1
|
Xie C, Chen J, Yang S, Ye F, Lin Z, Xu Y, Yang Y, Tong L. Risk Factors for Prognosis of Lung Cancer Patients Receiving Anlotinib Treatment: A Retrospective Cohort Study. THE CLINICAL RESPIRATORY JOURNAL 2025; 19:e70051. [PMID: 39924314 PMCID: PMC11807704 DOI: 10.1111/crj.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 10/30/2024] [Accepted: 01/29/2025] [Indexed: 02/11/2025]
Abstract
PURPOSE Anlotinib is widely used in the treatment of lung cancer. However, there remains a lack of predictive biomarkers to effectively gauge the response to anlotinib therapy. We conducted a retrospective study to preliminarily explore potential risk factors that might predict outcomes in lung cancer patients undergoing anlotinib treatment. PATIENTS AND METHODS We retrospectively analyzed lung cancer patients treated with anlotinib at our hospital between 1 June 2018 and 1 June 2021. Data were gathered from electronic medical records. Demographic and clinical characteristics of patients, progression-free survival (PFS), and overall survival (OS) were described. Predictive factors related to treatment efficacy were preliminarily analyzed using Cox regression and Kaplan-Meier survival analyses. RESULTS After adjusting for potential confounders, clinical stage IV (hazard ratio [HR] = 2.52, 95% confidence interval [CI], 1.09-5.82, p = 0.0311), N-terminal fragment brain natriuretic peptides (NT-pro-BNP) > 300 pg/mL (HR = 2.54, 95% CI, 1.17-5.52, p = 0.0183), and neuron-specific enolase (NSE) > 16.3 ng/mL (HR = 1.70, 95% CI, 1.03-2.81, p = 0.0389) were associated with shorter OS, whereas age (HR = 0.96, 95% CI, 0.94-0.99, p = 0.0055) was associated with a longer PFS in fully adjusted model. Kaplan-Meier analyses of cumulative risk factors (clinical stage IV, NT-pro-BNP > 300 pg/mL, and NSE > 16.3 ng/mL) indicated that patients with a greater number of coexisting risk factors had significantly shorter OS (p < 0.0001). CONCLUSION Clinical stage IV, NT-pro-BNP level, and NSE level were identified as independent prognostic factors for lung cancer patients undergoing anlotinib treatment. Patients with multiple high-risk factors may derive limited benefit from anlotinib.
Collapse
Affiliation(s)
- Congyi Xie
- Department of Pulmonary MedicineZhongshan Hospital (Xiamen), Fudan UniversityXiamenFujianPeople's Republic of China
- Xiamen Clinical Research Center for Cancer TherapyXiamenFujianPeople's Republic of China
| | - Jinzhan Chen
- Department of Pulmonary MedicineZhongshan Hospital (Xiamen), Fudan UniversityXiamenFujianPeople's Republic of China
| | - Shuwen Yang
- Department of Pulmonary MedicineZhongshan Hospital (Xiamen), Fudan UniversityXiamenFujianPeople's Republic of China
| | - Feiyang Ye
- College of Computer and Data ScienceFuzhou UniversityFuzhouFujianPeople's Republic of China
| | - Zhenyang Lin
- Department of Thoracic SurgeryZhongshan Hospital (Xiamen), Fudan UniversityXiamenFujianPeople's Republic of China
| | - Yijiao Xu
- Department of Pulmonary MedicineZhongshan Hospital (Xiamen), Fudan UniversityXiamenFujianPeople's Republic of China
| | - Yimin Yang
- Department of Vascular SurgeryZhongshan Hospital, Fudan UniversityShanghaiPeople's Republic of China
| | - Lin Tong
- Department of Pulmonary MedicineZhongshan Hospital (Xiamen), Fudan UniversityXiamenFujianPeople's Republic of China
- Department of Pulmonary and Critical Care MedicineZhongshan Hospital, Fudan UniversityShanghaiPeople's Republic of China
| |
Collapse
|
2
|
Evyapan G, Senturk NC, Celik IS. Ornidazole Inhibits the Angiogenesis and Migration Abilities of Non-small Cell Lung Cancer (NSCLC) via Downregulation of VEGFA/VEGFR2/NRP-1 and PI3K/AKT/mTOR Pathways. Cell Biochem Biophys 2024; 82:3277-3285. [PMID: 38886281 DOI: 10.1007/s12013-024-01358-x] [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] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Around the world, non-small cell lung cancer (NSCLC) is the leading cause of cancer-related deaths among all cancers. Despite advancements in new therapeutic approaches over the past few decades, the five-year survival rate still remains disappointing. The lack of effective anti-angiogenic and anti-migration drugs is the biggest obstacle to the treatment of metastatic lung cancer. Therefore, there is a need to develop new and effective therapeutic compounds targeting anti-angiogenic and anti-migration pathways for the treatment of lung cancer. Ornidazole is a nitroimidazole agent widely used in the treatment of parasitic infections such as trichomonas vaginalis, amebiasis and giardiasis. This study aimed to investigate the anti-proliferative, anti-angiogenic and anti-mitotic activities of the anti-parasitic drug Ornidazole in two human lung cancer cell lines (A549, H1299). METHODS We determined the effects of Ornidazole, on cell viability, apoptosis, migration, angiogenesis and metastatic ability against NSCLC in lung cancer cell lines. Its action on the mRNA and protein expression levels of VEGFA, VEGFR2, NRP1, Casp9, Casp3, Bax, Bcl-2, PIK3CA, AKT, MTOR, PTEN and FOX3A was assessed. Furthermore, in this study the effects on cell migration, cell viability and proliferation was evaluated through wound healing, MTT and Crystal violet assays. RESULTS This study demonstrated that Ornidazole effectively reduces cell viability and migration ability, inhibits angiogenesis and metastatic abilities in NSCLC cells. CONCLUSIONS In conclusion, these results may shed light on the treatment of NSCLC, and we suggest the anti-parasitic drug Ornidazole as a new agent with potential anti-angiogenic and anti-mitotic activity by interfering with the molecular pathways that trigger tumor angiogenesis and migration.
Collapse
Affiliation(s)
- Gulsah Evyapan
- Department of Medical Biology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey.
| | - Nesrin Cetinel Senturk
- Department of Medical Biology, Faculty of Medicine, Cukurova University, Balcali- Adana, Turkey
| | - Ibrahim Seyfettin Celik
- Department Of Medical Services And Techniques, Kahramanmaraş Health Services Vocational School, Pathology Laboratory Techniques Pr., Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| |
Collapse
|
3
|
Kim B, Park YY, Lee JH. CXCL10 promotes melanoma angiogenesis and tumor growth. Anim Cells Syst (Seoul) 2024; 28:453-465. [PMID: 39268223 PMCID: PMC11391877 DOI: 10.1080/19768354.2024.2402024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/24/2024] [Accepted: 09/01/2024] [Indexed: 09/15/2024] Open
Abstract
Upregulation of CXC motif chemokine 10 (CXCL10) in melanoma patients has been found to be associated with melanoma progression. However, the role of endogenous CXCL10 from the host in melanoma tumor growth remains unclear. In the present study, we found that host-derived endogenous CXCL10 production was dramatically augmented during subcutaneous B16F10 melanoma tumor growth and that host ablation of CXCL10 in Cxcl10-/- mice showed a decrease in both angiogenesis and tumor growth of B16F10 melanoma in vivo. Several signaling pathways involved in production of pro-angiogenic factors and tumor growth were activated by CXCL10 in B16F10 melanoma cells. CXCL10 increased expression of pro-angiogenic factors, such as vascular endothelial growth factor (VEGF), platelet-derived growth factor subunit-B (PDGF-B), fibroblast growth factor 2 (FGF2), hepatocyte growth factor (HGF), and angiopoietin 2 (Angpt2), in B16F10 melanoma cells, resulting in enhanced tube formation and proliferation of human umbilical vein endothelial cells in vitro. In addition, CXCL10 directly enhanced B16F10 melanoma tumor growth in an in vitro three-dimensional cell culture system. Together, our findings reveal that amplified host-derived endogenous CXCL10 is critical for B16F10 melanoma angiogenesis and tumor growth. Therefore, CXCL10 might represent a therapeutic target for melanoma.
Collapse
Affiliation(s)
- Bongjun Kim
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yun-Yong Park
- Department of life Science, Chung-Ang University, Seoul, Republic of Korea
| | - Jong-Ho Lee
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, Republic of Korea
- Department of Biomedical Sciences, Dong-A University, Busan, Republic of Korea
| |
Collapse
|
4
|
Wu Z, Zhou P, Zhao Y, Wang J, Gao S. Efficacy and safety of anlotinib in combination with immune checkpoint inhibitors or not as advanced non-small cell lung cancer treatment: a systematic review and network meta-analysis. Transl Cancer Res 2024; 13:2451-2463. [PMID: 38881944 PMCID: PMC11170544 DOI: 10.21037/tcr-23-1483] [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: 11/13/2023] [Accepted: 03/25/2024] [Indexed: 06/18/2024]
Abstract
Background Non-small cell lung cancer (NSCLC) remains a leading cause of cancer mortality. Combined anlotinib and immune checkpoint inhibitors (ICIs) therapy may have synergistic antitumor effects in NSCLC. This study aimed to comparing the efficacy and safety of anlotinib and ICIs treatment, monotherapy and combination in NSCLC. Methods We performed a systematic review and network meta-analysis of 14 studies involving 4,308 NSCLC patients across four regimens: anlotinib, ICIs, anlotinib plus ICIs, and placebo. Efficacy outcomes were progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and disease control rate (DCR). Safety outcomes included treatment-related adverse events (TRAEs), TRAE grade three or higher (TRAE ≥3). Analyses were performed in RevMan 5.3 and R 3.5.1 (gemtc package). P<0.05 or effect estimate with 95% confidence interval (CI) that did not include 1 indicated statistical significance. Results Fourteen publications involving 4,308 patients across four treatment regimens (anlotinib, ICIs, anlotinib plus ICIs, placebo) were included. For PFS, network meta-analysis showed all three interventions significantly improved PFS versus placebo. Anlotinib plus ICIs demonstrated the greatest PFS improvement [hazard ratio (HR) =0.24; 95% CI: 0.14, 0.36], followed by anlotinib (HR =0.37; 95% CI: 0.23, 0.58), and ICIs (HR =0.43; 95% CI: 0.27, 0.67). For OS, compared to placebo, anlotinib plus ICIs showed the greatest OS improvement (HR =0.52; 95% CI: 0.33, 0.74), followed by anlotinib (HR =0.66; 95% CI: 0.47, 0.95), and ICIs (HR =0.72; 95% CI: 0.54, 0.97). For ORR, anlotinib plus ICIs demonstrated the greatest improvement versus placebo [odds ratio (OR) =5.29; 95% CI: 3.32, 8.58], followed by anlotinib (OR =4.38; 95% CI: 2.42, 8.19), and ICIs (OR =2.17; 95% CI: 1.65, 2.89). For DCR, anlotinib plus ICIs showed the greatest improvement versus placebo (OR =13.32; 95% CI: 4.99, 45.09), followed by anlotinib (OR =5.56; 95% CI: 2.17, 14.38), and ICIs (OR =3.46; 95% CI: 1.29, 10.85). Compared to placebo, anlotinib was associated with the highest risk of TRAEs (OR =3.67, 95% CI: 1.12, 15.77), followed by ICIs (OR =1.83; 95% CI: 1.26, 2.69). Due to lack of data on anlotinib plus ICIs, no comparison was conducted. For grade ≥3 TRAEs, compared to placebo, anlotinib increased the risk (OR =3.67; 95% CI: 1.12, 15.77), while anlotinib plus ICIs (OR =2.45; 95% CI: 0.51, 11.6) and ICIs (OR =1.29; 95% CI: 0.33, 4.38) did not increase the risk. Conclusions Anlotinib combined with ICIs demonstrates improved efficacy over monotherapy for NSCLC treatment, without increased adverse events.
Collapse
Affiliation(s)
- Zhengyu Wu
- Clinical Research Center, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
| | - Peng Zhou
- Clinical Research Center, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
| | - Yanan Zhao
- Clinical Research Center, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
| | - Junping Wang
- Clinical Research Center, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
| | - Shan Gao
- College of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
5
|
Dou XJ, Ma RY, Ren DW, Liu Q, Yan P. Effectiveness and Safety of Anlotinib Combined with PD-1 Blockades in Patients with Previously Immunotherapy Treated Advanced Non-Small Cell Lung Cancer: A Retrospective Exploratory Study. LUNG CANCER (AUCKLAND, N.Z.) 2024; 15:29-40. [PMID: 38560413 PMCID: PMC10979677 DOI: 10.2147/lctt.s444884] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 02/18/2024] [Indexed: 04/04/2024]
Abstract
Objective This study aimed to investigate the effectiveness and tolerability of anlotinib plus PD-1 blockades in patients with previously immunotherapy treated advanced non-small-cell lung cancer (NSCLC). Methods A total of 67 patients with previously immunotherapy treated advanced NSCLC who received anlotinib plus PD-1 blockades in clinical practice were screened retrospectively. All the PD-1 blockades used in this study were approved in China and consisted of sintilimab, camrelizumab, tislelizumab and pembrolizumab. Effectiveness and safety of anlotinib plus PD-1 blockades were assessed, and all patients were followed up regularly. Clinical significance between response status to previous immune-related treatment regimens and therapeutic outcomes of anlotinib plus PD-1 blockades was further explored. Results The best overall response among the 67 patients suggested that a partial response was observed in 16 patients, stable disease was noted in 41 patients and progressive disease was found in 10 patients, which yielded an objective response rate of 23.9% (95% CI: 14.3-35.9%) and a disease control rate of 85.1% (95% CI: 74.3-92.6%). Prognostic outcomes indicated that the median progression-free survival (PFS) was 6.1 months (95% CI: 2.37-9.83) and the median overall survival (OS) was 16.5 months (95% CI: 10.73-22.27). Exploratory analysis highlighted that patients who were intolerant to previous immune-related regimens (17 patients) might have a superior prognosis (median OS: 22.3 months vs 12.5 months, P=0.024). Additionally, adverse reactions with any grades during anlotinib plus PD-1 blockades administration were observed in 62 patients (92.5%), of which 31 patients (46.3%) had ≥grade 3 adverse reactions. Most common adverse reactions were fatigue, hypertension, diarrhea and hepatotoxicity. Conclusion Anlotinib plus PD-1 blockades demonstrated promising effectiveness and tolerable safety in patients with previously immunotherapy treated advanced NSCLC. Those who were intolerant to previous immune-related regimens might benefit significantly from treatment with anlotinib plus PD-1 blockades. This conclusion should be confirmed in future studies.
Collapse
Affiliation(s)
- Xue-Jun Dou
- Department of Thoracic Surgery, Aerospace Center Hospital, Beijing, 100049, People’s Republic of China
| | - Run-Yang Ma
- Department of Thoracic Surgery, Aerospace Center Hospital, Beijing, 100049, People’s Republic of China
| | - De-Wang Ren
- Department of Thoracic Surgery, Aerospace Center Hospital, Beijing, 100049, People’s Republic of China
| | - Qiang Liu
- Department of Thoracic Surgery, Peking University International Hospital, Beijing, 102206, People’s Republic of China
| | - Peng Yan
- Department of Respiratory Medicine, China Aerospace Science & Industry Corporation 731 Hospital, Beijing, 100071, People’s Republic of China
| |
Collapse
|
6
|
Lei X, Li Z, Huang M, Huang L, Huang Y, Lv S, Zhang W, Chen Z, Ke Y, Li S, Chen J, Yang X, Deng Q, Liu J, Yu X. Gli1-mediated tumor cell-derived bFGF promotes tumor angiogenesis and pericyte coverage in non-small cell lung cancer. J Exp Clin Cancer Res 2024; 43:83. [PMID: 38493151 PMCID: PMC10944600 DOI: 10.1186/s13046-024-03003-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 03/04/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Tumor angiogenesis inhibitors have been applied for non-small cell lung cancer (NSCLC) therapy. However, the drug resistance hinders their further development. Intercellular crosstalk between lung cancer cells and vascular cells was crucial for anti-angiogenenic resistance (AAD). However, the understanding of this crosstalk is still rudimentary. Our previous study showed that Glioma-associated oncogene 1 (Gli1) is a driver of NSCLC metastasis, but its role in lung cancer cell-vascular cell crosstalk remains unclear. METHODS Conditioned medium (CM) from Gli1-overexpressing or Gli1-knockdown NSCLC cells was used to educate endothelia cells and pericytes, and the effects of these media on angiogenesis and the maturation of new blood vessels were evaluated via wound healing assays, Transwell migration and invasion assays, tube formation assays and 3D coculture assays. The xenograft model was conducted to establish the effect of Gli1 on tumor angiogenesis and growth. Angiogenic antibody microarray analysis, ELISA, luciferase reporte, chromatin immunoprecipitation (ChIP), bFGF protein stability and ubiquitination assay were performed to explore how Gli1 regulate bFGF expression. RESULTS Gli1 overexpression in NSCLC cells enhanced the endothelial cell and pericyte motility required for angiogenesis required for angiogenesis. However, Gli1 knockout in NSCLC cells had opposite effect on this process. bFGF was critical for the enhancement effect on tumor angiogenesis. bFGF treatment reversed the Gli1 knockdown-mediated inhibition of angiogenesis. Mechanistically, Gli1 increased the bFGF protein level by promoting bFGF transcriptional activity and protein stability. Importantly, suppressing Gli1 with GANT-61 obviously inhibited angiogenesis. CONCLUSION The Gli1-bFGF axis is crucial for the crosstalk between lung cancer cells and vascular cells. Targeting Gli1 is a potential therapeutic approach for NSCLC angiogenesis.
Collapse
Affiliation(s)
- Xueping Lei
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences &The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Zhan Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences &The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Manting Huang
- Zhongshan Hospital of Traditional Chinese Medicine, Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, 528400, PR, China
| | - Lijuan Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences &The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Yong Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences &The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Sha Lv
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences &The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Weisong Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences &The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Zhuowen Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences &The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Yuanyu Ke
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences &The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Songpei Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences &The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Jingfei Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences &The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Xiangyu Yang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences &The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Qiudi Deng
- GMU-GIBH Joint School of Life Sciences, Joint Laboratory for Cell Fate Regulation and Diseases, The Guangdong-Hong Kong-Macau, Guangzhou Medical University, Guangzhou, 511436, PR, China.
| | - Junshan Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China.
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, People's Republic of China.
| | - Xiyong Yu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences &The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China.
| |
Collapse
|
7
|
Arunrungvichian K, Vajragupta O, Hayakawa Y, Pongrakhananon V. Targeting Alpha7 Nicotinic Acetylcholine Receptors in Lung Cancer: Insights, Challenges, and Therapeutic Strategies. ACS Pharmacol Transl Sci 2024; 7:28-41. [PMID: 38230275 PMCID: PMC10789132 DOI: 10.1021/acsptsci.3c00138] [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: 07/04/2023] [Revised: 11/26/2023] [Accepted: 12/01/2023] [Indexed: 01/18/2024]
Abstract
Alpha7 nicotinic acetylcholine receptor (α7 nAChR) is an ion-gated calcium channel that plays a significant role in various aspects of cancer pathogenesis, particularly in lung cancer. Preclinical studies have elucidated the molecular mechanism underlying α7 nAChR-associated lung cancer proliferation, chemotherapy resistance, and metastasis. Understanding and targeting this mechanism are crucial for developing therapeutic interventions aimed at disrupting α7 nAChR-mediated cancer progression and improving treatment outcomes. Drug research and discovery have determined natural compounds and synthesized chemical antagonists that specifically target α7 nAChR. However, approved α7 nAChR antagonists for clinical use are lacking, primarily due to challenges related to achieving the desired selectivity, efficacy, and safety profiles required for effective therapeutic intervention. This comprehensive review provided insights into the molecular mechanisms associated with α7 nAChR and its role in cancer progression, particularly in lung cancer. Furthermore, it presents an update on recent evidence about α7 nAChR antagonists and addresses the challenges encountered in drug research and discovery in this field.
Collapse
Affiliation(s)
- Kuntarat Arunrungvichian
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
- Unit
of Compounds Library for Drug Discovery, Mahidol University, Bangkok 10400, Thailand
| | - Opa Vajragupta
- Research
Affairs, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Yoshihiro Hayakawa
- Institute
of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Varisa Pongrakhananon
- Department
of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Preclinical
Toxicity and Efficacy Assessment of Medicines and Chemicals Research
Unit, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
8
|
Yang Y, Wang Q, Zhan F. Unraveling the Action Mechanism of Tubeimoside-1 against Tumor Microvessels via Network Pharmacology and Experimental Validation. J Cancer 2024; 15:955-965. [PMID: 38230220 PMCID: PMC10788730 DOI: 10.7150/jca.90391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/27/2023] [Indexed: 01/18/2024] Open
Abstract
Objective: Tubeimoside-1 (TBMS1) is a plant-derived triterpenoid saponin that exhibits pharmacological properties and anti-tumor effects, but the anti-tumor microvessels of action of TBMS1 remains to be completely elucidated. This study aims to verify the effect of TBMS1 on tumor microvessels and its underlying mechanism. Methods: A SKOV3 xenografted mouse model were constructed to evaluate the anti-tumor microvessels of TBMS1 in vivo, followed by function assays to verify the effects of TBMS1 on the proliferation, cell cycle, migration, and tubule formation of vascular endothelial cells in vitro. Next, based on network pharmacology, the drug/disease-target protein-protein interaction (PPI) networks, biological functions and gene enrichment analyses were performed to predict the underlying mechanism. Finally, molecules and pathways associated with tumor trans-endothelial migration were identified. Results: TBMS1 treatment effectively reduced tumor microvessel density in ovarian cancer model and inhibited the proliferation, cell cycle, migration, and induced apoptosis of vascular endothelial cells in vitro. Network pharmacological data suggested that tumor cell adhesion and trans-endothelial migration may participate in antiangiogenic effects of TBMS1. By endothelial adhesion and permeability assay, we identified that tumor adhesion and the permeability of endothelial monolayers were reduced by TBMS1. Furthermore, adhesion protein (VCAM-1and ICAM-1) and tight junction (TJ) proteins (VE-cadhsion, ZO-1 and claudin-5) were found to be regulated. Finally, Akt, Erk1/2, Stat3 and NF-κB signaling were decreased by TBMS1 treatment. Conclusion: To sum up, our findings strongly suggest that clinical application of TBSM1 may serve as a vasoactive drug treatment to suppress tumor progression.
Collapse
Affiliation(s)
- YinRong Yang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University (Qingdao), Qingdao, Shandong 266035, China
| | - Qian Wang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University (Qingdao), Qingdao, Shandong 266035, China
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong 250012, China
| | - FengXia Zhan
- Department of Clinical Laboratory, Shandong University School Hospital, Jinan, Shandong, 250012, China
| |
Collapse
|
9
|
Gorbunova AS, Zamaraev AV, Yapryntseva MA, Kovaleva OV, Tchevkina EM, Turkina MV, Zhivotovsky B, Kopeina GS. Prognostic signature based on mitochondria quality control proteins for the prediction of lung adenocarcinoma patients survival. Cell Death Discov 2023; 9:352. [PMID: 37749074 PMCID: PMC10519931 DOI: 10.1038/s41420-023-01649-x] [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: 06/14/2023] [Revised: 08/30/2023] [Accepted: 09/13/2023] [Indexed: 09/27/2023] Open
Abstract
Lung cancer is the leading cause of cancer mortality worldwide. In recent years, the incidence of lung cancer subtype lung adenocarcinoma (LUAD) has steadily increased. Mitochondria, as a pivotal site of cell bioenergetics, metabolism, cell signaling, and cell death, are often dysregulated in lung cancer cells. Mitochondria maintenance and integrity depend on mitochondrial quality control proteins (MQCPs). During lung cancer progression, the levels of MQCPs could change and promote cancer cell adaptation to the microenvironment and stresses. Here, univariate and multivariate proportional Cox regression analyses were applied to develop a signature based on the level of MQCPs (dimeric form of BNIP3, DRP1, and SIRT3) in tumorous and non-tumorous samples of 80 patients with LUAD. The MQCP signature could be used to separate the patients with LUAD into high- and low-risk groups. Survival analysis indicated that patients in the high-risk group had dramatically shorter overall survival compared with the low-risk patients. Moreover, a nomogram combining clinicopathologic features and the MQCP signature was constructed and validated to predict 1-, 3-, and 5-year overall survival of the patients. Thus, this study presents a novel signature based on MQCPs as a reliable prognostic tool to predict overall survival for patients with LUAD.
Collapse
Affiliation(s)
- Anna S Gorbunova
- Faculty of Medicine, MV Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Alexey V Zamaraev
- Faculty of Medicine, MV Lomonosov Moscow State University, 119991, Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - Maria A Yapryntseva
- Faculty of Medicine, MV Lomonosov Moscow State University, 119991, Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - Olga V Kovaleva
- Department of Oncogenes Regulation, NN Blokhin Medical Research Center of Oncology, 115478, Moscow, Russia
| | - Elena M Tchevkina
- Department of Oncogenes Regulation, NN Blokhin Medical Research Center of Oncology, 115478, Moscow, Russia
| | - Maria V Turkina
- Faculty of Medicine and Heath Sciences, Department of Clinical and Experimental Medicine, Linköpings Universitet, 58185, Linkoping, Sweden
| | - Boris Zhivotovsky
- Faculty of Medicine, MV Lomonosov Moscow State University, 119991, Moscow, Russia.
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia.
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 17177, Stockholm, Sweden.
| | - Gelina S Kopeina
- Faculty of Medicine, MV Lomonosov Moscow State University, 119991, Moscow, Russia.
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia.
| |
Collapse
|
10
|
Liu ZL, Chen HH, Zheng LL, Sun LP, Shi L. Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct Target Ther 2023; 8:198. [PMID: 37169756 PMCID: PMC10175505 DOI: 10.1038/s41392-023-01460-1] [Citation(s) in RCA: 408] [Impact Index Per Article: 204.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/20/2023] [Accepted: 04/20/2023] [Indexed: 05/13/2023] Open
Abstract
Angiogenesis, the formation of new blood vessels, is a complex and dynamic process regulated by various pro- and anti-angiogenic molecules, which plays a crucial role in tumor growth, invasion, and metastasis. With the advances in molecular and cellular biology, various biomolecules such as growth factors, chemokines, and adhesion factors involved in tumor angiogenesis has gradually been elucidated. Targeted therapeutic research based on these molecules has driven anti-angiogenic treatment to become a promising strategy in anti-tumor therapy. The most widely used anti-angiogenic agents include monoclonal antibodies and tyrosine kinase inhibitors (TKIs) targeting vascular endothelial growth factor (VEGF) pathway. However, the clinical benefit of this modality has still been limited due to several defects such as adverse events, acquired drug resistance, tumor recurrence, and lack of validated biomarkers, which impel further research on mechanisms of tumor angiogenesis, the development of multiple drugs and the combination therapy to figure out how to improve the therapeutic efficacy. Here, we broadly summarize various signaling pathways in tumor angiogenesis and discuss the development and current challenges of anti-angiogenic therapy. We also propose several new promising approaches to improve anti-angiogenic efficacy and provide a perspective for the development and research of anti-angiogenic therapy.
Collapse
Affiliation(s)
- Zhen-Ling Liu
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Huan-Huan Chen
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Li-Li Zheng
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Li-Ping Sun
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China.
| | - Lei Shi
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China.
| |
Collapse
|
11
|
Lahiri A, Maji A, Potdar PD, Singh N, Parikh P, Bisht B, Mukherjee A, Paul MK. Lung cancer immunotherapy: progress, pitfalls, and promises. Mol Cancer 2023; 22:40. [PMID: 36810079 PMCID: PMC9942077 DOI: 10.1186/s12943-023-01740-y] [Citation(s) in RCA: 450] [Impact Index Per Article: 225.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/22/2022] [Indexed: 02/23/2023] Open
Abstract
Lung cancer is the primary cause of mortality in the United States and around the globe. Therapeutic options for lung cancer treatment include surgery, radiation therapy, chemotherapy, and targeted drug therapy. Medical management is often associated with the development of treatment resistance leading to relapse. Immunotherapy is profoundly altering the approach to cancer treatment owing to its tolerable safety profile, sustained therapeutic response due to immunological memory generation, and effectiveness across a broad patient population. Different tumor-specific vaccination strategies are gaining ground in the treatment of lung cancer. Recent advances in adoptive cell therapy (CAR T, TCR, TIL), the associated clinical trials on lung cancer, and associated hurdles are discussed in this review. Recent trials on lung cancer patients (without a targetable oncogenic driver alteration) reveal significant and sustained responses when treated with programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) checkpoint blockade immunotherapies. Accumulating evidence indicates that a loss of effective anti-tumor immunity is associated with lung tumor evolution. Therapeutic cancer vaccines combined with immune checkpoint inhibitors (ICI) can achieve better therapeutic effects. To this end, the present article encompasses a detailed overview of the recent developments in the immunotherapeutic landscape in targeting small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). Additionally, the review also explores the implication of nanomedicine in lung cancer immunotherapy as well as the combinatorial application of traditional therapy along with immunotherapy regimens. Finally, ongoing clinical trials, significant obstacles, and the future outlook of this treatment strategy are also highlighted to boost further research in the field.
Collapse
Affiliation(s)
- Aritraa Lahiri
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, 741246, India
| | - Avik Maji
- Department of Radiation Oncology, N. R. S. Medical College & Hospital, 138 A.J.C. Bose Road, Kolkata, 700014, India
| | - Pravin D Potdar
- Department of Molecular Medicine and Stem Cell Biology, Jaslok Hospital and Research Centre, Mumbai, 400026, India
| | - Navneet Singh
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Purvish Parikh
- Department of Clinical Hematology, Mahatma Gandhi Medical College and Hospital, Jaipur, Rajasthan, 302022, India
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, 400012, India
| | - Bharti Bisht
- Division of Thoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Anubhab Mukherjee
- Esperer Onco Nutrition Pvt Ltd, 4BA, 4Th Floor, B Wing, Gundecha Onclave, Khairani Road, Sakinaka, Andheri East, Mumbai, Maharashtra, 400072, India.
| | - Manash K Paul
- Department of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Microbiology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
12
|
Samarth N, Gulhane P, Singh S. Immunoregulatory framework and the role of miRNA in the pathogenesis of NSCLC - A systematic review. Front Oncol 2022; 12:1089320. [PMID: 36620544 PMCID: PMC9811680 DOI: 10.3389/fonc.2022.1089320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
With a 5-year survival rate of only 15%, non-small cell lung cancer (NSCLC), the most common kind of lung carcinoma and the cause of millions of deaths annually, has drawn attention. Numerous variables, such as disrupted signaling caused by somatic mutations in the EGFR-mediated RAS/RAF/MAPK, PI3K/AKT, JAK/STAT signaling cascade, supports tumour survival in one way or another. Here, the tumour microenvironment significantly contributes to the development of cancer by thwarting the immune response. MicroRNAs (miRNAs) are critical regulators of gene expression that can function as oncogenes or oncosuppressors. They have a major influence on the occurrence and prognosis of NSCLC. Though, a myriad number of therapies are available and many are being clinically tested, still the drug resistance, its adverse effect and toxicity leading towards fatality cannot be ruled out. In this review, we tried to ascertain the missing links in between perturbed EGFR signaling, miRNAs favouring tumorigenesis and the autophagy mechanism. While connecting all the aforementioned points multiple associations were set, which can be targeted in order to combat NSCLC. Here, we tried illuminating designing synthetically engineered circuits with the toggle switches that might lay a prototype for better therapeutic paradigm.
Collapse
Affiliation(s)
| | | | - Shailza Singh
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune, India
| |
Collapse
|
13
|
Cancer-derived exosomal miR-197-3p confers angiogenesis via targeting TIMP2/3 in lung adenocarcinoma metastasis. Cell Death Dis 2022; 13:1032. [PMID: 36494333 PMCID: PMC9734149 DOI: 10.1038/s41419-022-05420-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 11/01/2022] [Accepted: 11/08/2022] [Indexed: 12/13/2022]
Abstract
Cancer-derived exosomal miRNAs are implicated in tumorigenesis and development of lung adenocarcinoma (LUAD). The objective of this study is to unravel the biological function of exosomal miR-197-3p in LUAD metastasis. qRT-PCR showed that elevated miR-197-3p in LUAD tissues was positively correlated with LUAD metastasis. CCK-8, tube formation, transwell and wound healing assays revealed that exosomal miR-197-3p from LUAD cells promoted the proliferation, angiogenesis and migration of HUVECs in vitro. LUAD cells-derived exosomal miR-197-3p also facilitated tumor growth and angiogenesis in LUAD cells-derived tumor xenograft model. TIMP2 and TIMP3 were identified as target genes of miR-197-3p in HUVECs by bioinformatics analysis and luciferase reporter assay. Functional studies illustrated that exosomal miR-197-3p promoted angiogenesis and migration via targeting TIMP2 and TIMP3 in HUVECs. In vivo data further supported that exosomal miR-197-3p promoted lung metastasis via TIMP2/3-mediated angiogenesis. In conclusion, LUAD cells-derived exosomal miR-197-3p conferred angiogenesis via targeting TIMP2/3 in LUAD metastasis.
Collapse
|
14
|
Hatami E, B Nagesh PK, Sikander M, Dhasmana A, Chauhan SC, Jaggi M, Yallapu MM. Tannic Acid Exhibits Antiangiogenesis Activity in Nonsmall-Cell Lung Cancer Cells. ACS OMEGA 2022; 7:23939-23949. [PMID: 35847334 PMCID: PMC9281317 DOI: 10.1021/acsomega.2c02727] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Nonsmall-cell lung cancer (NSCLC) is the most common type of lung cancer, with a dismal prognosis. NSCLC is a highly vascularized tumor, and chemotherapy is often hampered by the development of angiogenesis. Therefore, suppression of angiogenesis is considered a potential treatment approach. Tannic acid (TA), a natural polyphenol, has been demonstrated to have anticancer properties in a variety of cancers; however, its angiogenic properties have yet to be studied. Hence, in the current study, we investigated the antiproliferative and antiangiogenic effects of TA on NSCLC cells. The (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) (MTS) assay revealed that TA induced a dose- and time-dependent decrease in the proliferation of A549 and H1299 cells. However, TA had no significant toxicity effects on human bronchial epithelial cells. Clonogenicity assay revealed that TA suppressed colony formation ability in NSCLC cells in a dose-dependent manner. The anti-invasiveness and antimigratory potential of TA were confirmed by Matrigel and Boyden chamber studies, respectively. Importantly, TA also decreased the ability of human umbilical vein endothelial cells (HUVEC) to form tube-like networks, demonstrating its antiangiogenic properties. Extracellular vascular endothelial growth factor (VEGF) release was reduced in TA-treated cells compared to that in control cells, as measured by the enzyme-linked immunosorbent assay (ELISA). Overall, these results demonstrate that TA can induce antiproliferative and antiangiogenic effects against NSCLC.
Collapse
Affiliation(s)
- Elham Hatami
- Department
of Pharmaceutical Sciences, University of
Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department
of Bioengineering, University of California, Los Angeles, California 90095, United States
| | - Prashanth K. B Nagesh
- Department
of Pharmaceutical Sciences, University of
Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Laboratory
of Signal Transduction, Memorial Sloan Kettering
Cancer Center, New York, New York 10065, United States
- Department
of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Mohammed Sikander
- Department
of Pharmaceutical Sciences, University of
Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department
of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South
Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Anupam Dhasmana
- Department
of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South
Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Subhash C. Chauhan
- Department
of Pharmaceutical Sciences, University of
Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department
of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South
Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Meena Jaggi
- Department
of Pharmaceutical Sciences, University of
Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department
of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South
Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Murali M. Yallapu
- Department
of Pharmaceutical Sciences, University of
Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department
of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South
Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- . Tel: 956-296-1734
| |
Collapse
|
15
|
Efficacy and safety of anlotinib with and without EGFR-TKIs or immunotherapy in the treatment of elder patients with non-small-cell lung cancer: a retrospective study. BMC Pulm Med 2022; 22:179. [PMID: 35524294 PMCID: PMC9074279 DOI: 10.1186/s12890-022-01981-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 05/03/2022] [Indexed: 12/03/2022] Open
Abstract
Background Anlotinib is a multitarget tyrosine kinase inhibitor for treating patients with advanced non-small cell lung cancer (NSCLC). We aimed to assess the efficacy and safety of anlotinib in elder patients with advanced NSCLC. Methods Elder patients with advanced NSCLC who received anlotinib were enrolled. They were all age ≥ 65 years and with demonstrated records of EGFR gene status. All patients had received treatment with anlotinib or immune checkpoint inhibitors (ICIs)/EGFR-TKIs. The efficacy was evaluated according to the efficacy evaluation criteria for solid tumors (RECIST 1.1). Common Adverse Events Evaluation Criteria (CTCAE 4.03) were used to evaluate adverse drug reactions. Results A total of 91 patients were included in this study. We divided the patients into two groups (EGFR wild type: 60 patients; EGFR mutation: 31 patients). Among EGFR negative patients, the progression-free survival (PFS) for anlotinib monotherapy and anlotinib combination ICI therapy was 3.2 months and 5.0 months, respectively (P = 0.012). The difference in overall survival (OS) between monotherapy and combination therapy was also significant (9.5 vs. 18.4 months, respectively P = 0.010). Interestingly, we further analyzed differences between patients with hypertension and without hypertension, and found that hypertension was associated with better prognosis (5.7 vs. 1.4 months, P < 0.0001). In the EGFR mutation group, the PFS for anlotinib and EGFR-TKI combination treatment indicated better efficacy than that of anlotinib monotherapy (1.83 months vs. 7.03 months, respectively, P = 0.001). The median OS for monotherapy and combination therapy in the EGFR mutation group showed no statistical difference (28.34 months vs. 31.37 months, P = 0.223). The most common adverse reactions were hypertension, fatigue, and hand-foot syndrome, mainly of grade 1 or 2. No significant increase in adverse reactions was observed in patients ≥ 70 years of age. Conclusions Anlotinib treatment and combination regimens resulted in good efficacy and controllable adverse reactions in elder patients with advanced NSCLC.
Collapse
|
16
|
Qin X, Liu M, Xu C, Xing B, Xu X, Wu Y, Ding H, Zhao Q. ZDQ-0620, a Novel Phosphatidylinositol 3-Kinase Inhibitor, Inhibits Colorectal Carcinoma Cell Proliferation and Suppresses Angiogenesis by Attenuating PI3K/AKT/mTOR Pathway. Front Oncol 2022; 12:848952. [PMID: 35311154 PMCID: PMC8924359 DOI: 10.3389/fonc.2022.848952] [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: 01/05/2022] [Accepted: 02/10/2022] [Indexed: 12/11/2022] Open
Abstract
The PI3K/AKT pathway plays a central role in human cancers, aberrant activation of this pathway is associated with tumorigenesis, cancer progression and angiogenesis. Based on the importance of the PI3K/AKT pathway in malignancies, we developed a 4-aminoquinazoline derivative, ZDQ-0620, initially envisioned as a novel pan-PI3K inhibitor. This study aimed to evaluate the potential target of ZDQ-0620 and its anticancer effect in human colorectal carcinoma (CRC). PI3K-kinase activity test showed IC50 of ZDQ-0620 against PI3Ka was 0.5 nM; molecular docking, CETSA assay and western blotting was further performed to predict ZDQ-0620 was a PI3K/AKT pathway inhibitor by targeting PI3K. To identify the effect of ZDQ-0620 on CRC cells, Sulforhodamine B (SRB) assay, flow cytometry, and Cell morphology analysis were conducted. The results showed that ZDQ-0620 inhibited the proliferation, migration and invasion of CRC cells, induced apoptosis through G0/G1 cell cycle arrest and mitochondrial pathway. Additionally, ZDQ-0620 inhibited the migration and tube formation of human umbilical vein endothelial cells (HUVECs). In vivo, neovascularization of rat aortic ring and chick chorioallantoic membrane (CAM) induced by VEGF was diminished when treated with ZDQ-0620. These results indicate that ZDQ-0620 induce apoptosis and anti-angiogenesis via inhibits the PI3K/AKT pathway. We suggest that the great potential of ZDQ-0620 as an effective treatment candidate against CRC.
Collapse
Affiliation(s)
- Xiaochun Qin
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China.,Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Mingyue Liu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Chang Xu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Bo Xing
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiangbo Xu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Yuting Wu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Huaiwei Ding
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Qingchun Zhao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China.,Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
17
|
Suo J, Sun Y, Fu Y, Xiu W, Zhang X, Wang Y, Zhu J. A Retrospective Analysis of the Effect of Anlotinib in Patients With Lung Cancer With or Without Previous Antiangiogenic Therapy. Front Oncol 2022; 11:788837. [PMID: 35004313 PMCID: PMC8732369 DOI: 10.3389/fonc.2021.788837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 11/24/2021] [Indexed: 11/23/2022] Open
Abstract
Objective The purpose of this study was to initially investigate the effect of previous antiangiogenic therapy (bevacizumab and endostatin) on the efficacy of anlotinib in patients with advanced or metastatic lung cancer (LC). Methods We retrospectively collected the clinical data of patients with LC treated with anlotinib and divided them into group A (treated with anlotinib after the failure of previous antiangiogenic drugs and group B (no prior use of antiangiogenic drugs). We used propensity score matching (PSM) for confounding factors between the groups. Progression-free survival (PFS) and overall survival (OS) were also recorded. Results A total of 160 patients were included in the analysis. The median OS in groups A and group B was 11.8 months and 16.1 months (P=0.120), whereas the median PFS was 3.1 months and 4.7 months (P=0.009), respectively. Moreover, the objective response rate (ORR) of the two groups was 9.6% and 10.4% (P=0.874), and the disease control rate (DCR) was 71.1% and 80.5% (P=0.165). After PSM (n=46), baseline characteristics were comparable between groups A and B. Furthermore, the median OS of the two groups was 14.6 months and 16.2 months (P=0.320), whereas the median PFS was 3.5 months and 4.5 months (P=0.040), respectively. Moreover, the ORR of the two groups were 13.0% and 10.9% (P=0.748), and the DCR were 78.3% and 82.6% (P=0.599), respectively. Conclusions Previous antiangiogenic treatments may affect the PFS of patients who receive anlotinib later, but it might not affect the patient’s ORR and OS.
Collapse
Affiliation(s)
- Jiaojiao Suo
- Department of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Sun
- Radiation Physics Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Fu
- Department of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Weigang Xiu
- Department of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Xuanwei Zhang
- Department of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Wang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, West China 2nd University Hospital, Sichuan University, Chengdu, China.,Key laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Jiang Zhu
- Department of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Chis AF, Râjnoveanu RM, Man MA, Todea DA, Chis BA, Stancu B, Arghir IA, Alexescu TG, Pop CM. Increased Vascular Endothelial Growth Factor Serum Level and the Role of +936C/T Gene Polymorphism in Chronic Obstructive Pulmonary Disease. Medicina (B Aires) 2021; 57:medicina57121351. [PMID: 34946296 PMCID: PMC8707840 DOI: 10.3390/medicina57121351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/30/2021] [Accepted: 12/05/2021] [Indexed: 11/16/2022] Open
Abstract
Background and Objectives: Chronic obstructive pulmonary disease (COPD) represents a debilitating disease, with rising morbidity and mortality. Vascular endothelial growth factor (VEGF) plays a major role in angiogenesis, vascular permeability, and airway remodeling. The purpose of this study was to investigate the relationship between VEGF serum levels and VEGF +936 C/T gene polymorphism (rs3025039) with COPD, for the first time in a Romanian population. Materials and Methods: In total, 120 participants from Transylvania were included in this case-control study. Serum levels of VEGF were determined using an enzyme-linked immune-sorbent assay and rs3025039 was investigated by high molecular weight genomic deoxyribonucleic acid (DNA). Spirometric values, arterial blood gas analysis, and the Six Minute Walk Test (6MWT) outcome were also determined. Results: The serum level of VEGF was higher in the COPD group versus controls (p < 0.001), with a positive correlation with the 6MWT outcome. No significant difference was observed in the VEGF serum levels between VEGF +936C/T genotypes. There was no difference in the VEGF +936C/T genotype between COPD patients and healthy subjects (chi2 test p = 0.92, OR = 1.04, 95%CI = 0.41–2.62), but the presence of the T allele was significantly linked to the presence of COPD (chi2 test p = 0.02, OR = 2.36, 95%CI = 1.12–4.97). Conclusions: Higher VEGF serum levels were found in moderate and severe COPD and were positively correlated with the distance in the 6MWT. No significant difference was found between CC, CT, and TT genotypes of rs3025039 and the presence of COPD. The presence of the T allele was found to be linked to COPD and also to the degree of airway obstruction.
Collapse
Affiliation(s)
- Ana Florica Chis
- Department of Pneumology, “Iuliu Haţieganu” University of Medicine and Pharmacy Cluj-Napoca, 8th Victor Babeș Street, 400012 Cluj, Romania; (A.F.C.); (R.-M.R.); (M.A.M.); (D.A.T.); (C.M.P.)
- “Leon Daniello” Clinical Hospital of Pneumology, 6th Bogdan Petriceicu Hașdeu Street, 400332 Cluj, Romania
| | - Ruxandra-Mioara Râjnoveanu
- Department of Pneumology, “Iuliu Haţieganu” University of Medicine and Pharmacy Cluj-Napoca, 8th Victor Babeș Street, 400012 Cluj, Romania; (A.F.C.); (R.-M.R.); (M.A.M.); (D.A.T.); (C.M.P.)
- “Leon Daniello” Clinical Hospital of Pneumology, 6th Bogdan Petriceicu Hașdeu Street, 400332 Cluj, Romania
| | - Milena Adina Man
- Department of Pneumology, “Iuliu Haţieganu” University of Medicine and Pharmacy Cluj-Napoca, 8th Victor Babeș Street, 400012 Cluj, Romania; (A.F.C.); (R.-M.R.); (M.A.M.); (D.A.T.); (C.M.P.)
- “Leon Daniello” Clinical Hospital of Pneumology, 6th Bogdan Petriceicu Hașdeu Street, 400332 Cluj, Romania
| | - Doina Adina Todea
- Department of Pneumology, “Iuliu Haţieganu” University of Medicine and Pharmacy Cluj-Napoca, 8th Victor Babeș Street, 400012 Cluj, Romania; (A.F.C.); (R.-M.R.); (M.A.M.); (D.A.T.); (C.M.P.)
- “Leon Daniello” Clinical Hospital of Pneumology, 6th Bogdan Petriceicu Hașdeu Street, 400332 Cluj, Romania
| | - Bogdan Augustin Chis
- 2nd Department of Internal Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 8th Victor Babeș Street, 400012 Cluj, Romania
- Correspondence: ; Tel.: +40-740047253
| | - Bogdan Stancu
- 2nd Department of General Surgery, “Iuliu Hațieganu” University of Medicine and Pharmacy, 8th Victor Babeș Street, 400012 Cluj, Romania;
| | - Ioan Anton Arghir
- 4th Department of Clinical Medical Disciplines II, “Ovidius” University of Medicine and Pharmacy, Mamaia Boulevard, No. 124, 900527 Constanța, Romania;
| | - Teodora Gabriela Alexescu
- 5th Departament Internal Medicine, 4th Medical Clinic, University of Medicine and Pharmacy, 400015 Cluj, Romania;
| | - Carmen Monica Pop
- Department of Pneumology, “Iuliu Haţieganu” University of Medicine and Pharmacy Cluj-Napoca, 8th Victor Babeș Street, 400012 Cluj, Romania; (A.F.C.); (R.-M.R.); (M.A.M.); (D.A.T.); (C.M.P.)
- “Leon Daniello” Clinical Hospital of Pneumology, 6th Bogdan Petriceicu Hașdeu Street, 400332 Cluj, Romania
| |
Collapse
|
19
|
Cheng Y, Zhang T, Xu Q. Therapeutic advances in non-small cell lung cancer: Focus on clinical development of targeted therapy and immunotherapy. MedComm (Beijing) 2021; 2:692-729. [PMID: 34977873 PMCID: PMC8706764 DOI: 10.1002/mco2.105] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 02/05/2023] Open
Abstract
Lung cancer still contributes to nearly one-quarter cancer-related deaths in the past decades, despite the rapid development of targeted therapy and immunotherapy in non-small cell lung cancer (NSCLC). The development and availability of comprehensive genomic profiling make the classification of NSCLC more precise and personalized. Most treatment decisions of advanced-stage NSCLC have been made based on the genetic features and PD-L1 expression of patients. For the past 2 years, more than 10 therapeutic strategies have been approved as first-line treatment for certain subgroups of NSCLC. However, some major challenges remain, including drug resistance and low rate of overall survival. Therefore, we discuss and review the therapeutic strategies of NSCLC, and focus on the development of targeted therapy and immunotherapy in advanced-stage NSCLC. Based on the latest guidelines, we provide an updated summary on the standard treatment for NSCLC. At last, we discussed several potential therapies for NSCLC. The development of new drugs and combination therapies both provide promising therapeutic effects on NSCLC.
Collapse
Affiliation(s)
- Yuan Cheng
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Tao Zhang
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Qing Xu
- Department of OncologyShanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| |
Collapse
|
20
|
Qin X, Liu M, Wu Y, Wang S, Lian S, Jia H, Wu Q, Ding H, Zhao Q. Dual blocking of PI3K and mTOR signaling by DHW-221, a novel benzimidazole derivative, exerts antitumor activity in human non-small cell lung cancer. Clin Transl Med 2021; 11:e514. [PMID: 34586727 PMCID: PMC8473641 DOI: 10.1002/ctm2.514] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 01/17/2023] Open
Affiliation(s)
- Xiaochun Qin
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Mingyue Liu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Yuting Wu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Shu Wang
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Siheng Lian
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Hui Jia
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Qiong Wu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Huaiwei Ding
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Qingchun Zhao
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China.,Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
21
|
Yin L, Liu X, Shao X, Feng T, Xu J, Wang Q, Hua S. The role of exosomes in lung cancer metastasis and clinical applications: an updated review. J Transl Med 2021; 19:312. [PMID: 34281588 PMCID: PMC8287779 DOI: 10.1186/s12967-021-02985-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/10/2021] [Indexed: 02/07/2023] Open
Abstract
Lung cancer is the leading cause of cancer-associated deaths accounting for 24% of all cancer deaths. As a crucial phase of tumor progression, lung cancer metastasis is linked to over 70% of these mortalities. In recent years, exosomes have received increasing research attention in their role in the induction of carcinogenesis and metastasis in the lung. In this review, recent studies on the contribution of exosomes to lung cancer metastasis are discussed, particularly highlighting the role of lung tumor-derived exosomes in immune system evasion, epithelial-mesenchymal transition, and angiogenesis, and their involvement at both the pre-metastatic and metastatic phases. The clinical application of exosomes as therapeutic drug carriers, their role in antitumor drug resistance, and their utility as predictive biomarkers in diagnosis and prognosis are also presented. The metastatic activity, a complex multistep process of cancer cell invasion, survival in blood vessels, attachment and subsequent colonization of the host's organs, is integrated with exosomal effects. Exosomes act as functional mediating factors in cell-cell communication, influencing various steps of the metastatic cascade. To this end, lung cancer cell-derived exosomes enhance cell proliferation, angiogenesis, and metastasis, regulate drug resistance, and antitumor immune activities during lung carcinogenesis, and are currently being explored as an important component in liquid biopsy assessment for diagnosing lung cancer. These nano-sized extracellular vesicles are also being explored as delivery vehicles for therapeutic molecules owing to their unique properties of biocompatibility, circulatory stability, decreased toxicity, and tumor specificity. The current knowledge of the role of exosomes highlights an array of exosome-dependent pathways and cargoes that are ripe for exploiting therapeutic targets to treat lung cancer metastasis, and for predictive value assessment in diagnosis, prognosis, and anti-tumor drug resistance.
Collapse
Affiliation(s)
- Lei Yin
- Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, 215000, People's Republic of China.
| | - Xiaotian Liu
- Department of Anesthesiology, Children's Hospital of Soochow University, Suzhou, 215000, People's Republic of China.
| | - Xuejun Shao
- Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, 215000, People's Republic of China
| | - Tao Feng
- Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, 215000, People's Republic of China
| | - Jun Xu
- Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, 215000, People's Republic of China
| | - Qi Wang
- Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, 215000, People's Republic of China
| | - Shenghao Hua
- Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, 215000, People's Republic of China
| |
Collapse
|
22
|
Liao L, Cen B, Li G, Wei Y, Wang Z, Huang W, He S, Yuan Y, Ji A. A bivalent cyclic RGD-siRNA conjugate enhances the antitumor effect of apatinib via co-inhibiting VEGFR2 in non-small cell lung cancer xenografts. Drug Deliv 2021; 28:1432-1442. [PMID: 34236267 PMCID: PMC8274511 DOI: 10.1080/10717544.2021.1937381] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The vascular endothelial growth factor receptor 2 (VEGFR2) is considered to be a pivotal target for anti-tumor therapy against angiogenesis of non-small cell lung cancer (NSCLC). However, effective and low-toxicity targeted therapies to inhibit VEGFR2 are still lacking. Here, biRGD–siVEGFR2 conjugate comprising murine VEGFR2 siRNA and [cyclo(Arg-Gly-Asp-D-Phe-Lys)-Ahx]2-Glu-PEG-MAL (biRGD) peptide which selectively binds to integrin αvβ3 receptors expressing on neovascularization endothelial cell was synthesized. The anti-tumor activity and renal toxicity of biRGD–siVEGFR2 or its combination therapy with low-dose apatinib were investigated on NSCLC xenografts. The immunogenicity of biRGD–siVEGFR2 was also evaluated in C57BL/6J mice. In vivo, intravenously injected biRGD–siVEGFR2 substantially inhibited NSCLC growth with a marked reduction of vessels and a down-regulation of VEGFR2 in tumor tissue. Furthermore, biRGD–siVEGFR2 in combination with low-dose apatinib achieved powerful anti-tumor effect with less nephrotoxicity compared with the regular dose of apatinib. Besides, no obvious immunogenicity of biRGD–siVEGFR2 was found. These findings demonstrate that biRGD–siVEGFR2 conjugate can be used as a new candidate for the treatment of NSCLC and its combination therapy with apatinib may also provide a novel strategy for cancer treatment in clinic.
Collapse
Affiliation(s)
- Lumin Liao
- Department of Pharmacy, The Seventh Affiliated Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Bohong Cen
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Guoxian Li
- Department of Pharmacy, The Seventh Affiliated Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Yuanyi Wei
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhen Wang
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wen Huang
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuai He
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yawei Yuan
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Aimin Ji
- Department of Pharmacy, The Seventh Affiliated Hospital, Southern Medical University, Foshan, Guangdong, China
| |
Collapse
|
23
|
Chen R, Lu FY, Liu B, Huang J, Zhou M, Dai R, Guo Y. Absolute Neutrophil Count in the Peripheral Blood Predicts Prognosis in Lung Cancer Patients Treated with Anlotinib. Cancer Manag Res 2021; 13:3619-3627. [PMID: 33976572 PMCID: PMC8106457 DOI: 10.2147/cmar.s307368] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/01/2021] [Indexed: 01/10/2023] Open
Abstract
Purpose Anlotinib is a multi-targeted tyrosine kinase inhibitor that inhibits tumor angiogenesis and cell proliferation. It is widely used as a third-line therapy for lung cancer. However, reliable prognostic biomarkers for predicting the efficacy of anlotinib are lacking. We conducted a retrospective study to investigate the prognostic value of serological inflammatory biomarkers in anlotinib treatment. Patients and Methods Patients with advanced lung cancer treated with anlotinib monotherapy were enrolled. Cox regression was conducted to analyze the significant factors related to progression-free survival (PFS) and overall survival (OS). The objective response rate (ORR) was compared based on the median cut-off value of the significant inflammation index. Meanwhile, we created survival curves to compare the two groups and performed receiver operating characteristic curve analysis to assess the predictive ability of the inflammation index. Results Among a total of 71 patients, the median PFS was 5.5 months and the median OS was 9.5 months. The ORR and disease control rate were 16.9% and 84.5%, respectively. According to univariate and multivariate analyses, absolute neutrophil count (ANC) was the only indicator associated with both PFS (hazard ratio [HR] =1.095, 95% confidence interval [CI] 1.030–1.163, P=0.003) and OS (HR=1.057, 95% CI 1.003–1.113, P=0.037). In the group with ANC ≥4.58, the ORR was relatively lower (8.1% vs 26.5%, P=0.057), but not statistically significant; PFS and OS were relatively shorter (median PFS 5.0 [95% CI 4.4–9.6] vs 7.0 months [95% CI 4.4–5.7], P=0.024 and median OS 7.3 [95% CI 4.7–10.0] vs 17.6 months [95% CI 12.3–22.9], P < 0.001). ANC had a relatively high discriminatory ability to predict 10-month survival, with an area under the curve of 0.729, sensitivity of 82.5%, and specificity of 67.7%. Conclusion Elevated pre-treatment ANC was associated with a poor prognosis. Patients with lower peripheral blood levels of ANC might benefit from anlotinib.
Collapse
Affiliation(s)
- Rong Chen
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Fang-Ying Lu
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Bing Liu
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jingwen Huang
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Min Zhou
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Ranran Dai
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yi Guo
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
24
|
Zhang M, Quan H, Fu L, Li Y, Fu H, Lou L. Third-generation EGFR inhibitor HS-10296 in combination with famitinib, a multi-targeted tyrosine kinase inhibitor, exerts synergistic antitumor effects through enhanced inhibition of downstream signaling in EGFR-mutant non-small cell lung cancer cells. Thorac Cancer 2021; 12:1210-1218. [PMID: 33656275 PMCID: PMC8046080 DOI: 10.1111/1759-7714.13902] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/06/2021] [Accepted: 02/06/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND As a highly heterogeneous disease, lung cancer has a multitude of cellular components and patterns of gene expression which are not dependent on a single mutation or signaling pathway. Thus, using combined drugs to treat lung cancer may be a practical strategy. METHODS The combined antitumor effects of HS-10296, a third-generation EGFR inhibitor targeting EGFR T790M mutation, with the multitargeted tyrosine kinase inhibitor (TKI) famitinib in non-small cell lung cancer (NSCLC) were evaluated by in vitro methods such as cell proliferation, apoptosis, angiogenesis assays, and in vivo animal efficacy studies. RESULTS Famitinib strengthened the effects of HS-10296 on inhibiting proliferation and inducing apoptosis of NSCLC cells, possibly by synergistic inhibition of AKT and ERK phosphorylation. Meanwhile, HS-10296 significantly potentiated the effects of famitinib on inhibiting the proliferation and migration of HUVEC, which may be through synergistic inhibition of ERK phosphorylation in HUVEC, suggesting that HS-10296 may improve the inhibition of angiogenesis by famitinib. Moreover, combination of HS-10296 and famitinib exerted synergistic antitumor activity in NCI-H1975 and PC-9 xenograft models, and this effect may be accomplished by synergistic inhibition of phosphorylation of AKT and ERK and tumor angiogenesis in tumor tissues. CONCLUSIONS Collectively, our results indicate that HS-10296 and famitinib exhibit significant synergistic antitumor activity, suggesting that the third-generation EGFR inhibitor combined with VEGFR inhibitor provides a promising strategy in the treatment of EGFR-mutant NSCLC.
Collapse
Affiliation(s)
- Mi Zhang
- School of Life Sciences, Shanghai UniversityShanghaiChina
- Shanghai Institute of Materia Medica, Chinese Academy of SciencesShanghaiChina
| | - Haitian Quan
- Shanghai Institute of Materia Medica, Chinese Academy of SciencesShanghaiChina
| | - Li Fu
- Shanghai Institute of Materia Medica, Chinese Academy of SciencesShanghaiChina
| | - Yun Li
- Shanghai Institute of Materia Medica, Chinese Academy of SciencesShanghaiChina
| | - Haoyu Fu
- Shanghai Institute of Materia Medica, Chinese Academy of SciencesShanghaiChina
| | - Liguang Lou
- Shanghai Institute of Materia Medica, Chinese Academy of SciencesShanghaiChina
| |
Collapse
|
25
|
Huang YS, Chen JLY, Chen HM, Yeh LH, Shih JY, Yen RF, Chang YC. Assessing tumor angiogenesis using dynamic contrast-enhanced integrated magnetic resonance-positron emission tomography in patients with non-small-cell lung cancer. BMC Cancer 2021; 21:348. [PMID: 33794813 PMCID: PMC8017855 DOI: 10.1186/s12885-021-08064-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/18/2021] [Indexed: 12/18/2022] Open
Abstract
Background Angiogenesis assessment is important for personalized therapeutic intervention in patients with non-small-cell lung cancer (NSCLC). This study investigated whether radiologic parameters obtained by dynamic contrast-enhanced (DCE)-integrated magnetic resonance-positron emission tomography (MR-PET) could be used to quantitatively assess tumor angiogenesis in NSCLC. Methods This prospective cohort study included 75 patients with NSCLC who underwent DCE-integrated MR-PET at diagnosis. The following parameters were analyzed: metabolic tumor volume (MTV), maximum standardized uptake value (SUVmax), reverse reflux rate constant (kep), volume transfer constant (Ktrans), blood plasma volume fraction (vp), extracellular extravascular volume fraction (ve), apparent diffusion coefficient (ADC), and initial area under the time-to-signal intensity curve at 60 s post enhancement (iAUC60). Serum biomarkers of tumor angiogenesis, including vascular endothelial growth factor-A (VEGF-A), angiogenin, and angiopoietin-1, were measured by enzyme-linked immunosorbent assays simultaneously. Results Serum VEGF-A (p = 0.002), angiogenin (p = 0.023), and Ang-1 (p < 0.001) concentrations were significantly elevated in NSCLC patients compared with healthy individuals. MR-PET parameters, including MTV, Ktrans, and kep, showed strong linear correlations (p < 0.001) with serum angiogenesis-related biomarkers. Serum VEGF-A concentrations (p = 0.004), MTV values (p < 0.001), and kep values (p = 0.029) were significantly higher in patients with advanced-stage disease (stage III or IV) than in those with early-stage disease (stage I or II). Patients with initial higher values of angiogenesis-related MR-PET parameters, including MTV > 30 cm3 (p = 0.046), Ktrans > 200 10− 3/min (p = 0.069), and kep > 900 10− 3/min (p = 0.048), may have benefited from angiogenesis inhibitor therapy, which thus led to significantly longer overall survival. Conclusions The present findings suggest that DCE-integrated MR-PET provides a reliable, non-invasive, quantitative assessment of tumor angiogenesis; can guide the use of angiogenesis inhibitors toward longer survival; and will play an important role in the personalized treatment of NSCLC.
Collapse
Affiliation(s)
- Yu-Sen Huang
- Department of Radiology, National Taiwan University College of Medicine, No. 7, Chung-Shan S. Rd., Taipei, 100, Taiwan.,Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
| | - Jenny Ling-Yu Chen
- Department of Radiology, National Taiwan University College of Medicine, No. 7, Chung-Shan S. Rd., Taipei, 100, Taiwan.,Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.,National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hsin-Ming Chen
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
| | - Li-Hao Yeh
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
| | - Jin-Yuan Shih
- Department of Internal Medicine National Taiwan University Hospital, Taipei, Taiwan
| | - Ruoh-Fang Yen
- Department of Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yeun-Chung Chang
- Department of Radiology, National Taiwan University College of Medicine, No. 7, Chung-Shan S. Rd., Taipei, 100, Taiwan. .,Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
26
|
Liang J, Gu W, Jin J, Zhang H, Chen Z, Tang Y, Zhang S, Yang S, Deng Y, Feng W. Efficacy and safety of apatinib as third- or further-line therapy for patients with advanced NSCLC: a retrospective study. Ther Adv Med Oncol 2021; 12:1758835920968472. [PMID: 33403012 PMCID: PMC7745562 DOI: 10.1177/1758835920968472] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/30/2020] [Indexed: 01/26/2023] Open
Abstract
Background Apatinib, an oral small-molecule angiogenesis inhibitor, selectively inhibits vascular endothelial growth factor receptor 2 (VEGFR-2), which inhibits vascular endothelial growth factor (VEGF) stimulated endothelial cell migration and proliferation and decreases tumour growth and metastasis. Recently, the efficacy of multi-target angiogenic drugs has been demonstrated for many cancers, including non-small-cell lung cancer (NSCLC). The aim of this retrospective study was to evaluate the clinical efficacy of apatinib in patients with advanced NSCLC. Patients and methods We conducted a retrospective analysis of 70 patients with advanced NSCLC who received second-line and later treatment from November 2015 to July 2017 with poor results. Out of the 70 patients, 36 patients received apatinib treatment after second-line or later treatment, whereas 34 patients in the control group did not receive further treatment. The patients were treated with oral apatinib 500 mg once a day every day for 4 weeks per cycle. Treatment was continued in responding and stable patients until disease progression or intolerable toxicity. The objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and side effects of the drug were recorded and reviewed. Results ORR, DCR, PFS, and OS were evaluated in 36 patients receiving apatinib and 34 patients in the control group. The ORR and DCR in patients receiving apatinib therapy were 22.2% and 77.8%, respectively. The median PFS and OS in the treatment group were 5.6 and 9.6 months, respectively. The median OS in the apatinib group was significantly longer than that in the control group (9.6 versus 3.8 months; p < 0.0001). In contrast, there were no differences in adverse reactions between the patients in the treatment and control groups. Conclusion Apatinib showed favourable efficacy and safety and can thus be used as a treatment option for patients with advanced NSCLC.
Collapse
Affiliation(s)
- Jianmiao Liang
- Department of Head and Neck/Thoracic Medical Oncology, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Weiguang Gu
- Oncology Department, Nanhai People's Hospital/The Second School of Clinical Medical, Southern Medical University, Foshan, Guangdong, China
| | - Jun Jin
- Department of Oncology, Guangdong Province Hospital of Combination of Traditional Chinese and Western Medicine, Foshan, Guangdong, China
| | - Hua Zhang
- Department of Head and Neck/Thoracic Medical Oncology, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Zecheng Chen
- Department of Head and Neck/Thoracic Medical Oncology, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Yicong Tang
- Department of Head and Neck/Thoracic Medical Oncology, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Shunda Zhang
- Department of Head and Neck/Thoracic Medical Oncology, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Shuang Yang
- Department of Head and Neck/Thoracic Medical Oncology, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Yanming Deng
- Department of Head and Neck/Thoracic Medical Oncology, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Weineng Feng
- Department of Head and Neck/Thoracic Medical Oncology, The First People's Hospital of Foshan, No. 81, North Lingnan Avenue, Chancheng District, Foshan City, Guangdong Province 528041, China
| |
Collapse
|
27
|
Daum S, Hagen H, Naismith E, Wolf D, Pircher A. The Role of Anti-angiogenesis in the Treatment Landscape of Non-small Cell Lung Cancer - New Combinational Approaches and Strategies of Neovessel Inhibition. Front Cell Dev Biol 2021; 8:610903. [PMID: 33469537 PMCID: PMC7813779 DOI: 10.3389/fcell.2020.610903] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Tumor progression depends primarily on vascular supply, which is facilitated by angiogenic activity within the malignant tissue. Non-small cell lung cancer (NSCLC) is a highly vascularized tumor, and inhibition of angiogenesis was projected to be a promising therapeutic approach. Over a decade ago, the first anti-angiogenic agents were approved for advanced stage NSCLC patients, however, they only produced a marginal clinical benefit. Explanations why anti-angiogenic therapies only show modest effects include the highly adaptive tumor microenvironment (TME) as well as the less understood characteristics of the tumor vasculature. Today, advanced methods of in-depth characterization of the NSCLC TME by single cell RNA sequencing (scRNA-Seq) and preclinical observations enable a detailed characterization of individual cancer landscapes, allowing new aspects for a more individualized inhibition of angiogenesis to be identified. Furthermore, the tumor vasculature itself is composed of several cellular subtypes, which closely interact with other cellular components of the TME, and show distinct biological functions such as immune regulation, proliferation, and organization of the extracellular matrix. With these new insights, combinational approaches including chemotherapy, anti- angiogenic and immunotherapy can be developed to yield a more target-oriented anti-tumor treatment in NSCLC. Recently, anti-angiogenic agents were also shown to induce the formation of high endothelial venules (HEVs), which are essential for the formation of tertiary lymphoid structures, and key components in triggering anti-tumor immunity. In this review, we will summarize the current knowledge of tumor-angiogenesis and corresponding anti-angiogenic therapies, as well as new aspects concerning characterization of tumor-associated vessels and the resulting new strategies for anti-angiogenic therapies and vessel inhibition in NSCLC. We will further discuss why anti-angiogenic therapies form an interesting backbone strategy for combinational therapies and how anti-angiogenic approaches could be further developed in a more personalized tumor-oriented fashion with focus on NSCLC.
Collapse
Affiliation(s)
- Sophia Daum
- Internal Medicine V, Department of Hematology and Oncology, Medical University Innsbruck, Innsbruck, Austria
| | - Hannes Hagen
- Internal Medicine V, Department of Hematology and Oncology, Medical University Innsbruck, Innsbruck, Austria
| | - Erin Naismith
- Internal Medicine V, Department of Hematology and Oncology, Medical University Innsbruck, Innsbruck, Austria
| | - Dominik Wolf
- Internal Medicine V, Department of Hematology and Oncology, Medical University Innsbruck, Innsbruck, Austria
- Medical Clinic 3, Department of Oncology, Hematology, Immunoncology and Rheumatology, University Hospital Bonn (UKB), Bonn, Germany
| | - Andreas Pircher
- Internal Medicine V, Department of Hematology and Oncology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
28
|
Yang Y, Luo H, Zheng XL, Ge H. The optimal immune checkpoint inhibitors combined with chemotherapy for advanced non-small-cell lung cancer: a systematic review and meta-analysis. Clin Transl Oncol 2020; 23:1117-1127. [PMID: 33211281 DOI: 10.1007/s12094-020-02502-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/15/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) plus chemotherapy (CT) have strikingly expanded the therapeutic landscape for advanced non-small cell lung cancer (NSCLC), but little is known about which is superior. We performed a meta-analysis that compared the efficacy and safety of PD-1 inhibitor + CT with PD-L1 inhibitor + CT. METHODS PubMed, Embase, Web of Science, Cochrane Library, and major international scientific meetings were searched for relevant randomized controlled trials (RCTs), and the indirect analysis was performed for PD-1 + CT vs PD-L1 + CT. The outcomes included progression-free survival (PFS), overall survival (OS), objective response rate (ORR) and treatment-related adverse events (TRAEs). RESULTS 8 phase III RCTs with 4253 patients comparing PD-1/PD-L1 + CT in NSCLC were included. The PD-1 + CT led to notably longer OS most in low/negative expression of PD-L1 for NSCLC patients compared with PD-L1 + CT. In terms of Grade 3-5 TRAEs, the results showed that PD-1 + CT and PD-L1 + CT exclusively increased the risk of adverse incidence than CT alone, especially for PD-L1 + CT (p < 0.00001). For subgroups including female, young patients, patients with nonsmoker, and EGFR/ALK wild-type, PD-1 + CT was associated with prolonged OS (p < 0.05). Meanwhile, for no liver metastasis of NSCLC patients, we found obviously OS advantage for patients treated with PD-1 + CT compared to PD-L1 + CT. CONCLUSIONS ICIs + CT seemed to be more effective first-line regimen and PD-1 + CT could be recommended as the first-rank therapy for advanced NSCLC patients with low/negative expression of PD-L1. However, we should be particularly vigilant about the occurrence of the Grade 3-5 TRAEs.
Collapse
Affiliation(s)
- Y Yang
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Dong Ming Road 127#, Zhengzhou, 450008, China
| | - H Luo
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Dong Ming Road 127#, Zhengzhou, 450008, China
| | - X L Zheng
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Dong Ming Road 127#, Zhengzhou, 450008, China
| | - H Ge
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Dong Ming Road 127#, Zhengzhou, 450008, China.
| |
Collapse
|
29
|
Zhong Y, Wei Q, Lu Y, Tang X, Wang Z, Chen L. Efficacy and safety of anlotinib in patients with advanced non-small cell lung cancer. J Thorac Dis 2020; 12:6016-6022. [PMID: 33209434 PMCID: PMC7656407 DOI: 10.21037/jtd-20-2855] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Non-small cell lung cancer (NSCLC) is the most common type of lung cancer and its incidence seriously affects human health. The purpose of this study was to evaluate the efficacy and safety of anlotinib in patients with advanced NSCLC. Methods A retrospective study was conducted on 150 patients with advanced NSCLC who were treated with anlotinib and discontinued treatment after disease progression or intolerance due to adverse events. Progression-free survival (PFS) of advanced NSCLC patients served as an endpoint. Kaplan-Meier survival curves were applied to evaluate the short-term efficacy of anlotinib treatment in advanced NSCLC patients. Results The median PFS of the whole 150-patient cohort was 5.0 months in (95% CI: 4.00–5.95), 5.0 months (95% CI: 3.0–6.00) in 90 patients with adenocarcinoma, and 4.5 months (95% CI: 4.00–7.00) in 60 patients with squamous cell carcinoma (P=0.676). The PFS was 6.5 months (95% CI: 4.00–8.80) and 4.5 months (95% CI: 4.00–5.60) in the first-/second-line and ≥ third-line patients, respectively (P=0.315). Following the Eastern Cooperative Oncology Group performance status (ECOG PS) score, the median PFS of 95 patients with a PS score 0–1 was 5.5 months (95% CI: 4.50–6.50), and the median PFS of 55 patients with a PS score ntswas 4.0 months (95% CI: 3.00–5.00) (P=0.221). For the 49 patients in the combination group the median PFS was 7.0 months (95% CI: 4.00–9.00), while that of the 101 patients in the anlotinib-alone group was 4.0 months in (95% CI: 2.80–5.50) (P=0.010). In a separate analysis of the combination group, the median PFS of anlotinib combined with chemotherapy, epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI), and immunotherapy was 5.5 months (95% CI: 4.00–9.00), 12.0 months (95% CI: 6.00–12.00), and 6.5 months (95% CI: 4.00–9.80), respectively (P=0.036). Conclusions Anlotinib exhibits good tolerance and performance in prolonging the PFS of patients and has considerable potential as a treatment for advanced NSCLC.
Collapse
Affiliation(s)
- Yuejiao Zhong
- Department of Medical Oncology, the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Qiang Wei
- Department of Ultrasonography, the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - You Lu
- Department of Intervention, the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xiuliang Tang
- Department of Ultrasonography, the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Zhongqiu Wang
- Department of Chest Surgery, the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Lingxiang Chen
- Department of Medical Oncology, the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| |
Collapse
|
30
|
O’Leary C, Gasper H, Sahin KB, Tang M, Kulasinghe A, Adams MN, Richard DJ, O’Byrne KJ. Epidermal Growth Factor Receptor (EGFR)-Mutated Non-Small-Cell Lung Cancer (NSCLC). Pharmaceuticals (Basel) 2020; 13:E273. [PMID: 32992872 PMCID: PMC7600164 DOI: 10.3390/ph13100273] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/18/2020] [Accepted: 09/23/2020] [Indexed: 12/22/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) mutations are the most common oncogenic drivers in non-small-cell lung cancer (NSCLC). Significant developments have taken place which highlight the differences in tumor biology that exist between the mutant and wild-type subtypes of NSCLC. Patients with advanced EGFR-mutant NSCLC have a variety of EGFR-targeting agents available proven to treat their disease. This has led to superior patient outcomes when used as a monotherapy over traditional cytotoxic systemic therapy. Attempts at combining EGFR agents with other anticancer systemic treatment options, such as chemotherapy, antiangiogenic agents, and immunotherapy, have shown varied outcomes. Currently, no specific combination stands out to cause a shift away from the use of single-agent EGFR inhibitors in the first-line setting. Similarly, adjuvant EGFR inhibitors, are yet to significantly add to patient overall survival if used at earlier timepoints in the disease course. Liquid biopsy is an evolving technology with potential promise of being incorporated into the management paradigm of this disease. Data are emerging to suggest that this technique may be capable of identifying early resistance mechanisms and consequential disease progression on the basis of the analysis of blood-based circulating tumor cells.
Collapse
Affiliation(s)
- Connor O’Leary
- Princess Alexandra Hospital, Brisbane 4000, Australia; (H.G.); (K.J.O.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane 4000, Australia; (K.B.S.); (M.T.); (A.K.); (M.N.A.); (D.J.R.)
- Cancer and Ageing Research Program, Translational Research Institute, Brisbane 4000, Australia
| | - Harry Gasper
- Princess Alexandra Hospital, Brisbane 4000, Australia; (H.G.); (K.J.O.)
| | - Katherine B. Sahin
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane 4000, Australia; (K.B.S.); (M.T.); (A.K.); (M.N.A.); (D.J.R.)
- Cancer and Ageing Research Program, Translational Research Institute, Brisbane 4000, Australia
| | - Ming Tang
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane 4000, Australia; (K.B.S.); (M.T.); (A.K.); (M.N.A.); (D.J.R.)
- Cancer and Ageing Research Program, Translational Research Institute, Brisbane 4000, Australia
| | - Arutha Kulasinghe
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane 4000, Australia; (K.B.S.); (M.T.); (A.K.); (M.N.A.); (D.J.R.)
- Cancer and Ageing Research Program, Translational Research Institute, Brisbane 4000, Australia
| | - Mark N. Adams
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane 4000, Australia; (K.B.S.); (M.T.); (A.K.); (M.N.A.); (D.J.R.)
- Cancer and Ageing Research Program, Translational Research Institute, Brisbane 4000, Australia
| | - Derek J. Richard
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane 4000, Australia; (K.B.S.); (M.T.); (A.K.); (M.N.A.); (D.J.R.)
- Cancer and Ageing Research Program, Translational Research Institute, Brisbane 4000, Australia
| | - Ken J. O’Byrne
- Princess Alexandra Hospital, Brisbane 4000, Australia; (H.G.); (K.J.O.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane 4000, Australia; (K.B.S.); (M.T.); (A.K.); (M.N.A.); (D.J.R.)
- Cancer and Ageing Research Program, Translational Research Institute, Brisbane 4000, Australia
| |
Collapse
|
31
|
Kim N, Kim HK, Lee K, Hong Y, Cho JH, Choi JW, Lee JI, Suh YL, Ku BM, Eum HH, Choi S, Choi YL, Joung JG, Park WY, Jung HA, Sun JM, Lee SH, Ahn JS, Park K, Ahn MJ, Lee HO. Single-cell RNA sequencing demonstrates the molecular and cellular reprogramming of metastatic lung adenocarcinoma. Nat Commun 2020; 11:2285. [PMID: 32385277 PMCID: PMC7210975 DOI: 10.1038/s41467-020-16164-1] [Citation(s) in RCA: 661] [Impact Index Per Article: 132.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 04/17/2020] [Indexed: 12/21/2022] Open
Abstract
Advanced metastatic cancer poses utmost clinical challenges and may present molecular and cellular features distinct from an early-stage cancer. Herein, we present single-cell transcriptome profiling of metastatic lung adenocarcinoma, the most prevalent histological lung cancer type diagnosed at stage IV in over 40% of all cases. From 208,506 cells populating the normal tissues or early to metastatic stage cancer in 44 patients, we identify a cancer cell subtype deviating from the normal differentiation trajectory and dominating the metastatic stage. In all stages, the stromal and immune cell dynamics reveal ontological and functional changes that create a pro-tumoral and immunosuppressive microenvironment. Normal resident myeloid cell populations are gradually replaced with monocyte-derived macrophages and dendritic cells, along with T-cell exhaustion. This extensive single-cell analysis enhances our understanding of molecular and cellular dynamics in metastatic lung cancer and reveals potential diagnostic and therapeutic targets in cancer-microenvironment interactions.
Collapse
Affiliation(s)
- Nayoung Kim
- Samsung Genome Institute, Samsung Medical Center, Seoul, 06351, Korea
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul, 06591, Korea
| | - Hong Kwan Kim
- Department of Thoracic and Cardiovascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Kyungjong Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 06351, Seoul, Korea
| | - Yourae Hong
- Samsung Genome Institute, Samsung Medical Center, Seoul, 06351, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences &Technology, Sungkyunkwan University, Seoul, 06355, Korea
| | - Jong Ho Cho
- Department of Thoracic and Cardiovascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Jung Won Choi
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Jung-Il Lee
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Yeon-Lim Suh
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Bo Mi Ku
- Samsung Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Hye Hyeon Eum
- Samsung Genome Institute, Samsung Medical Center, Seoul, 06351, Korea
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul, 06591, Korea
| | - Soyean Choi
- Samsung Genome Institute, Samsung Medical Center, Seoul, 06351, Korea
| | - Yoon-La Choi
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences &Technology, Sungkyunkwan University, Seoul, 06355, Korea
- Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Je-Gun Joung
- Samsung Genome Institute, Samsung Medical Center, Seoul, 06351, Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, 06351, Korea
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences &Technology, Sungkyunkwan University, Seoul, 06355, Korea
| | - Hyun Ae Jung
- Division of Haematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Jong-Mu Sun
- Division of Haematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Se-Hoon Lee
- Division of Haematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Jin Seok Ahn
- Division of Haematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Keunchil Park
- Division of Haematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Myung-Ju Ahn
- Division of Haematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea.
| | - Hae-Ock Lee
- Samsung Genome Institute, Samsung Medical Center, Seoul, 06351, Korea.
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea.
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul, 06591, Korea.
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences &Technology, Sungkyunkwan University, Seoul, 06355, Korea.
| |
Collapse
|
32
|
Non-Coding RNAs in Lung Tumor Initiation and Progression. Int J Mol Sci 2020; 21:ijms21082774. [PMID: 32316322 PMCID: PMC7215285 DOI: 10.3390/ijms21082774] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/07/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
Lung cancer is one of the deadliest forms of cancer affecting society today. Non-coding RNAs, such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), through the transcriptional, post-transcriptional, and epigenetic changes they impose, have been found to be dysregulated to affect lung cancer tumorigenesis and metastasis. This review will briefly summarize hallmarks involved in lung cancer initiation and progression. For initiation, these hallmarks include tumor initiating cells, immortalization, activation of oncogenes and inactivation of tumor suppressors. Hallmarks involved in lung cancer progression include metastasis and drug tolerance and resistance. The targeting of these hallmarks with non-coding RNAs can affect vital metabolic and cell signaling pathways, which as a result can potentially have a role in cancerous and pathological processes. By further understanding non-coding RNAs, researchers can work towards diagnoses and treatments to improve early detection and clinical response.
Collapse
|
33
|
Révész D, Engelhardt EG, Tamminga JJ, Schramel FMNH, Onwuteaka-Philipsen BD, van de Garde EMW, Steyerberg EW, de Vet HC, Coupé VMH. Needs with Regard to Decision Support Systems for Treating Patients with Incurable Non-small Cell Lung Cancer. JOURNAL OF CANCER EDUCATION : THE OFFICIAL JOURNAL OF THE AMERICAN ASSOCIATION FOR CANCER EDUCATION 2020; 35:345-351. [PMID: 30685832 PMCID: PMC7075822 DOI: 10.1007/s13187-019-1471-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Treatment decision-making for patients with incurable non-small cell lung cancer (NSCLC) is complex due to the rapidly increasing number of treatments and discovery of new biomarkers. Decision support systems (DSS) could assist thoracic oncologists (TO) weighing of the pros and cons of treatments in order to arrive at an evidence-based and personalized treatment advice. Our aim is to inventory (1) TO's needs with regard to DSS in the treatment of incurable (stage IIIB/IV) NSCLC patients, and (2) preferences regarding the development of future tools in this field. We disseminated an online inventory questionnaire among all members of the Section of Oncology within the Society of Physicians in Chest Medicine and Tuberculosis. Telephone interviews were conducted to better contextualize the findings from the questionnaire. In total, 58 TO completed the questionnaire and expressed a need for new DSS. They reported that it is important for tools to include genetic and immune markers, to be sufficiently validated, regularly updated, and time-efficient. Also, future DSS should incorporate multiple treatment options, integrate estimates of toxicity, quality of life and cost-effectiveness of treatments, enhance communication between caregivers and patients, and use IT solutions for a clear interface and continuous updating of tools. With this inventory among Dutch TO, we summarized the need for new DSS to aid treatment decision-making for patients with incurable NSCLC. To meet the expressed needs, substantial additional efforts will be required by DSS developers, above already existing tools.
Collapse
Affiliation(s)
- Dóra Révész
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, VU University Medical Center, De Boelelaan 1089a, PO Box 7057, 1081 HV Amsterdam, The Netherlands
| | - Ellen G. Engelhardt
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, VU University Medical Center, De Boelelaan 1089a, PO Box 7057, 1081 HV Amsterdam, The Netherlands
| | - Johannes J. Tamminga
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, VU University Medical Center, De Boelelaan 1089a, PO Box 7057, 1081 HV Amsterdam, The Netherlands
| | - Franz M. N. H. Schramel
- Department of Lung Diseases and Treatment, St. Antonius Hospital, Koekoekslaan 1, 3435 CM Nieuwegein, The Netherlands
| | - Bregje D. Onwuteaka-Philipsen
- Department of Public and Occupational Health, Amsterdam Public Health Research Institute, VU University Medical Center, PO Box 7057, 1081 HV Amsterdam, The Netherlands
| | - Ewoudt M. W. van de Garde
- Department of Clinical Pharmacy, St. Antonius Hospital, Koekoekslaan 1, 3435 CM Nieuwegein, The Netherlands
| | - Ewout W. Steyerberg
- Center for Medical Decision Sciences, Department of Public Health, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Henrica C.W. de Vet
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, VU University Medical Center, De Boelelaan 1089a, PO Box 7057, 1081 HV Amsterdam, The Netherlands
| | - Veerle M. H. Coupé
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, VU University Medical Center, De Boelelaan 1089a, PO Box 7057, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
34
|
Liu C, Zhang H, Li Y, Zhang Z, Shi R, Xu S, Zhu G, Wang P, Liu H, Chen J. [Apatinib Combined with CCI-779 Inhibits the Proliferation and Migration of Small Cell Lung Cancer NCI-H446 Cells In Vitro]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:216-222. [PMID: 32209188 PMCID: PMC7210093 DOI: 10.3779/j.issn.1009-3419.2020.104.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
背景与目的 肺癌是世界上最常见的恶性肿瘤,其中小细胞肺癌是恶性程度最高的亚型,具有生长迅速、早期转移和高度血管化等特点。阿帕替尼(Apatinib)是我国自主研发的血管内皮生长因子受体2抑制剂,在多种实体瘤中疗效显著。本研究旨在探讨Apatinib对小细胞肺癌细胞株NCI-H446的体外作用以及联合哺乳动物雷帕霉素靶蛋白(mammalian target of rapamycin, mTOR)抑制剂CCI-779对小细胞肺癌的体外作用。 方法 体外培养小细胞肺癌细胞株NCI-H446,CCK8法、细胞凋亡实验、细胞周期实验及Transwell实验检测Apatinib及联合mTOR抑制剂CCI-779对NCI-H446细胞增殖、凋亡、周期及迁移的影响;Western blot实验检测血管内皮生长因子受体和细胞周期相关蛋白的表达。 结果 CCK8实验结果显示高浓度Apatinib能抑制NCI-H446细胞增殖;细胞凋亡实验结果显示高浓度Apatinib诱导NCI-H446细胞凋亡;Transwell实验结果显示高浓度Apatinib抑制NCI-H446细胞迁移;联合mTOR抑制剂CCI-779后,低浓度Apatinib便能抑制NCI-H446细胞增殖和迁移,诱导细胞凋亡。 结论 Apatinib对小细胞肺癌细胞株NCI-H446的作用具有浓度依赖性特征,高浓度Apatinib能够抑制NCI-H446细胞增殖和迁移,诱导细胞凋亡,与mTOR抑制剂CCI-779联用能增加NCI-H446细胞对Apatinib的敏感性。
Collapse
Affiliation(s)
- Chao Liu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hongbing Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yongwen Li
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zihe Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ruifeng Shi
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Songlin Xu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Guangsheng Zhu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Pan Wang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hongyu Liu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jun Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
35
|
Qiang H, Chang Q, Xu J, Qian J, Zhang Y, Lei Y, Han B, Chu T. New advances in antiangiogenic combination therapeutic strategies for advanced non-small cell lung cancer. J Cancer Res Clin Oncol 2020; 146:631-645. [PMID: 32065262 DOI: 10.1007/s00432-020-03129-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 01/09/2020] [Indexed: 12/31/2022]
Abstract
PURPOSE Tumor growth relies on the sufficient blood supply and continuously requires new blood vessels to maintain, which lead to vascular abnormalities (Folkman, N Engl J Med 285:1182-1186, 1971). Antiangiogenic therapy has emerged with the goal of normalizing vasculature and tumor microenvironment (TME). Some antiangiogenic therapies combined with chemotherapy, targeted therapy or immunotherapy have been approved for clinical application. In this review, we summarize the recent advances of antiangiogenic combination therapeutic strategies in advanced NSCLC. METHODS References of this review are searched through PubMed and EMBASE and the abstracts of cancer conferences. The ClinicalTrials.gov database was used for relative trials. RESULTS Based on different mechanisms, antiangiogenic agents can be divided into monoclonal antibodies (mAbs), which mainly include bevacizumab and ramucirumab, and multi-target antiangiogenic tyrosine kinase inhibitors (TKIs) which include sunitinib, sorafenib, nintedanib, apatinib, anlotinib, fruquintinib, etc. In recent years, a number of large clinical studies have shown that antiangiogenic agents have conferred a significant overall survival (OS) benefit to patients with advanced non-small cell lung cancer (NSCLC). More and more evidences confirm that the combination of antiangiogenic agents with chemotherapy, targeted therapy and immunotherapy can improve the effect and prolong the survival of NSCLC patients. However, many problems about the application of antiangiogenic agents on advanced NSCLC patients still need to be explored. For example, the combination therapy of multi-target antiangiogenic agents is just beginning, and the biomarkers are not clear. CONCLUSIONS Antiangiogenic agents can achieve therapeutic benefit in advanced NSCLC patients and the combination of chemotherapy, targeted therapy or immunotherapy can lead to synergistic effect. However, exploring the best combination therapy and efficacy-related biomarkers needs further study.
Collapse
Affiliation(s)
- Huiping Qiang
- Department of Respiratory Medicine, Shanghai Chest Hospital, Jiaotong University, Huaihai West Road No. 241, Shanghai, 200030, People's Republic of China
| | - Qing Chang
- Department of Respiratory Medicine, Shanghai Chest Hospital, Jiaotong University, Huaihai West Road No. 241, Shanghai, 200030, People's Republic of China
| | - Jianlin Xu
- Department of Respiratory Medicine, Shanghai Chest Hospital, Jiaotong University, Huaihai West Road No. 241, Shanghai, 200030, People's Republic of China
| | - Jialin Qian
- Department of Respiratory Medicine, Shanghai Chest Hospital, Jiaotong University, Huaihai West Road No. 241, Shanghai, 200030, People's Republic of China
| | - Yanwei Zhang
- Department of Respiratory Medicine, Shanghai Chest Hospital, Jiaotong University, Huaihai West Road No. 241, Shanghai, 200030, People's Republic of China
| | - Yuqiong Lei
- Department of Respiratory Medicine, Shanghai Chest Hospital, Jiaotong University, Huaihai West Road No. 241, Shanghai, 200030, People's Republic of China
| | - Baohui Han
- Department of Respiratory Medicine, Shanghai Chest Hospital, Jiaotong University, Huaihai West Road No. 241, Shanghai, 200030, People's Republic of China.
| | - Tianqing Chu
- Department of Respiratory Medicine, Shanghai Chest Hospital, Jiaotong University, Huaihai West Road No. 241, Shanghai, 200030, People's Republic of China.
| |
Collapse
|
36
|
Systematic Development and Optimization of Inhalable Pirfenidone Liposomes for Non-Small Cell Lung Cancer Treatment. Pharmaceutics 2020; 12:pharmaceutics12030206. [PMID: 32121070 PMCID: PMC7150896 DOI: 10.3390/pharmaceutics12030206] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 12/24/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is a global disorder, treatment options for which remain limited with resistance development by cancer cells and off-target events being major roadblocks for current therapies. The discovery of new drug molecules remains time-consuming, expensive, and prone to failure in safety/efficacy studies. Drug repurposing (i.e., investigating FDA-approved drug molecules for use against new indications) provides an opportunity to shorten the drug development cycle. In this project, we propose to repurpose pirfenidone (PFD), an anti-fibrotic drug, for NSCLC treatment by encapsulation in a cationic liposomal carrier. Liposomal formulations were optimized and evaluated for their physicochemical properties, in-vitro aerosol deposition behavior, cellular internalization capability, and therapeutic potential against NSCLC cell lines in-vitro and ex-vivo. Anti-cancer activity of PFD-loaded liposomes and molecular mechanistic efficacy was determined through colony formation (1.5- to 2-fold reduction in colony growth compared to PFD treatment in H4006, A549 cell lines, respectively), cell migration, apoptosis and angiogenesis assays. Ex-vivo studies using 3D tumor spheroid models revealed superior efficacy of PFD-loaded liposomes against NSCLC, as compared to plain PFD. Hence, the potential of inhalable liposome-loaded pirfenidone in NSCLC treatment has been established in-vitro and ex-vivo, where further studies are required to determine their efficacy through in vivo preclinical studies followed by clinical studies.
Collapse
|
37
|
Ramadan WS, Zaher DM, Altaie AM, Talaat IM, Elmoselhi A. Potential Therapeutic Strategies for Lung and Breast Cancers through Understanding the Anti-Angiogenesis Resistance Mechanisms. Int J Mol Sci 2020; 21:565. [PMID: 31952335 PMCID: PMC7014257 DOI: 10.3390/ijms21020565] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/16/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
Breast and lung cancers are among the top cancer types in terms of incidence and mortality burden worldwide. One of the challenges in the treatment of breast and lung cancers is their resistance to administered drugs, as observed with angiogenesis inhibitors. Based on clinical and pre-clinical findings, these two types of cancers have gained the ability to resist angiogenesis inhibitors through several mechanisms that rely on cellular and extracellular factors. This resistance is mediated through angiogenesis-independent vascularization, and it is related to cancer cells and their microenvironment. The mechanisms that cancer cells utilize include metabolic symbiosis and invasion, and they also take advantage of neighboring cells like macrophages, endothelial cells, myeloid and adipose cells. Overcoming resistance is of great interest, and researchers are investigating possible strategies to enhance sensitivity towards angiogenesis inhibitors. These strategies involved targeting multiple players in angiogenesis, epigenetics, hypoxia, cellular metabolism and the immune system. This review aims to discuss the mechanisms of resistance to angiogenesis inhibitors and to highlight recently developed approaches to overcome this resistance.
Collapse
Affiliation(s)
- Wafaa S. Ramadan
- College of Medicine, University of Sharjah, Sharjah 27272, UAE; (W.S.R.); (D.M.Z.); (A.M.A.); (A.E.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, UAE
| | - Dana M. Zaher
- College of Medicine, University of Sharjah, Sharjah 27272, UAE; (W.S.R.); (D.M.Z.); (A.M.A.); (A.E.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, UAE
| | - Alaa M. Altaie
- College of Medicine, University of Sharjah, Sharjah 27272, UAE; (W.S.R.); (D.M.Z.); (A.M.A.); (A.E.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, UAE
| | - Iman M. Talaat
- College of Medicine, University of Sharjah, Sharjah 27272, UAE; (W.S.R.); (D.M.Z.); (A.M.A.); (A.E.)
- Pathology Department, Faculty of Medicine, Alexandria University, 21526 Alexandria, Egypt
| | - Adel Elmoselhi
- College of Medicine, University of Sharjah, Sharjah 27272, UAE; (W.S.R.); (D.M.Z.); (A.M.A.); (A.E.)
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
38
|
[Chinese Expert Consensus on Antiangiogenic Drugs for Advanced Non-small Cell Lung Cancer (2019 Edition)]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2019; 22:401-412. [PMID: 31315778 PMCID: PMC6712266 DOI: 10.3779/j.issn.1009-3419.2019.07.01] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
39
|
Liang H, Wang M. Prospect of immunotherapy combined with anti-angiogenic agents in patients with advanced non-small cell lung cancer. Cancer Manag Res 2019; 11:7707-7719. [PMID: 31616186 PMCID: PMC6699593 DOI: 10.2147/cmar.s212238] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 07/24/2019] [Indexed: 12/26/2022] Open
Abstract
In the latest years, some drugs have been approved by European Medicines Agency (EMA) and/or the US Food and Drug Administration (FDA) for the treatment of patients with advanced non-small cell lung cancer (NSCLC), particularly for the treatment of those who have no targeted gene mutations or who have progressed on previously targeted therapy or platinum-containing dual-agent chemotherapy. In general, these drugs fall into two categories: anti-angiogenic agents and immune checkpoint inhibitors (ICIs). Anti-angiogenic agents currently approved by the FDA and/or EMA for advanced NSCLC treatment include bevacizumab, nintedanib, and ramucirumab. Anlotinib has been approved in advanced NSCLC by Chinese Food and Drug Administration (CFDA). These anti-angiogenic agents can induce anti-angiogenesis by targeting vascular endothelial growth factor (VEGF)/VEGF2 or inhibiting multiple small molecules involved in angiogenic and proliferative pathways such as platelet-derived growth factor receptors (PDGFRs) and fibroblast growth factor receptors (FGFRs). Although these drugs show significant therapeutic efficacy, most patients inevitably experience disease progression resulting in death. ICIs approved by the FDA and/or EMA for advanced NSCLC treatment include nivolumab, pembrolizumab, and atezolizumab. These ICIs can significantly improve efficacy compared with standard chemotherapy by targeting programmed cell death protein 1 (PD-1) receptor or PD-2 receptor with longer response duration and acceptable toxicity. However, the response rate of ICIs is suboptimal, and only a few patients ultimately benefit from immunotherapy. So current efforts have focused on exploring new potential combinatorial strategies with synergistic antitumor activity. Here, we summarized the theoretical basis, current clinical data, and potential future perspective of immunotherapy combined with anti-angiogenic agents for advanced NSCLC.
Collapse
Affiliation(s)
- Hongge Liang
- Lung Cancer Center, Department of Respiratory Medicine, Peking Union Medical College Hospital, Beijing, 100730, People's Republic of China
| | - Mengzhao Wang
- Lung Cancer Center, Department of Respiratory Medicine, Peking Union Medical College Hospital, Beijing, 100730, People's Republic of China
| |
Collapse
|
40
|
Shao L, Wang W, Song Z, Zhang Y. The efficacy and safety of anlotinib treatment for advanced lung cancer. Onco Targets Ther 2019; 12:6549-6554. [PMID: 31616163 PMCID: PMC6699585 DOI: 10.2147/ott.s205674] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 07/21/2019] [Indexed: 02/06/2023] Open
Abstract
Objective Anlotinib is an oral novel multi-target tyrosine kinase inhibitor targeting vascular endothelial growth factor receptor, fibroblast growth factor receptor, platelet-derived growth factor receptor, and stem cell factor receptor (c-Kit). The aim of this study was to evaluate the efficacy and safety of anlotinib treatment in advanced lung cancer in the real world. Methods We evaluated the efficacy and toxicity of apatinib in patients with previously treated advanced lung cancer from 2018 to 2019 in Zhejiang Cancer Hospital. Survival analysis was performed by the Kaplan-Meier method. Results Fifty-eight patients were included in the present study. Thirty-one of these patients received anlotinib treatment as a third line and 27 patients received further therapy. All 58 patients had therapeutic evaluation and 46 patients acquired progression-free survival evaluation. Ten patients achieved partial response (PR), and 36 achieved stable disease (SD), representing a response rate of 17.2% and a disease control rate of 77.6%. Median progression-free survival was 3.3 months (95% CI 1.595-5.071). The toxicities associated with anlotinib were generally acceptable with a total grade 3/4 toxicity of 5.2%. The toxicities of anlotinib were generally tolerated and the common toxicities were hand-foot syndrome and hypertension. Conclusion In the third-line or more-line treatment of advanced lung cancer, anlotinib appears to have some activity when utilized as a salvage treatment. Adverse reactions are controllable.
Collapse
Affiliation(s)
- Lan Shao
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China
| | - Wenxian Wang
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China
| | - Zhengbo Song
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China
| | - Yiping Zhang
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
41
|
Li F, Wang Y, Chen WL, Wang DD, Zhou YJ, You BG, Liu Y, Qu CX, Yang SD, Chen MT, Zhang XN. Co-delivery of VEGF siRNA and Etoposide for Enhanced Anti-angiogenesis and Anti-proliferation Effect via Multi-functional Nanoparticles for Orthotopic Non-Small Cell Lung Cancer Treatment. Am J Cancer Res 2019; 9:5886-5898. [PMID: 31534526 PMCID: PMC6735374 DOI: 10.7150/thno.32416] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/25/2019] [Indexed: 12/13/2022] Open
Abstract
Targeting tumor angiogenesis pathway via VEGF siRNA (siVEGF) has shown great potential in treating highly malignant and metastatic non-small cell lung cancer (NSCLC). However, anti-angiogenic monotherapy lacked sufficient antitumor efficacy which suffered from malignant tumor proliferation. Therefore, the combined application of siVEGF and chemotherapeutic agents for simultaneous targeting of tumor proliferation and angiogenesis has been a research hotspot to explore a promising NSCLC therapy regimen. Methods: We designed, for the first time, a rational therapy strategy via intelligently co-delivering siVEGF and chemotherapeutics etoposide (ETO) by multi-functional nanoparticles (NPs) directed against the orthotopic NSCLC. These NPs consisted of cationic liposomes loaded with siVEGF and ETO and then coated with versatile polymer PEGylated histidine-grafted chitosan-lipoic acid (PHCL). We then comprehensively evaluated the anti-angiogenic and anti-proliferation efficiency in the in vitro tumor cell model and in bioluminescent orthotopic lung tumor bearing mice model. Results: The NPs co-delivering siVEGF and ETO exhibited tailor-made surface charge reversal features in mimicking tumor extracellular environment with improved internal tumor penetration capacity and higher cellular internalization. Furthermore, these NPs with flexible particles size triggered by intracellular acidic environment and redox environment showed pinpointed and sharp intracellular cargo release guaranteeing adequate active drug concentration in tumor cells. Enhanced VEGF gene expression silencing efficacy and improved tumor cell anti-proliferation effect were demonstrated in vitro. In addition, the PHCL layer improved the stability of these NPs in neutral environment allowing enhanced orthotopic lung tumor targeting efficiency in vivo. The combined therapy by siVEGF and ETO co-delivered NPs for orthotopic NSCLC simultaneously inhibited tumor proliferation and tumor angiogenesis resulting in more significant suppression of tumor growth and metastasis than monotherapy. Conclusion: Combined application of siVEGF and ETO by the multi-functional NPs with excellent and on-demand properties exhibited the desired antitumor effect on the orthotopic lung tumor. Our work has significant potential in promoting combined anti-angiogenesis therapy and chemotherapy regimen for clinical NSCLC treatment.
Collapse
|
42
|
Wu D, Nie J, Dai L, Hu W, Zhang J, Chen X, Ma X, Tian G, Han J, Han S, Long J, Wang Y, Zhang Z, Fang J. Salvage treatment with anlotinib for advanced non-small cell lung cancer. Thorac Cancer 2019; 10:1590-1596. [PMID: 31183998 PMCID: PMC6610258 DOI: 10.1111/1759-7714.13120] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 12/26/2022] Open
Abstract
Background This real‐world study assessed the efficacy and toxicity of anlotinib as salvage treatment in Chinese patients with advanced non‐small cell lung cancer (NSCLC). Methods The medical records of 81 patients with advanced NSCLC who had failed at least two lines of chemotherapy were retrospectively collected. All patients were administered anlotinib treatment until disease progression or intolerance as a result of adverse events. Survival curves were created using the Kaplan–Meier method. The log‐rank test was used for univariate analysis of progression‐free survival (PFS) between groups. Cox regression was used to estimate the statistically significant factors based on univariate analysis. Results The median PFS was five months (95% confidence interval [CI] 3.5–6.5). The objective response rate (ORR) was 7% and the disease control rate (DCR) was 84%. The following subgroups of patients had longer PFS (P < 0.05): squamous cell carcinoma, no brain or liver metastases, Eastern Cooperative Oncology Group performance status (ECOG PS) of 0–1, and no previous VEGF‐tyrosine kinase inhibitor treatment. The results of Cox regression indicated that an ECOG PS of 0–1 (hazard ratio 0.152, 95% CI 0.057–0.403; P = 0.00) and patients without brain metastases (hazard ratio 0.421, 95% CI 0.195–0.911; P = 0.028) had longer PFS following anlotinib treatment. Conclusion Anlotinib, which is well tolerated, plays a significant role in the salvage treatment of advanced NSCLC. Patients with advanced NSCLC with an ECOG PS of 0–1 and no brain metastases achieved longer PFS following anlotinib salvage treatment.
Collapse
Affiliation(s)
- Di Wu
- Department of Thoracic Oncology II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jun Nie
- Department of Thoracic Oncology II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Ling Dai
- Department of Thoracic Oncology II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Weiheng Hu
- Department of Thoracic Oncology II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jie Zhang
- Department of Thoracic Oncology II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaoling Chen
- Department of Thoracic Oncology II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiangjuan Ma
- Department of Thoracic Oncology II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Guangming Tian
- Department of Thoracic Oncology II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jindi Han
- Department of Thoracic Oncology II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Sen Han
- Department of Thoracic Oncology II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jieran Long
- Department of Thoracic Oncology II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yang Wang
- Department of Thoracic Oncology II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Ziran Zhang
- Department of Thoracic Oncology II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jian Fang
- Department of Thoracic Oncology II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
43
|
Jiang F, Yu Q, Chu Y, Zhu X, Lu W, Liu Q, Wang Q. MicroRNA-98-5p inhibits proliferation and metastasis in non-small cell lung cancer by targeting TGFBR1. Int J Oncol 2018; 54:128-138. [PMID: 30387848 PMCID: PMC6255066 DOI: 10.3892/ijo.2018.4610] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/09/2018] [Indexed: 12/15/2022] Open
Abstract
MicroRNAs (miRNAs or miRs) have recently emerged as key regulators of various types of cancer, including non‑small cell lung cancer (NSCLC). The disrupted expression of miRNAs is associated with tumorigenesis and metastasis; however, the underlying mechanisms remain unclear. In this study, we demonstrate that miR‑98‑5p is downregulated in NSCLC and that miR‑98‑5p deficiency is associated with an advanced clinical stage and metastasis. A dual‑luciferase reporter assay was performed to confirm that transforming growth factor beta receptor 1 (TGFBR1), a key stimulator of tumor proliferation and metastasis, was a direct target of miR‑98‑5p. miR‑98‑5p overexpression resulted in the downregulation of TGFBR1 and the suppression of the viability, proliferation, migration and invasion of A549 and H1299 cells. Furthermore, miR‑98‑5p was demonstrated to be an efficient suppressor of tumor growth in an A549 subcutaneous xenograft tumor mouse model. Finally, miR‑98‑5p overexpression exerted a significant anti‑metastatic effect in a mouse model of pulmonary metastasis. On the whole, the results of the present study suggest that miR‑98‑5p/TGFBR1 may serve as promising targets for NSCLC therapy.
Collapse
Affiliation(s)
- Feng Jiang
- Department of Cardiothoracic Surgery, Wujin People's Hospital of Changzhou, Changzhou, Jiangsu 213017, P.R. China
| | - Qiuhua Yu
- Department of Cardiothoracic Surgery, Wujin People's Hospital of Changzhou, Changzhou, Jiangsu 213017, P.R. China
| | - Ying Chu
- Central Laboratory, Wujin People's Hospital of Changzhou, Changzhou, Jiangsu 213017, P.R. China
| | - Xiaobo Zhu
- Department of Cardiothoracic Surgery, Wujin People's Hospital of Changzhou, Changzhou, Jiangsu 213017, P.R. China
| | - Wenbin Lu
- Department of Oncology, Wujin People's Hospital of Changzhou, Changzhou, Jiangsu 213017, P.R. China
| | - Qian Liu
- Department of Oncology, Wujin People's Hospital of Changzhou, Changzhou, Jiangsu 213017, P.R. China
| | - Qiang Wang
- Department of Cardiothoracic Surgery, Wujin People's Hospital of Changzhou, Changzhou, Jiangsu 213017, P.R. China
| |
Collapse
|
44
|
Miller HA, Frieboes HB. Evaluation of Drug-Loaded Gold Nanoparticle Cytotoxicity as a Function of Tumor Vasculature-Induced Tissue Heterogeneity. Ann Biomed Eng 2018; 47:257-271. [PMID: 30298374 DOI: 10.1007/s10439-018-02146-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 10/01/2018] [Indexed: 01/10/2023]
Abstract
The inherent heterogeneity of tumor tissue presents a major challenge to nanoparticle-mediated drug delivery. This heterogeneity spans from the molecular (genomic, proteomic, metabolomic) to the cellular (cell types, adhesion, migration) and to the tissue (vasculature, extra-cellular matrix) scales. In particular, tumor vasculature forms abnormally, inducing proliferative, hypoxic, and necrotic tumor tissue regions. As the vasculature is the main conduit for nanotherapy transport into tumors, vasculature-induced tissue heterogeneity can cause local inadequate delivery and concentration, leading to subpar response. Further, hypoxic tissue, although viable, would be immune to the effects of cell-cycle specific drugs. In order to enable a more systematic evaluation of such effects, here we employ computational modeling to study the therapeutic response as a function of vasculature-induced tumor tissue heterogeneity. Using data with three-layered gold nanoparticles loaded with cisplatin, nanotherapy is simulated interacting with different levels of tissue heterogeneity, and the treatment response is measured in terms of tumor regression. The results quantify the influence that varying levels of tumor vascular density coupled with the drug strength have on nanoparticle uptake and washout, and the associated tissue response. The drug strength affects the proportion of proliferating, hypoxic, and necrotic tissue fractions, which in turn dynamically affect and are affected by the vascular density. Higher drug strengths may be able to achieve stronger tumor regression but only if the intra-tumoral vascular density is above a certain threshold that affords sufficient transport. This study establishes an initial step towards a more systematic methodology to assess the effect of vasculature-induced tumor tissue heterogeneity on the response to nanotherapy.
Collapse
Affiliation(s)
- Hunter A Miller
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Hermann B Frieboes
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA. .,Department of Bioengineering, University of Louisville, Lutz Hall 419, Louisville, KY, 40292, USA. .,James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
45
|
Chen X, Zheng Q, Li W, Lu Y, Ni Y, Ma L, Fu Y. SOX5 induces lung adenocarcinoma angiogenesis by inducing the expression of VEGF through STAT3 signaling. Onco Targets Ther 2018; 11:5733-5741. [PMID: 30254466 PMCID: PMC6140741 DOI: 10.2147/ott.s176533] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background and objectives Angiogenesis is the main cause of lung adenocarcinoma (LAC) poor prognosis. This study aimed to investigate the effect of sex-determining region Y-box protein 5 (SOX5) expression on angiogenesis of LAC and explore its possible mechanism. Patients and methods The effect on angiogenesis was tested by tube formation assays using human umbilical vein endothelial cells cocultured with A549 cells. Lentivirus shRNA of SOX5 and lentivirus of SOX5 overexpression system were used to establish LAC cell lines, which expressed SOX5 of different levels. SOX5 downstream signaling targets were analyzed by real-time qPCR and Western blot. We collected 90 LAC cases and the tissues were examined by immunohistochemistry for SOX5 and vascular endothelial growth factor (VEGF). Results We found that SOX5 overexpression in A549 cells significantly promoted tube formation capacity of the cocultured human umbilical vein endothelial cells. SOX5 increased VEGF expression and signal transducer activator of transcription 3 phosphorylation; however, SOX5 had no effect on extracellular signal-regulated kinase and protein kinase B pathway. Furthermore, the expression of SOX5 and VEGF had a significantly positive correlation (r=0.399, P=0.001) according to the tissue microarray data. Conclusion These findings suggest that SOX5 induces angiogenesis by activating signal transducer activator of transcription 3/VEGF signaling and confer its candidacy as a potential therapeutic target in LAC.
Collapse
Affiliation(s)
- Xin Chen
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310003, P. R. China,
| | - Qi Zheng
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310003, P. R. China,
| | - Weidong Li
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310003, P. R. China,
| | - Yuan Lu
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310003, P. R. China,
| | - Yiming Ni
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310003, P. R. China,
| | - Liang Ma
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310003, P. R. China,
| | - Yufei Fu
- Zhejiang Key Laboratory of Gastro-Intestinal Pathophysiology, Zhejiang Hospital of Traditional Chinese Medicine, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P. R. China,
| |
Collapse
|
46
|
Cui J, Zhang Y, Su D, Li T, Li Y. Efficacy of combined icotinib and pemetrexed in EGFR mutant lung adenocarcinoma cell line xenografts. Thorac Cancer 2018; 9:1156-1165. [PMID: 30047610 PMCID: PMC6119608 DOI: 10.1111/1759-7714.12818] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/21/2018] [Accepted: 06/21/2018] [Indexed: 12/31/2022] Open
Abstract
Background The combination of EGFR tyrosine kinase inhibitors (TKIs) and chemotherapy is thought to increase treatment efficacy in non‐small‐cell lung cancer (NSCLC). This study investigated the efficacy and potential mechanisms of different combined modes of icotinib plus pemetrexed in EGFR‐mutant lung adenocarcinoma cell line xenograft models. Methods Nude mice were subcutaneously injected with EGFR‐mutant human lung adenocarcinoma cells (HCC827) and randomized into six treatment groups. Tumor xenograft volumes were monitored and recorded. Microvessel density (MVD) and proliferation and apoptosis rates were evaluated with CD34 positive cell counting, and Ki‐67 and caspase‐3 scores, respectively, and determined via immunohistochemistry. Thymidylate synthase (TS), EGFR, and downstream signaling molecule expression was detected by Western blotting. Results The volume and weight of tumor xenografts in the sequential pemetrexed followed by icotinib (Pem‐Ico) group and the concurrent icotinib and pemetrexed (Ico + Pem) group were significantly smaller than those in the control, pemetrexed (Pem), icotinib (Ico), and sequential icotinib followed by pemetrexed (Ico‐Pem) groups. Compared to other groups, a decrease in the MVD and proliferation rate and an increase in the apoptosis rate were observed in the Pem‐Ico and Ico + Pem groups. TS expression and EGFR, AKT, and MAPK phosphorylation were significantly reduced in the Pem‐Ico or Ico + Pem groups. Conclusions Pem‐Ico had additive antitumor activity in vivo, similar to Ico + Pem, both of which are suggested as potentially optimized strategies for treating EGFR‐mutant lung adenocarcinoma.
Collapse
Affiliation(s)
- Jiadong Cui
- Department of Respiratory Medicine, Qilu Hospital of Shandong University, Jinan, China.,Department of Respiratory Medicine, Dong'e County People's Hospital, Liaocheng, China
| | - Yan Zhang
- Department of Respiratory Medicine, Jinan No. 4 People's Hospital, Jinan, China
| | - Di Su
- Department of Respiratory Medicine, Dong'e County People's Hospital, Liaocheng, China
| | - Tao Li
- Department of Respiratory Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Yu Li
- Department of Respiratory Medicine, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
47
|
Rastegar M, Marjani HA, Yazdani Y, Shahbazi M, Golalipour M, Farazmandfar T. Investigating Effect of Rapamycin and Metformin on Angiogenesis in Hepatocellular Carcinoma Cell Line. Adv Pharm Bull 2018; 8:63-68. [PMID: 29670840 PMCID: PMC5899784 DOI: 10.15171/apb.2018.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 02/08/2018] [Accepted: 02/10/2018] [Indexed: 12/12/2022] Open
Abstract
Purpose: Human hepatocellular carcinoma is one of the most common causes of death in the world. Metformin and rapamycin may decrease the expression of VEGF protein and subsequently angiogenesis. The purpose of this study was to evaluate the effect of these two drugs on expression of VEGF protein and the cell proliferation in the hepatocellular carcinoma cell line (ATCC HB-8065). Methods: HepG2 was cultured in RPMI-1640 medium at 37°C for 48h as a pre-culture and then treated by different concentrations of metformin (0, 5, 10 and 20 mM) and rapamycin (0, 5, 10 and 20 nM) at different times (12, 24 and 48 h). Cell viability was assessed by the MTT assay. Total RNA was extracted by the Trizol reagent and VEGF gene expression was analyzed by quantitative real-time PCR and was calculated by 2–ΔCt method. The VEGF protein level was determined by Elisa assay. Finally, Apoptosis index was calculated by DAPI staining. Results: Metformin and rapamycin significantly decrease cancer cells viability (p<0.05). Rapamycin but not metformin decreases VEGF gene expression in HepG2 cells. Metformin and rapamycin significantly induce cell apoptosis in hepatocellular carcinoma (HCC) cells. Conclusion: Metformin and rapamycin have an anti-tumor effect on HCC. According to our data rapamycin might have an anti-angiogenesis effect via inhibition of VEGF expression. Our results provide an insight into future clinical strategies to improve chemotherapy outcomes in HCC.
Collapse
Affiliation(s)
- Mandana Rastegar
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Haji-Amin Marjani
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Yaghoub Yazdani
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Shahbazi
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Masoud Golalipour
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Touraj Farazmandfar
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
48
|
Xu X, Zhu S, Tao Z, Ye S. High circulating miR-18a, miR-20a, and miR-92a expression correlates with poor prognosis in patients with non-small cell lung cancer. Cancer Med 2018; 7:21-31. [PMID: 29266846 PMCID: PMC5773999 DOI: 10.1002/cam4.1238] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 09/14/2017] [Accepted: 09/23/2017] [Indexed: 12/16/2022] Open
Abstract
The purpose of this study was to assess the predictive value of angiogenic miRNAs for disease-free survival (DFS) and overall survival (OS) of patients with non-small cell lung cancer (NSCLC). In total, 196 patients with NSCLC (tumor lymph nodes metastasis (TNM) stage I-III) were enrolled and peripheral blood samples were collected. Total RNA was extracted from blood samples, and the relative expression levels of candidate miRNAs were evaluated by real time-polymerase chain reaction (RT-PCR). The median follow-up period was 56.7 months, and the final follow-up date was in August 2016. The median DFS of all patients was 30.0 (14.0-49.0) months, whereas the median OS was 41.5 (23.0-58.0) months. Furthermore, the 5-year DFS and OS rates were 11.3% and 32.3%, respectively. Kaplan-Meier (K-M) curves showed that high plasma miR-18a (P < 0.001), miR-20a (P < 0.001), miR-92a (P < 0.001), miR-126 (P < 0.001), miR-210 (P = 0.003), and miR-19a (P = 0.027) expressions levels correlated with a worse DFS. Moreover, patients with high plasma miR-18a, miR-20a, miR-92a, miR-210, and miR-126 expression levels had a shorter OS than patients with low expression levels of these miRNAs (all P <= 0.001). Furthermore, multivariate Cox regression analyses revealed that high plasma expression levels of miR-18a, miR-20a, and miR-92a as well as lymphatic node metastasis (all P < 0.001) were independent risk factors for both DFS and OS in patients with NSCLC. Thus, the circulating miR-18a, miR-20a, and miR-92a levels may serve as novel and promising prognostic biomarkers in patients with NSCLC.
Collapse
Affiliation(s)
- Xiaoxiao Xu
- Department of PneumologyThe Central Hospital of WuhanTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430014China
| | - Shan Zhu
- Department of PneumologyThe Central Hospital of WuhanTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430014China
| | - Zhaowu Tao
- Department of PneumologyThe Central Hospital of WuhanTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430014China
| | - Shenglan Ye
- Department of PneumologyThe Central Hospital of WuhanTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430014China
| |
Collapse
|
49
|
Gomes M, Archer F, Girard N, Gineys B, Dolmazon C, Bobet Erny A, Mornex JF, Leroux C. Blocked expression of key genes of the angiogenic pathway in JSRV-induced pulmonary adenocarcinomas. Vet Res 2017; 48:76. [PMID: 29137669 PMCID: PMC5686813 DOI: 10.1186/s13567-017-0480-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/20/2017] [Indexed: 02/06/2023] Open
Abstract
JSRV (Jaagsiekte Sheep Retrovirus) is a retrovirus inducing a transmissible lung adenocarcinoma in sheep and goats with predominantly lepidic and papillary lesions. This naturally occurring lung cancer in large animals shares many features with human pneumonic-type lung adenocarcinomas with predominant lepidic growth. The metastatic spread is rare in both human and animal cancers. This unique feature prompted us to decipher the angiogenesis pathway in these cancers. We focused on the levels of mRNA and proteins of genes implicated in the extension of JSRV-induced lung adenocarcinomas by studying their expression in lung cancers (n = 10) and normal lungs (n = 10) and in primary epithelial alveolar type II cells derived from cancers (n = 10) or normal lungs (n = 6). In parallel, we evaluated the levels of expression of key genes in lung tissues collected from lepidic (n = 13) or papillary (n = 5) human adenocarcinomas and, when available, adjacent normal lungs (n = 11). We measured the expression of the same key genes implicated in angiogenesis, lymphangiogenesis and degradation of the extracellular matrix. In ovine adenocarcinomas, VEGFR2 and VEGFD mRNA were downregulated in cancers; MMP9, TIMP1 and FGFR2 mRNA were overexpressed as compared to normal lungs. Importantly, VEGFA and VEGFR2 proteins were not expressed in JSRV-induced cancers. In human lepidic adenocarcinomas, VEGFA and VEGFR2 mRNA were weakly expressed and no VEGFR2 protein was detectable. Downregulation of key angiogenic players may contribute to the control of extra thoracic invasion of cancer cells in human and ovine pneumonic-type adenocarcinoma with predominant lepidic growth.
Collapse
Affiliation(s)
- Maryline Gomes
- IVPC UMR754, INRA, Univ Lyon, Université Claude Bernard Lyon 1, EPHE, Lyon, France
| | - Fabienne Archer
- IVPC UMR754, INRA, Univ Lyon, Université Claude Bernard Lyon 1, EPHE, Lyon, France
| | - Nicolas Girard
- IVPC UMR754, INRA, Univ Lyon, Université Claude Bernard Lyon 1, EPHE, Lyon, France.,Department of Respiratory Diseases, Hospices Civils de Lyon, Louis Pradel Hospital, Lyon, France
| | - Barbara Gineys
- IVPC UMR754, INRA, Univ Lyon, Université Claude Bernard Lyon 1, EPHE, Lyon, France
| | - Christine Dolmazon
- IVPC UMR754, INRA, Univ Lyon, Université Claude Bernard Lyon 1, EPHE, Lyon, France
| | - Alexandra Bobet Erny
- IVPC UMR754, INRA, Univ Lyon, Université Claude Bernard Lyon 1, EPHE, Lyon, France
| | - Jean-François Mornex
- IVPC UMR754, INRA, Univ Lyon, Université Claude Bernard Lyon 1, EPHE, Lyon, France.,Department of Respiratory Diseases, Hospices Civils de Lyon, Louis Pradel Hospital, Lyon, France
| | - Caroline Leroux
- IVPC UMR754, INRA, Univ Lyon, Université Claude Bernard Lyon 1, EPHE, Lyon, France.
| |
Collapse
|
50
|
Manzo A, Montanino A, Carillio G, Costanzo R, Sandomenico C, Normanno N, Piccirillo MC, Daniele G, Perrone F, Rocco G, Morabito A. Angiogenesis Inhibitors in NSCLC. Int J Mol Sci 2017; 18:E2021. [PMID: 28934120 PMCID: PMC5666703 DOI: 10.3390/ijms18102021] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/13/2017] [Accepted: 09/15/2017] [Indexed: 01/04/2023] Open
Abstract
Angiogenesis is a complex biological process that plays a relevant role in sustaining the microenvironment, growth, and metastatic potential of several tumors, including non-small cell lung cancer (NSCLC). Bevacizumab was the first angiogenesis inhibitor approved for the treatment of patients with advanced NSCLC in combination with chemotherapy; however, it was limited to patients with non-squamous histology and first-line setting. Approval was based on the results of two phase III trials (ECOG4599 and AVAIL) that demonstrated an improvement of about two months in progression-free survival (PFS) in both trials, and in the ECOG4599 trial, an improvement in overall survival (OS) also. Afterwards, other antiangiogenic agents, including sunitinib, sorafenib, and vandetanib have been unsuccessfully tested in first and successive lines. Recently, two new antiangiogenic agents (ramucirumab and nintedanib) produced a significant survival benefit in second-line setting. In the REVEL study, ramucirumab plus docetaxel prolonged the median OS of patients with any histology NSCLC when compared with docetaxel alone (10.4 versus 9.1 months, hazard ratio (HR) 0.857, p = 0.0235). In the LUME-Lung 1 study, nintedanib plus docetaxel prolonged the median PFS of patients with any tumor histology (p = 0.0019), and improved OS (12.6 versus 10.3 months) in patients with adenocarcinoma. As a result, it became a new option for the second-line treatment of patients with advanced NSCLC and adenocarcinoma histology. Identifying predictive biomarkers to optimize the benefit of antiangiogenic drugs remains an ongoing challenge.
Collapse
MESH Headings
- Adenocarcinoma/drug therapy
- Adenocarcinoma/metabolism
- Adenocarcinoma/mortality
- Adenocarcinoma/pathology
- Angiogenesis Inhibitors/therapeutic use
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Bevacizumab/therapeutic use
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/pathology
- Clinical Trials, Phase III as Topic
- Disease-Free Survival
- Docetaxel
- Humans
- Indoles/therapeutic use
- Lung Neoplasms/drug therapy
- Lung Neoplasms/metabolism
- Lung Neoplasms/mortality
- Lung Neoplasms/pathology
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/mortality
- Neovascularization, Pathologic/pathology
- Niacinamide/analogs & derivatives
- Niacinamide/therapeutic use
- Phenylurea Compounds/therapeutic use
- Piperidines/therapeutic use
- Pyrroles/therapeutic use
- Quinazolines/therapeutic use
- Sorafenib
- Sunitinib
- Taxoids/therapeutic use
- Ramucirumab
Collapse
Affiliation(s)
- Anna Manzo
- Thoracic Medical Oncology, Istituto Nazionale Tumori, "Fondazione G.Pascale"-IRCCS, 80131 Napoli, Italy.
| | - Agnese Montanino
- Thoracic Medical Oncology, Istituto Nazionale Tumori, "Fondazione G.Pascale"-IRCCS, 80131 Napoli, Italy.
| | - Guido Carillio
- Department of Oncology and Hematology, Azienda Ospedaliera Pugliese-Ciaccio, 88100 Catanzaro, Italy.
| | - Raffaele Costanzo
- Thoracic Medical Oncology, Istituto Nazionale Tumori, "Fondazione G.Pascale"-IRCCS, 80131 Napoli, Italy.
| | - Claudia Sandomenico
- Thoracic Medical Oncology, Istituto Nazionale Tumori, "Fondazione G.Pascale"-IRCCS, 80131 Napoli, Italy.
| | - Nicola Normanno
- Cellular Biology and Biotherapy, Research Department, Istituto Nazionale Tumori "Fondazione G.Pascale"-IRCCS, Napoli 80131, Italy.
| | - Maria Carmela Piccirillo
- Clinical Trials Unit, Istituto Nazionale Tumori, "Fondazione G.Pascale"-IRCCS, 80131 Napoli, Italy.
| | - Gennaro Daniele
- Clinical Trials Unit, Istituto Nazionale Tumori, "Fondazione G.Pascale"-IRCCS, 80131 Napoli, Italy.
| | - Francesco Perrone
- Clinical Trials Unit, Istituto Nazionale Tumori, "Fondazione G.Pascale"-IRCCS, 80131 Napoli, Italy.
| | - Gaetano Rocco
- Thoracic Surgery, Istituto Nazionale Tumori, "Fondazione G.Pascale"-IRCCS, 80131 Napoli, Italy.
| | - Alessandro Morabito
- Thoracic Medical Oncology, Istituto Nazionale Tumori, "Fondazione G.Pascale"-IRCCS, 80131 Napoli, Italy.
| |
Collapse
|