijing, China
| |
Collapse
|
30
|
Zhu Y, Lin Z, Wu C. Apatinib plus Radiotherapy on the Expression of CEA and VEGF in Advanced Oligometastatic Non-Small-Cell Lung Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2023; 2023:4242346. [PMID: 37089715 PMCID: PMC10118890 DOI: 10.1155/2023/4242346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/07/2022] [Accepted: 06/17/2022] [Indexed: 04/25/2023]
Abstract
Objective The purpose of this study was to evaluate the clinical efficacy of apatinib plus concurrent radiotherapy on carcinoma embryonic antigen (CEA) and vascular endothelial growth factor (VEGF) expression in patients with non-small-cell lung cancer (NSCLC) with oligometastases. Methods This is a prospective randomized controlled trial. Sixty-four patients with oligometastatic NSCLC who were treated in the Central South University Xiangya School of Medicine Affiliated Haikou Hospital from January 2017 to January 2019 were randomly assigned into the control group and the study group, with 32 cases in each group. The control group was treated with stereotactic body radiotherapy (SBRT), and the study group was treated with apatinib. Results The overall response rate (ORR) of the study group was significantly higher than that of the control group. The carcinoma embryonic antigen (CEA) and the vascular endothelial growth factor (VEGF) in the two groups were significantly decreased, with lower results in the study group compared to the control group. The 12-month and 24-month overall survival (OS) of the study group were significantly higher than those of the control group. There was no significant difference in progression-free survival (PFS) between the two groups. The median OS in the control group was 20.0 months, and the study group had not yet reached the median OS; the OS in the study group was significantly higher than that in the control group. There was no significant difference in adverse reactions between the two groups. Conclusion For patients with oligometastatic lung cancer, apatinib combined with chemotherapy can significantly improve clinical efficacy, reduce tumor marker expression, and extend overall survival with good safety profiles.
Collapse
Affiliation(s)
- Yanxing Zhu
- Department of Oncology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Zhiren Lin
- Department of Tumor Radiotherapy, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Chengde Wu
- Department of Thoracic Surgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| |
Collapse
|
31
|
Nanoparticle-Mediated Delivery of STAT3 Inhibitors in the Treatment of Lung Cancer. Pharmaceutics 2022; 14:pharmaceutics14122787. [PMID: 36559280 PMCID: PMC9781630 DOI: 10.3390/pharmaceutics14122787] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Lung cancer is a common malignancy worldwide, with high morbidity and mortality. Signal transducer and activator of transcription 3 (STAT3) is an important transcription factor that not only regulates different hallmarks of cancer, such as tumorigenesis, cell proliferation, and metastasis but also regulates the occurrence and maintenance of cancer stem cells (CSCs). Abnormal STAT3 activity has been found in a variety of cancers, including lung cancer, and its phosphorylation level is associated with a poor prognosis of lung cancer. Therefore, the STAT3 pathway may represent a promising therapeutic target for the treatment of lung cancer. To date, various types of STAT3 inhibitors, including natural compounds, small molecules, and gene-based therapies, have been developed through direct and indirect strategies, although most of them are still in the preclinical or early clinical stages. One of the main obstacles to the development of STAT3 inhibitors is the lack of an effective targeted delivery system to improve their bioavailability and tumor targetability, failing to fully demonstrate their anti-tumor effects. In this review, we will summarize the recent advances in STAT3 targeting strategies, as well as the applications of nanoparticle-mediated targeted delivery of STAT3 inhibitors in the treatment of lung cancer.
Collapse
|
32
|
Zhao X, Liu C, He X, Wang M, Zhang H, Cheng J, Wang H. Laminin-bound integrin α6β4 promotes non-small cell lung cancer progression via the activation of YAP/TAZ signaling pathway. Front Oncol 2022; 12:1015709. [PMID: 36276068 PMCID: PMC9583390 DOI: 10.3389/fonc.2022.1015709] [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: 08/10/2022] [Accepted: 09/23/2022] [Indexed: 11/26/2022] Open
Abstract
Laminin is an extracellular matrix multidomain trimeric glycoprotein, that has a potential role in tumor progression. Here, we studied the effects of non-small cell lung cancer (NSCLC) cells interaction on laminin and explored the underlying mechanism of laminin associated NSCLC progression. Culture of A549 and NCI-1299 cells on 2D collagen gels (containing laminin) significantly promoted the proliferative and tumorigenic characteristics, as well as cell invasion of tumor cells in vitro. Consistently, comparing the clinical NSCLC tumor tissues, a poor overall survival was observed in patients with high laminin expression. Mechanistically, the expression of integrin α6β4 was required for the pro-tumor effects of laminin. Meanwhile, we showed that the downstream signaling of integrin α6β4, involved the focal adhesion kinase (FAK)/Yes-Associated Protein (YAP)/TAZ signaling pathway. The activation of FAK/YAP/TAZ signaling pathway induced by laminin was validated in tumor tissues from NSCLC patients. Suppression of integrin α6β4/FAK/YAP/TAZ signaling pathway efficiently suppressed the laminin-induced tumor growth, and strengthened the anticancer effects of chemotherapy, describing a novel target for NSCLC treatment.
Collapse
Affiliation(s)
- Xiaopeng Zhao
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chuang Liu
- Department of Thoracic Surgery, The Fourth Central Hospital of Baoding City, Baoding, China
| | - Xu He
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Miao Wang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Haoran Zhang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jingge Cheng
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hongyan Wang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Hongyan Wang,
| |
Collapse
|
33
|
Luo Y, Hu S, Wang F, Yang J, Gong D, Xu W, Xu X, Min L. miR-137 represses migration and cell motility by targeting COX-2 in non-small cell lung cancer. Transl Cancer Res 2022; 11:3803-3813. [PMID: 36388045 PMCID: PMC9641119 DOI: 10.21037/tcr-22-2177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/28/2022] [Indexed: 09/15/2023]
Abstract
BACKGROUND Lung cancer is a common malignant tumor, with, non-small cell lung cancer (NSCLC) accounting for about 80-85% of cases. This study investigated the expression of miR-137 in NSCLC tissues and cells and its effects on the migration and invasion of NSCLC cells and related mechanisms. METHODS We collected the neoplastic and paracancerous tissues of NSCLC patients, detected the expression of miR-137 in NSCLC tissues and cell lines by real-time quantitative polymerase chain reaction (RT-qPCR), and analyzed the correlation between miR-137 expression and the clinicopathological features and survival of NSCLC. Following transfection with miR-137 mimic or inhibitor in NSCLC cell lines (A549 or H1299) to upregulate or downregulate the expression of miR-137, transwell assay was employed to detect the effects of miR-137 on migration or invasion. Online software was employed to predict and analyze the target gene of miR-137, and luciferase reporter gene system was adopted to validate it. The effects of miR-137 on the expressions of COX-2 and Epithelial-Mesenchymal Transition (EMT) related proteins were investigated by Western blot. RESULTS Compared to paracancerous tissues and BEAS-2B cells, the expressions of miR-137 in NSCLC tissues, A549 and H1299 cells were dramatically down-regulated (P<0.01). After transfection with miR-137 mimic or inhibitor in A549 and H1299 cells, the miR-137 expressions were markedly up-regulated or down-regulated (P<0.01), respectively. The number of migrating or invading cells was observably decreased or increased (P<0.01) after transfected with mimic or inhibitor, respectively, while relative luciferase activity was evidently decreased in cells co-transfected with miR-137 mimic and wild type recombined vector of 3'UTR of COX-2. While the expressions of COX-2 and E-cadherin were both substantially reduced in A549 cells treated with miR-137 mimic, that of vimentin was substantially raised. The expression of miR-137 correlated with smoking history, lymph node metastasis, and TNM clinical stage, and patients with high miR-137 expression had apparent longer survival. CONCLUSIONS The expression of miR-137 was significantly down-regulated in NSCLC tissues and cells, and correlated with NSCLC progress. miR-137 suppressed the migration and invasion of NSCLC cells through regulating EMT relative proteins by targeting COX-2. miR-137 is expected to become a novel biomarker and therapeutic target of NSCLC.
Collapse
Affiliation(s)
- Yutu Luo
- Clinical Medical School of Yangzhou University, Subei People’s Hospital of Jiangsu Province, Yangzhou, China
- Department of Pulmonary and Critical Care Medicine, Affiliated Taizhou Second People’s Hospital, Medical School of Yangzhou University, Taizhou, China
| | - Suwei Hu
- Medical Genetic Center, Affiliated Yangzhou Women and Children Hospital, Medical School of Yangzhou University, Yangzhou, China
| | - Fang Wang
- Clinical Medical School of Yangzhou University, Subei People’s Hospital of Jiangsu Province, Yangzhou, China
| | - Junjun Yang
- Clinical Medical School of Yangzhou University, Subei People’s Hospital of Jiangsu Province, Yangzhou, China
| | - Daohui Gong
- Clinical Medical School of Yangzhou University, Subei People’s Hospital of Jiangsu Province, Yangzhou, China
| | - Wenjing Xu
- Clinical Medical School of Yangzhou University, Subei People’s Hospital of Jiangsu Province, Yangzhou, China
| | - Xingxiang Xu
- Clinical Medical School of Yangzhou University, Subei People’s Hospital of Jiangsu Province, Yangzhou, China
| | - Lingfeng Min
- Clinical Medical School of Yangzhou University, Subei People’s Hospital of Jiangsu Province, Yangzhou, China
| |
Collapse
|
34
|
Buszka K, Ntzifa A, Owecka B, Kamińska P, Kolecka-Bednarczyk A, Zabel M, Nowicki M, Lianidou E, Budna-Tukan J. Liquid Biopsy Analysis as a Tool for TKI-Based Treatment in Non-Small Cell Lung Cancer. Cells 2022; 11:2871. [PMID: 36139444 PMCID: PMC9497234 DOI: 10.3390/cells11182871] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 12/03/2022] Open
Abstract
The treatment of non-small cell lung cancer (NSCLC) has recently evolved with the introduction of targeted therapy based on the use of tyrosine kinase inhibitors (TKIs) in patients with certain gene alterations, including EGFR, ALK, ROS1, BRAF, and MET genes. Molecular targeted therapy based on TKIs has improved clinical outcomes in a large number of NSCLC patients with advanced disease, enabling significantly longer progression-free survival (PFS). Liquid biopsy is an increasingly popular diagnostic tool for treating TKI-based NSCLC. The studies presented in this article show that detection and analysis based on liquid biopsy elements such as circulating tumor cells (CTCs), cell-free DNA (cfDNA), exosomes, and/or tumor-educated platelets (TEPs) can contribute to the appropriate selection and monitoring of targeted therapy in NSCLC patients as complementary to invasive tissue biopsy. The detection of these elements, combined with their molecular analysis (using, e.g., digital PCR (dPCR), next generation sequencing (NGS), shallow whole genome sequencing (sWGS)), enables the detection of mutations, which are required for the TKI treatment. Despite such promising results obtained by many research teams, it is still necessary to carry out prospective studies on a larger group of patients in order to validate these methods before their application in clinical practice.
Collapse
Affiliation(s)
- Karolina Buszka
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
| | - Aliki Ntzifa
- Analysis of Circulating Tumor Cells Lab, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Barbara Owecka
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Paula Kamińska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
| | - Agata Kolecka-Bednarczyk
- Department of Immunology, Chair of Pathomorphology and Clinical Immunology, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Maciej Zabel
- Division of Anatomy and Histology, University of Zielona Góra, 65-046 Zielona Góra, Poland
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Evi Lianidou
- Analysis of Circulating Tumor Cells Lab, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Joanna Budna-Tukan
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| |
Collapse
|
35
|
Wang W, Sun H, Ma X, Zhu T, Zhang H. Circ_0002476 regulates cell growth, invasion, and mtDNA damage in non-small cell lung cancer by targeting miR-1182/TFAM axis. Thorac Cancer 2022; 13:2867-2878. [PMID: 36056804 PMCID: PMC9575079 DOI: 10.1111/1759-7714.14631] [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: 07/04/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Many circular RNAs (circRNAs) have been identified as potential targets for cancer therapy. However, the role of circ_0002476 in non-small cell lung cancer (NSCLC) progression has not been explored. METHODS The expression levels of circ_0002476, microRNA (miR)-1182, and mitochondrial transcription factor A (TFAM) were detected by quantitative real-time polymerase chain reaction. Cell functions were measured by cell counting kit 8 assay, EdU assay, colony formation assay, flow cytometry and transwell assay. Mitochondrial DNA (mtDNA) damage was assessed by measuring mtDNA copy number and transcript levels of ND1 and ATP6. Protein expression was examined by western blot. The interaction between miR-1182 and circ_0002476 or TFAM was detected by dual-luciferase reporter assay and RNA pull-down assay. Animal experiments were performed to explore circ_0002476 role in vivo. Exosomes (Exs) were extracted and identified by transmission electron microscopy and nanoparticle tracking analysis. RESULTS Circ_0002476 was overexpressed in NSCLC tissues and cells. Circ_0002476 knockdown suppressed NSCLC cell proliferation and invasion, while promoted apoptosis and mtDNA damage. Circ_0002476 could sponge miR-1182, and miR-1182 inhibitor reversed the influence induced by circ_0002476 knockdown. Moreover, TFAM was targeted by miR-1182, and miR-1182 hindered NSCLC cell progression by regulating TFAM. Additionally, circ_0002476 silencing could reduce NSCLC tumor growth by miR-1182/TFAM. Further analyzed showed that Exs were involved in the transport of circ_0002476 between cells. CONCLUSION Taken together, our findings suggested that circ_0002476 might be a potential molecular target for NSCLC treatment.
Collapse
Affiliation(s)
- Weijie Wang
- Department of Thoracic Surgery, The Affiliated Xiangshan Hospital of Wenzhou Medial University, Ningbo, China
| | - Haiting Sun
- Department of Thoracic Surgery, The Affiliated Xiangshan Hospital of Wenzhou Medial University, Ningbo, China
| | - Xuan Ma
- Department of Thoracic Surgery, The Affiliated Xiangshan Hospital of Wenzhou Medial University, Ningbo, China
| | - Ting Zhu
- Department of Thoracic Surgery, The Affiliated Xiangshan Hospital of Wenzhou Medial University, Ningbo, China
| | - Haina Zhang
- Department of Thoracic Surgery, The Affiliated Xiangshan Hospital of Wenzhou Medial University, Ningbo, China
| |
Collapse
|
36
|
Wang J, Yang Q, Tang M, Liu W. Validation and analysis of expression, prognosis and immune infiltration of WNT gene family in non-small cell lung cancer. Front Oncol 2022; 12:911316. [PMID: 35957916 PMCID: PMC9359207 DOI: 10.3389/fonc.2022.911316] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Early diagnosis and prognosis prediction of non-small cell lung cancer (NSCLC) have been challenging. Signaling cascades involving the Wingless-type (WNT) gene family play important biological roles and show prognostic value in various cancers, including NSCLC. On this basis, this study aimed to investigate the significance of WNTs in the prognosis and tumor immunity in NSCLC by comprehensive analysis. Expression and methylation levels of WNTs were obtained from the ONCOMINE, TIMER, and UALCAN. The dataset obtained from The Cancer Genome Atlas (TCGA) was utilized for prognostic analysis. cBioPortal was used to perform genetic alterations and correlation analysis of WNTs. R software was employed for functional enrichment and pathway analysis, partial statistics, and graph drawing. TRRUST was used to find key transcription factors. GEPIA was utilized for the analysis of expression, pathological staging, etc. Correlative analysis of immune infiltrates from TIMER. TISIDB was used for further immune infiltration validation analysis. Compared with that of normal tissues, WNT2/2B/3A/4/7A/9A/9B/11 expressions decreased, while WNT3/5B/6/7B/8B/10A/10B/16 expressions increased in lung adenocarcinoma (LUAD); WNT2/3A/7A/11 expressions were lessened, while WNT2B/3/5A/5B/6/7B/10A/10B/16 expressions were enhanced in squamous cell lung cancer (LUSC). Survival analysis revealed that highly expressed WNT2B and lowly expressed WNT7A predicted better prognostic outcomes in LUAD and LUSC. In the study of immune infiltration levels, WNT2, WNT9B, and WNT10A were positively correlated with six immune cells in LUAD; WNT1, WNT2, and WNT9B were positively correlated with six immune cells in LUSC, while WNT7B was negatively correlated. Our study indicated that WNT2B and WNT7A might have prognostic value in LUAD, and both of them might be important prognostic factors in LUSC and correlated to immune cell infiltration in LUAD and LUSC to a certain extent. Considering the prognostic value of WNT2B and WNT7A in NSCLC, we validated their mRNA and protein expression levels in NSCLC by performing qRT-PCR, western blot, and immunohistochemical staining on NSCLC pathological tissues and cell lines. This study may provide some direction for the subsequent exploration of the prognostic value of the WNTs and their role as biomarkers in NSCLC.
Collapse
Affiliation(s)
- Jianglin Wang
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Qingping Yang
- Department of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Mengjie Tang
- Department of Pathology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wei Liu
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Wei Liu,
| |
Collapse
|
37
|
Chen L, Wan X, Shan X, Zha W, Fan R. Smart PROTACs Enable Controllable Protein Degradation for Precision Cancer Therapy. Mol Diagn Ther 2022; 26:283-291. [PMID: 35471699 DOI: 10.1007/s40291-022-00586-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2022] [Indexed: 10/18/2022]
Abstract
Proteolysis-targeting chimeras (PROTACs) are heterobifunctional chemicals that degrade proteins at the post-translational level, which represent an emerging therapeutic modality to fight cancer and other diseases. Although several PROTACs have now entered clinical trials, potential off-tissue side effects have resulted from nonspecific accumulation at non-cancerous sites after systemic administration, and this remains a major challenge. To this end, in the past 3 years, activatable PROTACs whose activity can only be launched on demand have gained tremendous momentum. In this review, we provide an overview of these new smart activatable PROTACs, which exert protein degradation action only in response to internal or external stimuli. We categorize these activatable PROTACs according to their activation mechanism contributed by different stimuli, including reduction-activatable, hypoxia-activatable, and enzyme-activatable PROTACs and photo-caged or photo-switchable PROTACs. The use of stimuli-responsive chemical blocks in these activatable PROTACs allows local activation of the antitumor effects while reducing the incidence of off-site side effects for precision cancer therapy. The design principle and category of smart PROTACs are introduced along with an overview of their therapeutic prospects and challenges.
Collapse
Affiliation(s)
- Lixia Chen
- Medical College of Nantong University, Nantong, China
| | - Xinqiang Wan
- Department of Gynaecology and Obstetrics, The Fourth Affiliated Hospital of Nantong University, The First People's Hospital of Yancheng, Yancheng, China
| | - Xiangxiang Shan
- Department of Geraeology, Yancheng City No. 1 People's Hospital, Yancheng, China
| | - Wenzhang Zha
- Department of General Surgery, The Fourth Affiliated Hospital of Nantong University, The First People's Hospital of Yancheng, 166 Yulong Road, Yancheng, 224001, China
| | - Rengen Fan
- Department of General Surgery, The Fourth Affiliated Hospital of Nantong University, The First People's Hospital of Yancheng, 166 Yulong Road, Yancheng, 224001, China.
| |
Collapse
|
38
|
Wu J, He X, Xiong Z, Shi L, Chen D, Feng Y, Wen Q. Bruceine H Mediates EGFR-TKI Drug Persistence in NSCLC by Notch3-Dependent β-Catenin Activating FOXO3a Signaling. Front Oncol 2022; 12:855603. [PMID: 35463301 PMCID: PMC9024338 DOI: 10.3389/fonc.2022.855603] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/08/2022] [Indexed: 11/13/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) targeting epidermal growth factor receptor (EGFR) protein serve as a critical pillar in the treatment of non-small cell lung cancer (NSCLC), but resistance is universal. Identifying the potential key factors of drug resistance to EGFR-TKIs is essential to treat patients with EGFR mutant lung cancer. Our research here shows that bruceine H suppressed the proliferation, migration, and invasion of lung cancer cells; inhibited the growth of human NSCLC cell xenografts; and enhanced the therapeutic effects of gefitinib in the PC-9/GR xenograft models, possibly by inhibiting Notch3. In order to analyze the potential targets of the combination of Notch3 and EGFR-TKIs on resistance to EGFR, we analyzed the differences of gene expression between NSCLC tissues and EGFR-driven gefitinib-resistant tumoral groups and then identify through the WGCNA key genes that may provide therapeutic targets for TKI-resistant lung cancer xenograft models. We confirmed that EGFR-TKI in combination with Notch3 inhibitor can inhibit the expression of β-catenin and enhance the level of FOXO3a, leading to improved recurrence-free survival and overall survival of the xenotransplantation model. These results support that the combination of gefitinib and bruceine H may provide a promising alternative strategy for treating acquired EGFR-TKI resistance in patients with NSCLC.
Collapse
Affiliation(s)
- Jiahui Wu
- Pharmacy, Jiangxi University of Chinese Medicine, Jiangxi, China
| | - Xiao He
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi, China
| | - Ziwei Xiong
- Pharmacy, Jiangxi University of Chinese Medicine, Jiangxi, China
| | - Lingyu Shi
- Pharmacy, Jiangxi University of Chinese Medicine, Jiangxi, China
| | - Daofeng Chen
- Pharmacy, Fudan University of Pharmacy, Shanghai, China
| | - Yulin Feng
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi, China
| | - Quan Wen
- Pharmacy, Jiangxi University of Chinese Medicine, Jiangxi, China
| |
Collapse
|
39
|
Zhang Y, Hu K, Qu Z, Xie Z, Tian F. ADAMTS8 inhibited lung cancer progression through suppressing VEGFA. Biochem Biophys Res Commun 2022; 598:1-8. [PMID: 35149432 DOI: 10.1016/j.bbrc.2022.01.110] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 01/27/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND ADAMTS8 expression has been identified to be low in many cancers including lung cancer. However, the specific functions and regulatory system of ADAMTS8 remain to be unveiled. PURPOSE To study the potential modulatory mechanism of ADAMTS8 in lung cancer in cell and xenograft mice models. METHODS Differential expression of ADAMTS8 in lung cancer was analyzed on online tools. So was the overall survival curve in association with ADAMTS8/VEGFA expression in lung cancer patients. RT-qPCR was applied to validate the ADAMTS8 expression in lung cancer cell lines H460 and A549, with the normal lung epithelial cell Beas-2b as a control. Thereafter, overexpressed and knockdown plasmids were constructed for transfection. Colony and flow cytometry methods were used for cell proliferation and apoptosis. RT-qPCR and Western blot methods validated the changes in VEGFA after ADAMTS8 regulation in cells. Tube formation and Transwell methods were applied to observe the changes in tube formation and migration in HUVECs induced by tumor conditioned medium (TCM). Stable-transfected cells were injected subcutaneously into nude mice. H&E and Immunohistochemistry were applied to analyze the pathological differences and protein changes of ADAMTS8, VEGFA and CD31. RESULTS High ADAMTS8 was correlated with high overall survival rate in lung cancer patients. ADAMTS8 was also abnormally downregulated in NSCLC cells. Upregulation of ADAMTS8 suppressed cell proliferation and enhanced apoptosis while downregulation of ADAMTS8 promoted cell proliferation and decreased apoptosis. VEGFA was negatively correlated with ADAMTS8 in lung cancer tissues. Upregulation of ADAMTS8 inhibited VEGFA in mRNA and protein levels. Further, knockdown of ADAMTS8 induced tube formation and migration of HUVECs and upregulation of ADAMTS8 inhibited this. In addition, upregulation of ADAMTS8 in nude mice inhibited tumor growth and also suppressed VEGFA and CD31 in tumors. CONCLUSION ADAMTS8 inhibited lung cancer progression through suppressing VEGFA in lung cancer.
Collapse
Affiliation(s)
- Yutian Zhang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Kang Hu
- Department of Microbiological Testing, Center for Disease Control and Prevention of Nanchong City, Sichuan, PR China.
| | - Ziyi Qu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Zhihong Xie
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Fei Tian
- Department of Oncology, The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| |
Collapse
|
40
|
Ren H, Yang B, Li M, Lu C, Li X. RAB26 contributes to the progression of non-small cell lung cancer after being transcriptionally activated by SMAD3. Bioengineered 2022; 13:8064-8075. [PMID: 35291909 PMCID: PMC9161862 DOI: 10.1080/21655979.2022.2051853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for 85% of all cases of lung cancer, which constitutes the leading cause of cancer mortality. RAB26, a member of Rab GTPase superfamily, has been suggested to play a role in the tumorigenesis of NSCLC. The present work aimed to explore whether and how RAB26 contributed to the progression of NSCLC. NSCLC cell line A549 was transfection with short hairpin RNA (shRNA) or overexpression (Ov) vector to knockdown RAB26 or overexpress SMAD3, respectively. Then the malignant processes of A549 cells including proliferation, migration, invasion and apoptosis were evaluated by CCK-8, colony formation, wound-healing, transwell and TUNEL assays, respectively. Expression of proteins involved in these processes was measured by western blot. A549 xenograft mice model was established to confirm the effect of RAB26 silence on NSCLC progression in vivo. The relationship between RAB26 and SMAD3 was analyzed by bioinformatics and then verified by dual-luciferase reporter and chromatin immunoprecipitation (ChIP) assays. We found that silence of RAB26 inhibited the proliferation, migration and invasion but promoted apoptosis of A549 cells. In vivo studies revealed that the tumor growth of A549 xenograft was markedly suppressed upon RAB26 silence. Moreover, it was confirmed that SMAD3 bound to the promoter of RAB26 and enhance its expression. Finally, we observed that overexpression of SMAD3 significantly blocked the inhibitory effect of RAB26 silence on NSCLC progression. Collectively, RAB26 may contribute to the progression of NSCLC after being transcriptionally activated by SMAD3.
Collapse
Affiliation(s)
- Haixia Ren
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Bo Yang
- Department of Thoracic Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Mingjiang Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Chunling Lu
- Department of Pharmacy, Liaoyang, Liaoning, China
| | - Xiaoping Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
41
|
Liu WW, Hu J, Zhao Y, Wang R, Han Q, Rong XZ, Wang SY, Wang EH, Wu MX, Wang S, Liu Y. PTP-PEST Regulated Membranous/Cytoplasmic Translocation of p120ctn in the Lung Cancer Resistance to Tyrosine Kinase Inhibitor. Appl Immunohistochem Mol Morphol 2022; 30:215-224. [PMID: 35030104 DOI: 10.1097/pai.0000000000001008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 12/10/2021] [Indexed: 12/24/2022]
Abstract
Our previous studies indicate that resistance induction using first-generation tyrosine kinase inhibitors (TKIs) in lung cancer is accompanied with p120-catenin (p120ctn) cytoplasmic translocation from the membrane. However, the molecular mechanism underlying p120ctn intracytoplasmic translocation has not yet been reported. We performed immunohistochemistry to detect the correlation of p120ctn distribution with protein tyrosine phosphatase non-receptor type 12 (PTP-PEST) and p120ctn Y335 phosphorylation levels in non-small cell lung cancer (NSCLC) patients. After resistance induction using first-generation TKIs in lung cancer cells, Western blotting and substrate trapping were used to assess PTP-PEST expression and its influence on p120ctn Y335 phosphorylation, as well as the role of p120ctn Y335 phosphorylation on the association of p120ctn with E-cadherin and p120ctn membrane/cytoplasm translocation. In 197 samples collected from NSCLC patients, cytoplasmic p120ctn and enhanced p120ctn Y335 phosphorylation were associated with decreased PTP-PEST. After resistance induction using gefitinib, decreased PTP-PEST expression was accompanied by enhanced phosphorylation of p120ctn Y335 and p120ctn translocated to the cytoplasm. In gefitinib-resistant cells, PTP-PEST overexpression restrained p120ctn Y335 phosphorylation and restored membrane p120ctn expression. PTP-PEST enhanced the interaction of p120ctn with E-cadherin and elevated p120ctn membrane expression. However, increased p120ctn-Y335F mutant had no effect on p120ctn interaction with E-cadherin and membrane/cytoplasm translocation compared with the control group. In conclusion, resistance to first-generation TKIs inhibited PTP-PEST expression, which promoted p120ctn-Y335 phosphorylation and reduced the interaction of p120ctn with E-cadherin, resulting in p120ctn cytoplasmic translocation.
Collapse
Affiliation(s)
- Wei-Wei Liu
- Department of Anesthesiology, The First Hospital of China Medical University
| | - Jing Hu
- Sujia Tuo Town Community Health Service Center, Beijing, PR China
| | - Yue Zhao
- Department of Pathology, The First Hospital of China Medical University and College of Basic Medical Sciences, China Medical University
| | - Rui Wang
- Department of Pathology, The First Hospital of China Medical University and College of Basic Medical Sciences, China Medical University
| | - Qiang Han
- Department of Pathology, The First Hospital of China Medical University and College of Basic Medical Sciences, China Medical University
| | - Xue-Zhu Rong
- Department of Pathology, The First Hospital of China Medical University and College of Basic Medical Sciences, China Medical University
| | - Si-Yao Wang
- Department of Pathology, The First Hospital of China Medical University and College of Basic Medical Sciences, China Medical University
| | - En-Hua Wang
- Department of Pathology, The First Hospital of China Medical University and College of Basic Medical Sciences, China Medical University
| | - Mei-Xi Wu
- China Medical University-The Queen's University of Belfast Joint College, Shenyang
| | - Si Wang
- Department of Medical Microbiology and Human Parasitology, College of Basic Medical Sciences, China Medical University
| | - Yang Liu
- Department of Pathology, The First Hospital of China Medical University and College of Basic Medical Sciences, China Medical University
| |
Collapse
|
42
|
Deng Q, Wu M, Deng J. USP36 promotes tumor growth of non-small cell lung cancer via increasing KHK-A expression by regulating c-MYC-hnRNPH1/H2 axis. Hum Cell 2022; 35:694-704. [PMID: 35133629 DOI: 10.1007/s13577-022-00677-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/18/2022] [Indexed: 11/30/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the most common subtype of lung cancer with poor prognosis. This study designated to figure out the effects of Ubiquitin Specific Peptidase 36 (USP36) on NSCLC. Data of this study demonstrated that upregulation of USP36 was observed in NSCLC tissues and cell lines. Overexpression of USP36 promoted NSCLC cell proliferation and inhibited NSCLC cell apoptosis. Knockdown of USP36 decreased Ketohexokinase A (KHK-A) and increased KHK-C expression at both RNA and protein levels. Expression of c-MYC and hnRNPH1/H2 was positively correlated with the expression of USP36. Upregulation of c-MYC reversed the downregulation of hnRNPH1/H2 induced inhibition of USP36. Overexpression of hnRNPH1/H2 reversed the downregulation of KHK-A induced inhibition of USP36. Results of in vivo xenograft model were consistent with the findings of in vitro experiments. In summary, overexpression of USP36 in NSCLC accelerated tumor growth through upregulation of KHK-A, which was medicated by stabilizing c-MYC to increase hnRNPH1/H2 expression.
Collapse
Affiliation(s)
- Qian Deng
- Department of Palliative Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Maolin Wu
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, 610075, Sichuan Province, China.
| | - Jing Deng
- Department of Otolaryngology, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, 610075, Sichuan Province, China.
| |
Collapse
|
43
|
Yao Q, Li Z, Chen D. Review of LINC00707: A Novel LncRNA and Promising Biomarker for Human Diseases. Front Cell Dev Biol 2022; 10:813963. [PMID: 35155429 PMCID: PMC8826578 DOI: 10.3389/fcell.2022.813963] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/11/2022] [Indexed: 12/22/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are a major type of noncoding RNA greater than 200 nucleotides in length involved in important regulatory processes. Abnormal expression of certain lncRNAs contributes to the pathogenesis of multiple diseases, including cancers. The lncRNA LINC00707 is located on chromosome 10p14 and is abnormally expressed in numerous disease types, and particularly in several types of cancer. High LINC00707 levels mediate a series of biological functions, including cell proliferation, apoptosis, metastasis, invasion, cell cycle arrest, inflammation, and even osteogenic differentiation. In this review, we discuss the main functions and underlying mechanisms of LINC00707 in different diseases and describe promising applications of LINC00707 in clinical settings.
Collapse
Affiliation(s)
- Qinfan Yao
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Zheng Li
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Dajin Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
- National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- *Correspondence: Dajin Chen,
| |
Collapse
|
44
|
Chang MT, Shanahan F, Nguyen TTT, Staben ST, Gazzard L, Yamazoe S, Wertz IE, Piskol R, Yang YA, Modrusan Z, Haley B, Evangelista M, Malek S, Foster SA, Ye X. Identifying transcriptional programs underlying cancer drug response with TraCe-seq. Nat Biotechnol 2022; 40:86-93. [PMID: 34531539 DOI: 10.1038/s41587-021-01005-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
Genetic and non-genetic heterogeneity within cancer cell populations represent major challenges to anticancer therapies. We currently lack robust methods to determine how preexisting and adaptive features affect cellular responses to therapies. Here, by conducting clonal fitness mapping and transcriptional characterization using expressed barcodes and single-cell RNA sequencing (scRNA-seq), we have developed tracking differential clonal response by scRNA-seq (TraCe-seq). TraCe-seq is a method that captures at clonal resolution the origin, fate and differential early adaptive transcriptional programs of cells in a complex population in response to distinct treatments. We used TraCe-seq to benchmark how next-generation dual epidermal growth factor receptor (EGFR) inhibitor-degraders compare to standard EGFR kinase inhibitors in EGFR-mutant lung cancer cells. We identified a loss of antigrowth activity associated with targeted degradation of EGFR protein and an essential role of the endoplasmic reticulum (ER) protein processing pathway in anti-EGFR therapeutic efficacy. Our results suggest that targeted degradation is not always superior to enzymatic inhibition and establish TraCe-seq as an approach to study how preexisting transcriptional programs affect treatment responses.
Collapse
Affiliation(s)
- Matthew T Chang
- Department of Computational Biology and Bioinformatics, Genentech Inc., South San Francisco, CA, USA
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA
| | - Frances Shanahan
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA
| | - Thi Thu Thao Nguyen
- Department of Computational Biology and Bioinformatics, Genentech Inc., South San Francisco, CA, USA
| | - Steven T Staben
- Department of Discovery Chemistry, Genentech Inc., South San Francisco, CA, USA
| | - Lewis Gazzard
- Department of Discovery Chemistry, Genentech Inc., South San Francisco, CA, USA
| | - Sayumi Yamazoe
- Department of Discovery Chemistry, Genentech Inc., South San Francisco, CA, USA
- Discovery Biotherapeutics, Bristol-Myers Squibb, Redwood City, CA, USA
| | - Ingrid E Wertz
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA, USA
| | - Robert Piskol
- Department of Computational Biology and Bioinformatics, Genentech Inc., South San Francisco, CA, USA
| | - Yeqing Angela Yang
- Department of Microchemistry, Proteomics and Lipidomics, Genentech Inc., South San Francisco, CA, USA
| | - Zora Modrusan
- Department of Microchemistry, Proteomics and Lipidomics, Genentech Inc., South San Francisco, CA, USA
| | - Benjamin Haley
- Department of Molecular Biology, Genentech Inc., South San Francisco, CA, USA
| | - Marie Evangelista
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA
| | - Shiva Malek
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA
| | - Scott A Foster
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA.
| | - Xin Ye
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA.
| |
Collapse
|
45
|
Zarczynska I, Gorska-Arcisz M, Cortez AJ, Kujawa KA, Wilk AM, Skladanowski AC, Stanczak A, Skupinska M, Wieczorek M, Lisowska KM, Sadej R, Kitowska K. p38 Mediates Resistance to FGFR Inhibition in Non-Small Cell Lung Cancer. Cells 2021; 10:cells10123363. [PMID: 34943871 PMCID: PMC8699485 DOI: 10.3390/cells10123363] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 12/16/2022] Open
Abstract
FGFR signalling is one of the most prominent pathways involved in cell growth and development as well as cancer progression. FGFR1 amplification occurs in approximately 20% of all squamous cell lung carcinomas (SCC), a predominant subtype of non-small cell lung carcinoma (NSCLC), indicating FGFR as a potential target for the new anti-cancer treatment. However, acquired resistance to this type of therapies remains a serious clinical challenge. Here, we investigated the NSCLC cell lines response and potential mechanism of acquired resistance to novel selective FGFR inhibitor CPL304110. We found that despite significant genomic differences between CPL304110-sensitive cell lines, their resistant variants were characterised by upregulated p38 expression/phosphorylation, as well as enhanced expression of genes involved in MAPK signalling. We revealed that p38 inhibition restored sensitivity to CPL304110 in these cells. Moreover, the overexpression of this kinase in parental cells led to impaired response to FGFR inhibition, thus confirming that p38 MAPK is a driver of resistance to a novel FGFR inhibitor. Taken together, our results provide an insight into the potential direction for NSCLC targeted therapy.
Collapse
Affiliation(s)
- Izabela Zarczynska
- Department of Molecular Enzymology and Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland; (I.Z.); (M.G.-A.); (A.C.S.)
| | - Monika Gorska-Arcisz
- Department of Molecular Enzymology and Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland; (I.Z.); (M.G.-A.); (A.C.S.)
| | - Alexander Jorge Cortez
- Department of Biostatistics and Bioinformatics, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102 Gliwice, Poland; (A.J.C.); (A.M.W.)
| | - Katarzyna Aleksandra Kujawa
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102 Gliwice, Poland; (K.A.K.); (K.M.L.)
| | - Agata Małgorzata Wilk
- Department of Biostatistics and Bioinformatics, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102 Gliwice, Poland; (A.J.C.); (A.M.W.)
- Department of Systems Biology and Engineering, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Andrzej Cezary Skladanowski
- Department of Molecular Enzymology and Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland; (I.Z.); (M.G.-A.); (A.C.S.)
| | - Aleksandra Stanczak
- Clinical Development Department, Celon Pharma S.A., Marymoncka 15, 05-152 Kazuń Nowy, Poland; (A.S.); (M.W.)
| | - Monika Skupinska
- Preclinical Development Departament, Celon Pharma S.A., Marymoncka 15, 05-152 Kazuń Nowy, Poland;
| | - Maciej Wieczorek
- Clinical Development Department, Celon Pharma S.A., Marymoncka 15, 05-152 Kazuń Nowy, Poland; (A.S.); (M.W.)
| | - Katarzyna Marta Lisowska
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeze Armii Krajowej 15, 44-102 Gliwice, Poland; (K.A.K.); (K.M.L.)
| | - Rafal Sadej
- Department of Molecular Enzymology and Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland; (I.Z.); (M.G.-A.); (A.C.S.)
- Correspondence: (R.S.); (K.K.)
| | - Kamila Kitowska
- Department of Molecular Enzymology and Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland; (I.Z.); (M.G.-A.); (A.C.S.)
- Correspondence: (R.S.); (K.K.)
| |
Collapse
|
46
|
Multicenter phase II trial of nintedanib plus docetaxel in second-line treatment in advanced non-squamous non-small cell lung cancer patients refractory to first-line platin-based chemotherapy (REFRACT GFPC 02-15 study). Lung Cancer 2021; 161:122-127. [PMID: 34583220 DOI: 10.1016/j.lungcan.2021.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/02/2021] [Accepted: 09/14/2021] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Advanced non-squamous non-small cell lung cancer (NsqNSCLC) progressing at the induction of a first-line of platin-based chemotherapy is a subgroup of patients with poor prognosis and few second-line treatment options. MATERIALS AND METHODS This single-stage phase II prospective multicenter open-label trial performed in platin-based refractory (i.e. progressing during induction phase of first-line platin-based chemotherapy) advanced NsqNSCLC assessed the efficacy of the nintedanib-docetaxel combination in second-line treatment. The primary endpoint was progression-free survival (PFS) rates at 12 weeks with a cut-off at 30% for ineffectiveness and 50% for minimal efficacy. RESULTS A total of 59 patients from 23 centers were included (mean age, 58.5 years; male gender, 73.6%; performance status 0-1, 100%; former/current smokers, 92.5%; adenocarcinoma, 92.5%, median platin-based first-line chemotherapy, 2). Nintedanib-docetaxel combination was administered for a median of 4 cycles. The rate of PFS at 12 weeks was 39.6% (95% CI, 28.2-56.8). Median PFS was 2.7 (95% CI, 1.4-4.1) months and one-year PFS was 11.8% (95% CI, 4.8-22.2). Median overall survival (OS) was 6.9 (95% CI, 4.3-8.2) months and 12-month OS was 32.1% (95% CI, 19.8-45.0); 18-month OS was 27.6% (95% CI, 16,1-40.4). Twenty-nine (53.7%) patients reported at least one serious treatment-related adverse events leading to permanent discontinuation of at least one study drug in 12 (22.2%) patients. CONCLUSION The predefined minimal efficacy was not demonstrated. However, a number of NsqNSCLC patients refractory to first-line platin-based chemotherapy appeared to benefit from this combination.
Collapse
|
47
|
Hou D, Li W, Wang S, Huang Y, Wang J, Tang W, Zhou L, Qi L, Wu N, Zhao S. Different Clinicopathologic and Computed Tomography Imaging Characteristics of Primary and Acquired EGFR T790M Mutations in Patients with Non-Small-Cell Lung Cancer. Cancer Manag Res 2021; 13:6389-6401. [PMID: 34413682 PMCID: PMC8370596 DOI: 10.2147/cmar.s323972] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/02/2021] [Indexed: 12/16/2022] Open
Abstract
Purpose Although patients with primary and acquired epidermal growth factor receptor (EGFR) T790M positive non-small-cell lung cancer (NSCLC) respond to osimertinib treatment, the optimal treatment strategy differs for these two groups of patients. This study aimed to compare the clinicopathologic and computed tomography (CT) imaging characteristics between primary and acquired EGFR T790M mutations in patients with NSCLC before treatment. Patients and Methods We enrolled two groups of patients with primary or acquired EGFR T790M mutation NSCLC (n = 103 per group) from January 2012 to December 2019. We analyzed their clinicopathologic and CT characteristics and differences between the groups. The groups were further categorized based on 21L858R and 19del to exclude the effect of coexistent mutations. Results Primary, compared to acquired, T790M mutation tends to coexist with 21L858R (P < 0.001), exhibiting earlier tumor stage (P < 0.001), higher differentiation (P = 0.029), higher proportion of lepidic subtype adenocarcinoma (P < 0.001), and significant associations with some CT features (multiple primary lung cancers, ground-glass opacity, air bronchogram, and vacuole sign [all P < 0.001]). The combined model, composed of clinicopathologic and conventional CT signature and CT-radiomic signature, showed good discriminative ability with the area under the receiver operating characteristic curve 0.90 and 0.91 in the training and validation datasets, respectively. The T790M mutation contributed to these differences independently of coexistent mutations. Conclusion We identified clinicopathologic and CT imaging differences between primary and acquired T790M mutations. These findings provide insights into developing future personalized T790M mutation status-based theranostic strategies.
Collapse
Affiliation(s)
- Donghui Hou
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Weihua Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Sicong Wang
- GE Healthcare, Life Sciences, Beijing, People's Republic of China
| | - Yao Huang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Jianwei Wang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Wei Tang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Lina Zhou
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Linlin Qi
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Ning Wu
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.,PET-CT Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Shijun Zhao
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
48
|
Testing EGFR with Idylla on Cytological Specimens of Lung Cancer: A Review. Int J Mol Sci 2021; 22:ijms22094852. [PMID: 34063720 PMCID: PMC8125729 DOI: 10.3390/ijms22094852] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/30/2021] [Accepted: 04/30/2021] [Indexed: 12/14/2022] Open
Abstract
The current standard of care for advanced non-small-cell lung cancer is based on detecting actionable mutations that can benefit from targeted therapy. Comprehensive genetic tests can have long turn-around times, and because EGFR mutations are the most prevalent actionable mutation, a quick detection would enable a prompt initiation of targeted therapy. Furthermore, the scarcity of diagnostic material means that sometimes only cytologic material is available. The Idylla™ EGFR assay is a real-time PCR–based method able to detect 51 EGFR mutations in 2.5 h. Idylla is validated for use only on FFPE sections, but some researchers described their experiences with cytological material. We reviewed the relevant literature, finding four articles describing 471 cases and many types of cytological input material: smears, cell-block sections, suspensions, and extracted DNA. The sensitivity, specificity, and limit of detection appear comparable to those obtained with histological input material, with one exception: the usage of scraped stained smears as input may reduce the accuracy of the test. In conclusion, usage of cytological material as input to the Idylla EGFR test is possible. A workflow where common mutations are tested first and fast, leaving rarer mutations for subsequent comprehensive profiling, seems the most effective approach.
Collapse
|
49
|
Identification of transcriptional subtypes in lung adenocarcinoma and squamous cell carcinoma through integrative analysis of microarray and RNA sequencing data. Sci Rep 2021; 11:8709. [PMID: 33888829 PMCID: PMC8062554 DOI: 10.1038/s41598-021-88209-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/08/2021] [Indexed: 02/02/2023] Open
Abstract
Classification of tumors into subtypes can inform personalized approaches to treatment including the choice of targeted therapies. The two most common lung cancer histological subtypes, lung adenocarcinoma and lung squamous cell carcinoma, have been previously divided into transcriptional subtypes using microarray data, and corresponding signatures were subsequently used to classify RNA-seq data. Cross-platform unsupervised classification facilitates the identification of robust transcriptional subtypes by combining vast amounts of publicly available microarray and RNA-seq data. However, cross-platform classification is challenging because of intrinsic differences in data generated using the two gene expression profiling technologies. In this report, we show that robust gene expression subtypes can be identified in integrated data representing over 3500 normal and tumor lung samples profiled using two widely used platforms, Affymetrix HG-U133 Plus 2.0 Array and Illumina HiSeq RNA sequencing. We tested and analyzed consensus clustering for 384 combinations of data processing methods. The agreement between subtypes identified in single-platform and cross-platform normalized data was then evaluated using a variety of statistics. Results show that unsupervised learning can be achieved with combined microarray and RNA-seq data using selected preprocessing, cross-platform normalization, and unsupervised feature selection methods. Our analysis confirmed three lung adenocarcinoma transcriptional subtypes, but only two consistent subtypes in squamous cell carcinoma, as opposed to four subtypes previously identified. Further analysis showed that tumor subtypes were associated with distinct patterns of genomic alterations in genes coding for therapeutic targets. Importantly, by integrating quantitative proteomics data, we were able to identify tumor subtype biomarkers that effectively classify samples on the basis of both gene and protein expression. This study provides the basis for further integrative data analysis across gene and protein expression profiling platforms.
Collapse
|
50
|
Gao R, Zhang Y, Hou W, Li J, Zhu G, Zhang X, Xu B, Wu Z, Wang H. Combination of first-line chemotherapy with Kanglaite injections versus first-line chemotherapy alone for advanced non-small-cell lung cancer: study protocol for an investigator-initiated, multicenter, open-label, randomized controlled trial. Trials 2021; 22:214. [PMID: 33731199 PMCID: PMC7966914 DOI: 10.1186/s13063-021-05169-w] [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: 06/14/2020] [Accepted: 03/04/2021] [Indexed: 12/24/2022] Open
Abstract
Background Non-small-cell lung cancer (NSCLC) is usually diagnosed at an advanced stage, and chemotherapy is the main treatment for this disease. Kanglaite injections (KLTi) have been widely used for the treatment of cancer in China. KLTi combined with chemotherapy could improve the short-term efficacy, quality of life, and performance status for NSCLC compared with chemotherapy alone. This trial aims to assess the long-term efficacy and safety of KLTi in combination with chemotherapy for the treatment of advanced NSCLC. Methods This will be an investigator-initiated multicenter open-label randomized controlled trial. We will randomly assign 334 eligible participants with stage IIIA-IV NSCLC to the treatment or control groups in a 1:1 ratio. Patients in both groups will be administered 4–6 cycles of first-line platinum-based double chemotherapy regimens. Patients with complete response, partial response, or stable disease after 4–6 cycles will receive non-platinum single-agent chemotherapy. Patients in the treatment group are to receive intravenous KLTi 200 ml per day continuously for 14 days, commencing on the first day of chemotherapy. The treatment will be discontinued at the time of disease progression or until unacceptable toxicity is noted. The follow-up will be conducted every 2 months until death, loss of follow-up, or 12 months from randomized enrollment. The primary outcome will be progression-free survival (PFS). The secondary outcomes will be the objective response rate, 1-year survival rate, quality of life, living ability, and blood lipids. The safety outcome will be the rate of adverse events. Discussion This study will be the first randomized controlled trial in which PFS is used as the primary outcome to test whether KLTi combined with first-line chemotherapy has superior efficacy and reduced toxicity compared to chemotherapy alone in advanced NSCLC. This will also be the first clinical study to observe the effects of KLTi on blood lipids. Trial registration ClinicalTrials.gov NCT03986528. Prospectively registered on 30 May 2019.
Collapse
Affiliation(s)
- Ruike Gao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wei Hou
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Guanghui Zhu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoxiao Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,China Academy of Chinese Medical Sciences, Beijing, China
| | - Bowen Xu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Zhe Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,China Academy of Chinese Medical Sciences, Beijing, China
| | - Heping Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|