1
|
Alam SK, Pandit A, Wang L, Mortazavi Farsani SS, Thiele BA, Manoj P, Aubry MC, Verma V, Rudin CM, Lo YC, Hoeppner LH. Dopamine D 2 receptor agonists abrogate neuroendocrine tumour angiogenesis to inhibit chemotherapy-refractory small cell lung cancer progression. Cell Death Dis 2025; 16:370. [PMID: 40346068 DOI: 10.1038/s41419-025-07693-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/11/2025]
Abstract
Small cell lung cancer (SCLC) is difficult to treat due to its aggressiveness, early metastasis, and rapid development of resistance to chemotherapeutic agents. Here, we show that treatment with a dopamine D2 receptor (D2R) agonist reduces tumour angiogenesis in multiple in vivo xenograft models of human SCLC, thereby reducing SCLC progression. An FDA-approved D2R agonist, cabergoline, also sensitized chemotherapy-resistant SCLC tumours to cisplatin and etoposide in patient-derived xenograft models of acquired chemoresistance in mice. Ex vivo, D2R agonist treatment decreased tumour angiogenesis through increased apoptosis of tumour-associated endothelial cells, creating a less favourable tumour microenvironment that limited cancer cell proliferation. In paired SCLC patient-derived specimens, D2R was expressed by tumour-associated endothelial cells obtained before treatment, but D2R was downregulated in SCLC tumours that had acquired chemoresistance. D2R agonist treatment of chemotherapy-resistant specimens restored expression of D2R. Activation of dopamine signalling is thus a new strategy for inhibiting angiogenesis in SCLC and potentially for combatting chemotherapy-refractory SCLC progression.
Collapse
Affiliation(s)
- Sk Kayum Alam
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Anuradha Pandit
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Li Wang
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | | | - Britteny A Thiele
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Parvathy Manoj
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marie Christine Aubry
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Vivek Verma
- The Hormel Institute, University of Minnesota, Austin, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ying-Chun Lo
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Luke H Hoeppner
- The Hormel Institute, University of Minnesota, Austin, MN, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
2
|
Zhang T, Tao L, Chen Y, Zhang S, Liu Y, Li Y, Wang R. Evaluation of Efficacy and Safety in First-Line Treatment Methods for Extensive-Stage Small Cell Lung Cancer: A Comprehensive Comparative Study of Chemotherapy, Targeted Therapy Combined With Chemotherapy, and Immunotherapy Combined With Chemotherapy. THE CLINICAL RESPIRATORY JOURNAL 2024; 18:e13819. [PMID: 39118429 PMCID: PMC11310407 DOI: 10.1111/crj.13819] [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: 11/03/2023] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Small cell lung cancer (SCLC) is a highly aggressive tumor with limited effectiveness in its standard chemotherapy treatment. Targeted antiangiogenic therapy and immune checkpoint inhibitors (ICIs) have demonstrated potential as alternative treatments for extensive-stage SCLC (ES-SCLC). However, there is insufficient comparative evidence available to determine the optimal first-line treatment option between ICIs plus chemotherapy and targeted antiangiogenic therapy plus chemotherapy. OBJECTIVE This study is aimed at analyzing clinical data from ES-SCLC patients treated at the First Affiliated Hospital of Bengbu Medical College between June 2021 and June 2023. The study compared the efficacy and safety of three first-line treatment regimens: standard chemotherapy, antiangiogenic therapy combined with chemotherapy, and immune combination therapy. METHODS Patients who met the inclusion criteria were divided into three groups: chemotherapy, immune combination therapy, and antiangiogenic therapy combined with chemotherapy. The study collected data on clinical characteristics, treatment regimens, and adverse reactions. The analysis included objective response rate (ORR), duration of response (DoR), disease control rate (DCR), progression-free survival (PFS), and treatment safety. RESULTS A total of 101 patients were included in the study, with 49 receiving chemotherapy alone, 19 receiving antiangiogenic therapy, and 33 receiving immune combination therapy. The ORRs were 78.9% for antiangiogenic therapy, 72.7% for immune combination therapy, and 42.9% for chemotherapy alone. The median PFS was 8.0 months for antiangiogenic therapy, 7.8 months for immune combination therapy, and 5.2 months for chemotherapy alone. Both combination therapy groups demonstrated superior efficacy compared to chemotherapy alone. CONCLUSION Targeted combined chemotherapy and immune combination chemotherapy showed superior efficacy as first-line treatments for ES-SCLC compared to chemotherapy alone, with manageable adverse reactions.
Collapse
Affiliation(s)
- Tiantian Zhang
- Departments of Medical OncologyThe First Affiliated Hospital of Bengbu Medical CollegeBengbuAnhuiPeople's Republic of China
| | - Lu Tao
- Departments of Medical OncologyThe First Affiliated Hospital of Bengbu Medical CollegeBengbuAnhuiPeople's Republic of China
| | - Yufo Chen
- Departments of Medical OncologyThe First Affiliated Hospital of Bengbu Medical CollegeBengbuAnhuiPeople's Republic of China
| | - Shanshan Zhang
- Departments of Medical OncologyThe First Affiliated Hospital of Bengbu Medical CollegeBengbuAnhuiPeople's Republic of China
| | - Yang Liu
- Departments of Medical OncologyThe First Affiliated Hospital of Bengbu Medical CollegeBengbuAnhuiPeople's Republic of China
| | - Yumei Li
- Departments of Medical OncologyThe First Affiliated Hospital of Bengbu Medical CollegeBengbuAnhuiPeople's Republic of China
| | - Rui Wang
- Departments of Medical OncologyThe First Affiliated Hospital of Bengbu Medical CollegeBengbuAnhuiPeople's Republic of China
- Anhui Provincial Key Laboratory of Cancer Translational MedicineThe First Affiliated Hospital of Bengbu Medical CollegeBengbuAnhuiPeople's Republic of China
| |
Collapse
|
3
|
Hou Y, Wang H, Wu J, Guo H, Chen X. Dissecting the pleiotropic roles of reactive oxygen species (ROS) in lung cancer: From carcinogenesis toward therapy. Med Res Rev 2024; 44:1566-1595. [PMID: 38284170 DOI: 10.1002/med.22018] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/14/2023] [Accepted: 01/10/2024] [Indexed: 01/30/2024]
Abstract
Lung cancer is a major cause of morbidity and mortality. The specific pulmonary structure to directly connect with ambient air makes it more susceptible to damage from airborne toxins. External oxidative stimuli and endogenous reactive oxygen species (ROS) play a crucial role in promoting lung carcinogenesis and development. The biological properties of higher ROS levels in tumor cells than in normal cells make them more sensitive and vulnerable to ROS injury. Therefore, the strategy of targeting ROS has been proposed for cancer therapy for decades. However, it is embarrassing that countless attempts at ROS-based therapies have had very limited success, and no FDA approval in the anticancer list was mechanistically based on ROS manipulation. Even compared with the untargetable proteins, such as transcription factors, ROS are more difficult to be targeted due to their chemical properties. Thus, the pleiotropic roles of ROS provide therapeutic potential for anticancer drug discovery, while a better dissection of the mechanistic action and signaling pathways is a prerequisite for future breakthroughs. This review discusses the critical roles of ROS in cancer carcinogenesis, ROS-inspired signaling pathways, and ROS-based treatment, exemplified by lung cancer. In particular, an eight considerations rule is proposed for ROS-targeting strategies and drug design and development.
Collapse
Affiliation(s)
- Ying Hou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
| | - Heng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Hongwei Guo
- Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Research and Evaluation of Bioactive Molecules & College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
- Department of Pharmaceutical Sciences, University of Macau, Taipa, Macao, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macao, China
| |
Collapse
|
4
|
Aung TM, Ngamjarus C, Proungvitaya T, Saengboonmee C, Proungvitaya S. Biomarkers for prognosis of meningioma patients: A systematic review and meta-analysis. PLoS One 2024; 19:e0303337. [PMID: 38758750 PMCID: PMC11101050 DOI: 10.1371/journal.pone.0303337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 04/23/2024] [Indexed: 05/19/2024] Open
Abstract
Meningioma is the most common primary brain tumor and many studies have evaluated numerous biomarkers for their prognostic value, often with inconsistent results. Currently, no reliable biomarkers are available to predict the survival, recurrence, and progression of meningioma patients in clinical practice. This study aims to evaluate the prognostic value of immunohistochemistry-based (IHC) biomarkers of meningioma patients. A systematic literature search was conducted up to November 2023 on PubMed, CENTRAL, CINAHL Plus, and Scopus databases. Two authors independently reviewed the identified relevant studies, extracted data, and assessed the risk of bias of the studies included. Meta-analyses were performed with the hazard ratio (HR) and 95% confidence interval (CI) of overall survival (OS), recurrence-free survival (RFS), and progression-free survival (PFS). The risk of bias in the included studies was evaluated using the Quality in Prognosis Studies (QUIPS) tool. A total of 100 studies with 16,745 patients were included in this review. As the promising markers to predict OS of meningioma patients, Ki-67/MIB-1 (HR = 1.03, 95%CI 1.02 to 1.05) was identified to associate with poor prognosis of the patients. Overexpression of cyclin A (HR = 4.91, 95%CI 1.38 to 17.44), topoisomerase II α (TOP2A) (HR = 4.90, 95%CI 2.96 to 8.12), p53 (HR = 2.40, 95%CI 1.73 to 3.34), vascular endothelial growth factor (VEGF) (HR = 1.61, 95%CI 1.36 to 1.90), and Ki-67 (HR = 1.33, 95%CI 1.21 to 1.46), were identified also as unfavorable prognostic biomarkers for poor RFS of meningioma patients. Conversely, positive progesterone receptor (PR) and p21 staining were associated with longer RFS and are considered biomarkers of favorable prognosis of meningioma patients (HR = 0.60, 95% CI 0.41 to 0.88 and HR = 1.89, 95%CI 1.11 to 3.20). Additionally, high expression of Ki-67 was identified as a prognosis biomarker for poor PFS of meningioma patients (HR = 1.02, 95%CI 1.00 to 1.04). Although only in single studies, KPNA2, CDK6, Cox-2, MCM7 and PCNA are proposed as additional markers with high expression that are related with poor prognosis of meningioma patients. In conclusion, the results of the meta-analysis demonstrated that PR, cyclin A, TOP2A, p21, p53, VEGF and Ki-67 are either positively or negatively associated with survival of meningioma patients and might be useful biomarkers to assess the prognosis.
Collapse
Affiliation(s)
- Tin May Aung
- Centre of Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Chetta Ngamjarus
- Department of Epidemiology and Biostatistics, Faculty of Public Health, Khon Kaen University, Khon Kaen, Thailand
| | - Tanakorn Proungvitaya
- Centre of Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Charupong Saengboonmee
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Siriporn Proungvitaya
- Centre of Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
5
|
Goggins E, Mironchik Y, Kakkad S, Jacob D, Wildes F, Bhujwalla ZM, Krishnamachary B. Reprogramming of VEGF-mediated extracellular matrix changes through autocrine signaling. Cancer Biol Ther 2023; 24:2184145. [PMID: 37389973 PMCID: PMC10012930 DOI: 10.1080/15384047.2023.2184145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 01/20/2023] [Accepted: 01/30/2023] [Indexed: 03/11/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) plays key roles in angiogenesis, vasculogenesis, and wound healing. In cancers, including triple negative breast cancer (TNBC), VEGF has been associated with increased invasion and metastasis, processes that require cancer cells to traverse through the extracellular matrix (ECM) and establish angiogenesis at distant sites. To further understand the role of VEGF in modifying the ECM, we characterized VEGF-mediated changes in the ECM of tumors derived from TNBC MDA-MB-231 cells engineered to overexpress VEGF. We established that increased VEGF expression by these cells resulted in tumors with reduced collagen 1 (Col1) fibers, fibronectin, and hyaluronan. Molecular characterization of tumors identified an increase of MMP1, uPAR, and LOX, and a decrease of MMP2, and ADAMTS1. α-SMA, a marker of cancer associated fibroblasts (CAFs), increased, and FAP-α, a marker of a subset of CAFs associated with immune suppression, decreased with VEGF overexpression. Analysis of human data from The Cancer Genome Atlas Program confirmed mRNA differences for several molecules when comparing TNBC with high and low VEGF expression. We additionally characterized enzymatic changes induced by VEGF overexpression in three different cancer cell lines that clearly identified autocrine-mediated changes, specifically uPAR, in these enzymes. Unlike the increase of Col1 fibers and fibronectin mediated by VEGF during wound healing, in the TNBC model, VEGF significantly reduced key protein components of the ECM. These results further expand our understanding of the role of VEGF in cancer progression and identify potential ECM-related targets to disrupt this progression.
Collapse
Affiliation(s)
- Eibhlin Goggins
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yelena Mironchik
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Samata Kakkad
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Desmond Jacob
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Flonne Wildes
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zaver M. Bhujwalla
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Balaji Krishnamachary
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
6
|
Vimalraj S, Hariprabu KNG, Rahaman M, Govindasami P, Perumal K, Sekaran S, Ganapathy D. Vascular endothelial growth factor-C and its receptor-3 signaling in tumorigenesis. 3 Biotech 2023; 13:326. [PMID: 37663750 PMCID: PMC10474002 DOI: 10.1007/s13205-023-03719-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 07/13/2023] [Indexed: 09/05/2023] Open
Abstract
The cancer-promoting ligand vascular endothelial growth factor-C (VEGF-C) activates VEGF receptor-3 (VEGFR-3). The VEGF-C/VEGFR-3 axis is expressed by a range of human tumor cells in addition to lymphatic endothelial cells. Activating the VEGF-C/VEGFR-3 signaling enhances metastasis by promoting lymphangiogenesis and angiogenesis inside and around tumors. Stimulation of VEGF-C/VEGFR-3 signaling promotes tumor metastasis in tumors, such as ovarian, renal, pancreatic, prostate, lung, skin, gastric, colorectal, cervical, leukemia, mesothelioma, Kaposi sarcoma, and endometrial carcinoma. We discuss and update the role of VEGF-C/VEGFR-3 signaling in tumor development and the research is still needed to completely comprehend this multifunctional receptor.
Collapse
Affiliation(s)
- Selvaraj Vimalraj
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology, Madras, Chennai, India
| | | | - Mostafizur Rahaman
- Department of Chemistry, College of Science, King Saud University, P. O. Box 2455, Riyadh, 11451 Saudi Arabia
| | - Periyasami Govindasami
- Department of Chemistry, College of Science, King Saud University, P. O. Box 2455, Riyadh, 11451 Saudi Arabia
| | - Karthikeyan Perumal
- Department of Chemistry and Biochemistry, The Ohio State University, 151 W. Woodruff Ave, Columbus, OH 43210 USA
| | - Saravanan Sekaran
- Department of Prosthodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu 600 077 India
| | - Dhanraj Ganapathy
- Department of Prosthodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu 600 077 India
| |
Collapse
|
7
|
Zhang Q, Zhao M, Lin S, Han Q, Ye H, Peng F, Li L. Prediction of prognosis and immunotherapy response in lung adenocarcinoma based on CD79A, DKK1 and VEGFC. Heliyon 2023; 9:e18503. [PMID: 37534013 PMCID: PMC10392102 DOI: 10.1016/j.heliyon.2023.e18503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 07/14/2023] [Accepted: 07/19/2023] [Indexed: 08/04/2023] Open
Abstract
Background Tumor immune microenvironment (TIME) is crucial for tumor initiation, progression, and metastasis; however, its relationship with lung adenocarcinoma (LUAD) is unknown. Traditional predictive models screen for biomarkers that are too general and infrequently associated with immune genes. Methods RNA sequencing data of LUAD patients and immune-related gene sets were retrieved from public databases. Using the common genes shared by The Cancer Genome Atlas (TCGA) and Immunology Database and Analysis Portal (ImmPort), differential gene expression analysis, survival analysis, Lasso regression analysis, and univariate and multivariate Cox regression analyses were performed to generate a novel risk score model. LUAD cohort in International Cancer Genome Consortium (ICGC), GSE68465 cohort in Gene Expression Omnibus (GEO) and an immunohistochemical assay were used to validate the key genes constructed risk score. The LUAD-related prognosis, clinical indicators, immune infiltrate characteristics, response to immunotherapy, and response to chemotherapeutic agents in different risk groups were evaluated by CIBERSORT, ImmuCellAI, pRRophetic and other tools. Results The risk score model was constructed using CD79a molecule (CD79A), Dickkopf WNT signaling pathway inhibitor 1 (DKK1), and vascular endothelial growth factor C (VEGFC). High risk score was identified as a negative predictor for overall survival (OS) in subgroup analyses with tumor stage, TNM classification, therapy outcome, and ESTIMATE scores (P < 0.05). Low risk score was positively associated with plasma cells, memory B cells, CD8 T cells, regulatory T cells and γδT cells (P < 0.05). In low-risk group, programmed cell death 1 receptor (PD1), cytotoxic T-lymphocyte associated protein 4 (CTLA4), and lymphocyte activating 3 (LAG3) and indoleamine 2,3-dioxygenase (IDO) were more robustly expressed (P < 0.05). The treatment responses of immune checkpoint blockade (ICB) therapy and chemotherapy were likewise superior in low-risk group (P < 0.05). In immunohistochemical analysis, the tumor group had significantly higher levels of CD79A, DKK1, and VEGFC than the adjacent normal group (P < 0.01). Conclusions CD79A, DKK1 and VEGFC are important differential genes related to LUAD, risk score could reliably predict prognosis, composition of TIME and immunotherapy responses in LUAD patients. The excellent performance of the risk model shows its strong and broad application potential.
Collapse
Affiliation(s)
- Qilong Zhang
- Department of Pharmacy, Zhejiang Hospital, Hangzhou, Zhejiang 310007, China
| | - Mingyuan Zhao
- Department of Pathology, Zhejiang Hospital, Hangzhou, Zhejiang 310007, China
| | - Shuangyan Lin
- Department of Pathology, Zhejiang Hospital, Hangzhou, Zhejiang 310007, China
| | - Qi Han
- Department of Pharmacy, Zhejiang Hospital, Hangzhou, Zhejiang 310007, China
| | - He Ye
- Department of Pharmacy, Zhejiang Hospital, Hangzhou, Zhejiang 310007, China
| | - Fang Peng
- Department of Pathology, Zhejiang Hospital, Hangzhou, Zhejiang 310007, China
| | - Li Li
- Department of Pharmacy, Zhejiang Hospital, Hangzhou, Zhejiang 310007, China
| |
Collapse
|
8
|
Chen S, Pan TY, Wu X, Li T, Wei Y, He HL, Zhou XM, Wang Q, Zhu JP. Uses of Vascular Endothelial Growth Factor C as a Lung Adenocarcinoma Prognostic Biomarker. World J Oncol 2023; 14:51-59. [PMID: 36896001 PMCID: PMC9990733 DOI: 10.14740/wjon1520] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 12/16/2022] [Indexed: 03/01/2023] Open
Abstract
Background Lung adenocarcinoma (LUAD) is the most common type of lung cancer and a leading cause of death worldwide. Vascular endothelial growth factor C (VEGF-C) has been identified as a prognosis prediction marker for LUAD. However, VEGF-C protein expression does not appear to significantly relate to LUAD patient survival in several studies. Methods We carried out a bioinformatic analysis to review the effect of VEGF-C mRNA expression on LUAD patient outcomes. GEPIA, UALCAN, TCGAportal, OncoLnc, LCE, GeneMANIA, Metascape, ImmuCellAI, and GSCA online databases were utilized. The expression levels of VEGF-C mRNA between normal tissue and LUAD tissue, overall survival (OS) analysis, function analysis, tumor microenvironment and drug sensitivity were conducted in the current study. Results We found that the expression level of VEGF-C mRNA was significantly lower in LUAD than normal tissue. Low expression of VEGF-C mRNA was also associated with better OS. VEGF-C expression was correlated with both NF1 and TP53 mutation status. No relationship was observed between VEGF-C and Tr1 or CD4 T-cell infiltrate scores. Additionally, VEGF-C was associated with epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor resistance. The sensitivity of 5-fluorouracil was positively correlated with VEGF-C, and the sensitivity of TGX221 was negatively correlated with VEGF-C. The activity of BI-2536 and BRD-A94377914 was positively correlated with VEGF-C. Conclusion Novel LUAD prognostic biomarkers such as VEGF-C mRNA may aid diagnosis and treatment, and may help identify optimal LUAD populations for therapeutic treatments.
Collapse
Affiliation(s)
- Shi Chen
- Department of Respiratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.,These authors contributed equally to this article
| | - Ting Yu Pan
- Department of Respiratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.,These authors contributed equally to this article
| | - Xiao Wu
- Department of Respiratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Tian Li
- Department of Respiratory Medicine of the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yu Wei
- Department of Respiratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Hai Lang He
- Department of Respiratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Xian Mei Zhou
- Department of Respiratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Qian Wang
- Department of Respiratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Ji Ping Zhu
- Department of Respiratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| |
Collapse
|
9
|
Therapeutic strategies for non-small cell lung cancer: Experimental models and emerging biomarkers to monitor drug efficacies. Pharmacol Ther 2023; 242:108347. [PMID: 36642389 DOI: 10.1016/j.pharmthera.2023.108347] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/15/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
While new targeted therapies have considerably changed the treatment and prognosis of non-small cell lung cancer (NSCLC), they are frequently unsuccessful due to primary or acquired resistances. Chemoresistance is a complex process that combines cancer cell intrinsic mechanisms including molecular and genetic abnormalities, aberrant interactions within the tumor microenvironment, and the pharmacokinetic characteristics of each molecule. From a pharmacological point of view, two levers could improve the response to treatment: (i) developing tools to predict the response to chemo- and targeted therapies and (ii) gaining a better understanding of the influence of the tumor microenvironment. Both personalized medicine approaches require the identification of relevant experimental models and biomarkers to understand and fight against chemoresistance mechanisms. After describing the main therapies in NSCLC, the scope of this review will be to identify and to discuss relevant in vitro and ex vivo experimental models that are able to mimic tumors. In addition, the interests of these models in the predictive responses to proposed therapies will be discussed. Finally, this review will evaluate the involvement of novel secreted biomarkers such as tumor DNA or micro RNA in predicting responses to anti-tumor therapies.
Collapse
|
10
|
Nanomodulation and nanotherapeutics of tumor-microenvironment. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
11
|
Zheng HR, Jiang AM, Gao H, Liu N, Zheng XQ, Fu X, Ruan ZP, Tian T, Liang X, Yao Y. The efficacy and safety of anlotinib combined with platinum-etoposide chemotherapy as first-line treatment for extensive-stage small cell lung cancer: A Chinese multicenter real-world study. Front Oncol 2022; 12:894835. [PMID: 36203439 PMCID: PMC9531009 DOI: 10.3389/fonc.2022.894835] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundPatients with extensive-stage small-cell lung cancer (ES-SCLC) have high recurrence rates and bleak prognosis. This multicenter real-world study aimed to explore the efficacy and safety of anlotinib combined with platinum-etoposide chemotherapy as the first-line treatment of ES-SCLC.MethodsPathologically confirmed ES-SCLC patients receiving anlotinib plus platinum-etoposide chemotherapy as the first-line treatment were enrolled in this retrospective study. The primary endpoint of this study was progression-free survival (PFS), and secondary endpoints included overall survival (OS), objective response rate (ORR), disease control rate (DCR), and adverse reactions. The Cox regression analyses were employed to investigate the independent prognostic factors for OS and PFS of these individuals.ResultsIn total, 58 patients were included in this study. The median PFS was 6.0 months [95% confidence interval (CI): 3.5-8.5], and the median OS was 10.5 months (95%CI 8.7-12.3). Thirty-four patients achieved partial response (PR), 18 patients achieved stable disease (SD), and 6 patients achieved progressive disease (PD). The ORR and DCR were 58.6% and 89.6%. The main treatment-related adverse reactions were generally tolerated. Myelosuppression (44.8%) was the most common adverse reaction, followed by hypertension (41.4%), fatigue (34.5%), gastrointestinal reaction (32.7%), and hand-foot syndrome (24.1%). Multivariate analysis showed that post-medication hand-foot syndrome [PFS 8.5 vs. 5.5 months, Hazards Ratio (HR)=0.23, 95%CI 0.07-0.72, P =0.012] was the independent predictor of PFS, and hypertension (OS 15.9 vs. 8.3 months, HR=0.18, 95%CI 0.05-0.58, P =0.005) was the independent predictor of OS.ConclusionAnlotinib combined with platinum-etoposide chemotherapy as the first-line treatment for ES-SCLC appears to be effective and well-tolerated in the real-world. Well-designed large-scale prospective studies are urgently needed in the future to verify our findings.
Collapse
Affiliation(s)
- Hao-Ran Zheng
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Department of Medical Oncology, Xi’an No.3 Hospital, Xi’an, China
| | - Ai-Min Jiang
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Huan Gao
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Na Liu
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiao-Qiang Zheng
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiao Fu
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Zhi-Ping Ruan
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Tao Tian
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xuan Liang
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yu Yao
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Yu Yao,
| |
Collapse
|
12
|
Go SI, Park S, Kang MH, Kim HG, Kang JH, Kim JH, Lee GW. Endothelial activation and stress index (EASIX) as a predictive biomarker in small cell lung cancer. Cancer Biomark 2022; 35:217-225. [PMID: 36120771 DOI: 10.3233/cbm-220032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Endothelial activation and insult may contribute to the aggressive clinical course of small-cell lung cancer (SCLC); however, no predictive biomarker for this pathogenesis has been identified. OBJECTIVE To evaluate the clinical impact of the endothelial activation and stress index (EASIX) in SCLC. METHODS In this retrospective study, the EASIX was calculated from measurements of serum lactate dehydrogenase, creatinine, and platelet levels. A total of 264 patients with SCLC treated with platinum-based chemotherapy were stratified into high and low EASIX groups. RESULTS Complete and objective response rates in the limited-stage (LD) were 19.5% vs. 33.3% (P= 0.050) and 85.4% vs. 97.9% (P= 0.028) in the high and low EASIX groups, respectively. There was no significant difference in the response rate between the two groups in the extensive-stage (ED). The median overall survival was 9.8 vs. 40.5 months in LD (P< 0.001) and 7.2 vs. 11.9 months in ED (P< 0.001) in the high and low EASIX groups, respectively. In multivariate analyses, a high EASIX level was an independent prognostic factor for worse progression-free and overall survival irrespective of stage. CONCLUSION EASIX may be a potential predictive biomarker of SCLC.
Collapse
Affiliation(s)
- Se-Il Go
- Division of Hematology-Oncology, Department of Internal Medicine, Institute of Health Science, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Korea
| | - Sungwoo Park
- Division of Hematology-Oncology, Department of Internal Medicine, Institute of Health Science, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, Korea
| | - Myoung Hee Kang
- Division of Hematology-Oncology, Department of Internal Medicine, Institute of Health Science, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Korea
| | - Hoon-Gu Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Institute of Health Science, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Korea
| | - Jung Hun Kang
- Division of Hematology-Oncology, Department of Internal Medicine, Institute of Health Science, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, Korea
| | - Jung Hoon Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Institute of Health Science, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, Korea
| | - Gyeong-Won Lee
- Division of Hematology-Oncology, Department of Internal Medicine, Institute of Health Science, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, Korea
| |
Collapse
|
13
|
Yuan E, Liu K, Lee J, Tsung K, Chow F, Attenello FJ. Modulating glioblastoma chemotherapy response: Evaluating long non-coding RNA effects on DNA damage response, glioma stem cell function, and hypoxic processes. Neurooncol Adv 2022; 4:vdac119. [PMID: 36105389 PMCID: PMC9466271 DOI: 10.1093/noajnl/vdac119] [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] [Indexed: 01/29/2023] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive primary adult brain tumor, with an estimated annual incidence of 17 000 new cases in the United States. Current treatments for GBM include chemotherapy, surgical resection, radiation therapy, and antiangiogenic therapy. However, despite the various therapeutic options, the 5-year survival rate remains at a dismal 5%. Temozolomide (TMZ) is the first-line chemotherapy drug for GBM; however, poor TMZ response is one of the main contributors to the dismal prognosis. Long non-coding RNAs (lncRNAs) are nonprotein coding transcripts greater than 200 nucleotides that have been implicated to mediate various GBM pathologies, including chemoresistance. In this review, we aim to frame the TMZ response in GBM via exploration of the lncRNAs mediating three major mechanisms of TMZ resistance: (1) regulation of the DNA damage response, (2) maintenance of glioma stem cell identity, and (3) exploitation of hypoxia-associated responses.
Collapse
Affiliation(s)
- Edith Yuan
- Corresponding Author: Edith Yuan, BA, Keck School of Medicine, University of Southern California, 1200 North State St. Suite 3300, Los Angeles, CA 90033, USA ()
| | - Kristie Liu
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Justin Lee
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kathleen Tsung
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Frances Chow
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Frank J Attenello
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
14
|
Zheng HR, Jiang AM, Gao H, Liu N, Zheng XQ, Fu X, Zhang R, Ruan ZP, Tian T, Liang X, Yao Y. The Efficacy and Safety of Anlotinib in Extensive-Stage Small Cell Lung Cancer: A Multicenter Real-World Study. Cancer Manag Res 2022; 14:2273-2287. [PMID: 35942069 PMCID: PMC9356751 DOI: 10.2147/cmar.s364125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Anlotinib, an antiangiogenic multi-target tyrosine kinase inhibitor (TKI), has shown favorable anticancer efficacy and acceptable safety in treating extensive-stage small cell lung cancer (ES-SCLC) in some clinical studies. This research aimed to explore the real-world efficacy and safety of anlotinib in ES-SCLC. Methods Pathologically confirmed ES-SCLC patients receiving anlotinib were enrolled for this retrospective study. The primary endpoint of this study was progression-free survival (PFS), and secondary endpoints included overall survival (OS), objective response rate (ORR), disease control rate (DCR), and adverse reactions. Results In total, 202 patients were included in this study. The median PFS of all patients was 4.8 months [95% confidence interval (CI): 3.9–5.7], and the median OS was 7.6 months (95% CI 6.5–8.7). Respectively, the overall ORR and DCR were 30.2% and 87.1%. The univariate and multivariate Cox regression analyses revealed that patients with Eastern Cooperative Oncology Group Performance Status (ECOG PS) ≤1, plus chemotherapy or immunotherapy, plus radiotherapy, and post-medication hypertension might have longer PFS and OS. The PFS and OS were significantly prolonged in combination group than that in monotherapy group [PFS 6.0 vs 3.6 months, hazards ratio (HR)=0.49, 95% CI 0.34–0.70, P < 0.001; OS 9.2 vs 4.8 months, HR = 0.48, 95% CI 0.32–0.72, P < 0.001]. The main treatment-related adverse reactions were generally tolerated. The incidence of adverse reactions in combination group was higher than that in monotherapy group (75.0% vs 52.6%, P = 0.001). The most common adverse reaction was hypertension, followed by hand-foot syndrome and fatigue, regardless of monotherapy or combination group. Conclusion Anlotinib is effective and well tolerated in patients with ES-SCLC in the real-world. The clinical efficacy of anlotinib combined with chemotherapy or immunotherapy is better than that of monotherapy. Further investigations are needed for prospective studies with larger sample size.
Collapse
Affiliation(s)
- Hao-Ran Zheng
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Department of Medical Oncology, Xi’an No.3 Hospital, Xi’an, Shaanxi, People’s Republic of China
| | - Ai-Min Jiang
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Huan Gao
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Na Liu
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Xiao-Qiang Zheng
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Xiao Fu
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Rui Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Zhi-Ping Ruan
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Tao Tian
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Xuan Liang
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Correspondence: Xuan Liang; Yu Yao, Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, No. 277 Yanta West Road, Xi’an, Shaanxi, 710061, People’s Republic of China, Tel +86-29-85324600, Fax +86-29-85324086, Email ;
| | - Yu Yao
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| |
Collapse
|
15
|
Peng W, Cao L, Chen L, Lin G, Zhu B, Hu X, Lin Y, Zhang S, Jiang M, Wang J, Li J, Li C, Shao L, Du H, Hou T, Chen Z, Xiang J, Pu X, Li J, Xu F, Loong H, Wu L. Comprehensive Characterization of the Genomic Landscape in Chinese Pulmonary Neuroendocrine Tumors Reveals Prognostic and Therapeutic Markers (CSWOG-1901). Oncologist 2022; 27:e116-e125. [PMID: 35641209 PMCID: PMC8895731 DOI: 10.1093/oncolo/oyab044] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 10/07/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Pulmonary neuroendocrine tumors (pNETs) include typical carcinoid (TC), atypical carcinoid (AC), large cell neuroendocrine carcinoma (LCNEC), and small cell lung carcinoma (SCLC). The optimal treatment strategy for each subtype remains elusive, partly due to the lack of comprehensive understanding of their molecular features. We aimed to explore differential genomic signatures in pNET subtypes and identify potential prognostic and therapeutic biomarkers. METHODS We investigated genomic profiles of 57 LCNECs, 49 SCLCs, 18 TCs, and 24 ACs by sequencing tumor tissues with a 520-gene panel and explored the associations between genomic features and prognosis. RESULTS Both LCNEC and SCLC displayed higher mutation rates for TP53, PRKDC, SPTA1, NOTCH1, NOTCH2, and PTPRD than TC and AC. Small cell lung carcinoma harbored more frequent co-alterations in TP53-RB1, alterations in PIK3CA and SOX2, and mutations in HIF-1, VEGF and Notch pathways. Large cell neuroendocrine carcinoma (12.7 mutations/Mb) and SCLC (11.9 mutations/Mb) showed higher tumor mutational burdens than TC (2.4 mutations/Mb) and AC (7.1 mutations/Mb). 26.3% of LCNECs and 20.8% of ACs harbored alterations in classical non-small cell lung cancer driver genes. The presence of alterations in the homologous recombination pathway predicted longer progression-free survival in advanced LCNEC patients with systemic therapy (P = .005) and longer overall survival (OS) in SCLC patients with resection (P = .011). The presence of alterations in VEGF (P = .048) and estrogen (P = .018) signaling pathways both correlated with better OS in patients with resected SCLC. CONCLUSION We performed a comprehensive genomic investigation on 4 pNET subtypes in the Chinese population. Our data revealed distinctive genomic signatures in subtypes and provided new insights into the prognostic and therapeutic stratification of pNETs.
Collapse
Affiliation(s)
- Wenying Peng
- The Second Department of Thoracic Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Liming Cao
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Likun Chen
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Gen Lin
- Department of Thoracic Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, People’s Republic of China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Xiaohua Hu
- The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Yingcheng Lin
- Cancer Hospital of Shantou University Medical College, Shantou, People’s Republic of China
| | - Sheng Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Meilin Jiang
- The Second Department of Thoracic Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Jingyi Wang
- The Second Department of Thoracic Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Junjun Li
- Department of Pathology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Chao Li
- Department of Pathology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, People’s Republic of China
| | - Lin Shao
- Burning Rock Biotech, Guangzhou, People’s Republic of China
| | - Haiwei Du
- Burning Rock Biotech, Guangzhou, People’s Republic of China
| | - Ting Hou
- Burning Rock Biotech, Guangzhou, People’s Republic of China
| | - Zhiqiu Chen
- Burning Rock Biotech, Guangzhou, People’s Republic of China
| | - Jianxing Xiang
- Burning Rock Biotech, Guangzhou, People’s Republic of China
| | - Xingxiang Pu
- The Second Department of Thoracic Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Jia Li
- The Second Department of Thoracic Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Fang Xu
- The Second Department of Thoracic Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Herbert Loong
- Department of Clinical Oncology, Deputy Medical Director, Phase 1 Clinical Trials Centre, Chinese University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Lin Wu
- The Second Department of Thoracic Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| |
Collapse
|
16
|
Chen Y, Jin Y, Hu X, Chen M. Infiltrating T lymphocytes in the tumor microenvironment of small cell lung cancer: a state of knowledge review. J Cancer Res Clin Oncol 2022; 148:881-895. [PMID: 34997864 DOI: 10.1007/s00432-021-03895-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 12/19/2021] [Indexed: 10/19/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have brought new hope for the treatment of patients with small cell lung cancer (SCLC) over the past decades. However, the overall response rate is limited, and is lower than that in non-small cell lung cancer (NSCLC). This is in part because of the lack of pre-existing tumor-infiltrating T lymphocytes (TITLs), especially cytotoxic T cells (CTLs), in the SCLC tumor microenvironment (TME), resulting in insufficient anti-tumor immune response. To unleash the full potential of ICIs, the trafficking and infiltration of TITLs to the tumor is necessary and tightly regulated, the highly immunosuppressive tumor microenvironment blunts the infiltration and function of TITLs that reach the tumor in SCLC. Here, we review the characteristics of TITLs, the effects of various factors on T cell infiltration, and possible strategies to restore or promote T cell infiltration in the TME of SCLC.
Collapse
Affiliation(s)
- Yamei Chen
- Zhejiang Key Laboratory of Radiation Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
| | - Ying Jin
- Zhejiang Key Laboratory of Radiation Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China.,Department of Medical Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
| | - Xiao Hu
- Zhejiang Key Laboratory of Radiation Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China. .,Department of Radiation Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang, China.
| | - Ming Chen
- Zhejiang Key Laboratory of Radiation Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China. .,Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
17
|
Valadares BN, Stephano MA. Small cell lung cancer: an overview of the targets. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e19114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
18
|
Seebacher NA, Krchniakova M, Stacy AE, Skoda J, Jansson PJ. Tumour Microenvironment Stress Promotes the Development of Drug Resistance. Antioxidants (Basel) 2021; 10:1801. [PMID: 34829672 PMCID: PMC8615091 DOI: 10.3390/antiox10111801] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/29/2021] [Accepted: 11/08/2021] [Indexed: 01/18/2023] Open
Abstract
Multi-drug resistance (MDR) is a leading cause of cancer-related death, and it continues to be a major barrier to cancer treatment. The tumour microenvironment (TME) has proven to play an essential role in not only cancer progression and metastasis, but also the development of resistance to chemotherapy. Despite the significant advances in the efficacy of anti-cancer therapies, the development of drug resistance remains a major impediment to therapeutic success. This review highlights the interplay between various factors within the TME that collectively initiate or propagate MDR. The key TME-mediated mechanisms of MDR regulation that will be discussed herein include (1) altered metabolic processing and the reactive oxygen species (ROS)-hypoxia inducible factor (HIF) axis; (2) changes in stromal cells; (3) increased cancer cell survival via autophagy and failure of apoptosis; (4) altered drug delivery, uptake, or efflux and (5) the induction of a cancer stem cell (CSC) phenotype. The review also discusses thought-provoking ideas that may assist in overcoming the TME-induced MDR. We conclude that stressors from the TME and exposure to chemotherapeutic agents are strongly linked to the development of MDR in cancer cells. Therefore, there remains a vast area for potential research to further elicit the interplay between factors existing both within and outside the TME. Elucidating the mechanisms within this network is essential for developing new therapeutic strategies that are less prone to failure due to the development of resistance in cancer cells.
Collapse
Affiliation(s)
| | - Maria Krchniakova
- Department of Experimental Biology, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic;
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| | - Alexandra E. Stacy
- Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia;
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic;
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| | - Patric J. Jansson
- Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia;
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St. Leonards, NSW 2065, Australia
| |
Collapse
|
19
|
Gao X, Peng L, Zhang L, Huang K, Yi C, Li B, Meng X, Li J. Real-world efficacy and safety of anlotinib as third- or further-line treatment in refractory small cell lung cancer. J Cancer Res Clin Oncol 2021; 148:2661-2671. [PMID: 34748028 DOI: 10.1007/s00432-021-03848-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/30/2021] [Indexed: 01/10/2023]
Abstract
PURPOSE As a novel antiangiogenic multi-target tyrosine kinase inhibitor recently approved in China, anlotinib has exhibited promising anticancer efficacy and acceptable safety profile in the salvage treatment of small cell lung cancer (SCLC) in clinical trials. Here we retrospectively investigated the efficacy and safety of anlotinib as third- or further-line treatment in patients with refractory SCLC. PATIENTS AND METHODS A total of 40 patients with refractory SCLC treated with anlotinib monotherapy were included in this study. The clinicopathological data, treatment information, survival data and safety data were retrospectively collected. Survival curves were constructed using the Kaplan-Meier method. Univariate analysis was performed by log-rank testing. RESULTS Altogether, 40 patients of extensive-stage SCLC or progressive limited-stage SCLC received anlotinib monotherapy as third- or further-line treatment from July 2018 to June 2020. Four patients achieved partial response (PR), 14 patients achieved stable disease (SD), no complete response (CR) was recorded, and 22 patients experienced progressive disease (PD). The disease control rate (DCR) was 45.0%. The median progression-free survival (PFS) was 3.0 months (95% CI 2.241-3.759), and the median overall survival (OS) was 7.8 months (95% CI 3.190-12.410). The common adverse effects (AEs) included hypertension, fatigue, anorexia, cough, rash and nausea. Grade 3 treatment-related AEs occurred in 3 (7.5%) patients. One patient interrupted anlotinib treatment due to repeated grade 1 epistaxis. Univariate analysis revealed that patients without liver metastases, previously treated with radiotherapy or with Eastern Cooperative Oncology Group (ECOG) scores of 0 or 1 had longer OS with anlotinib treatment. Cox regression analysis demonstrated that patients without liver metastases and patients with ECOG score ≤ 1 had longer PFS, while patients without liver metastases had longer OS. CONCLUSION Anlotinib is beneficial to refractory SCLC as third- or further-line treatment, especially in patients without liver metastasis and with better physical status. Related adverse effects are tolerable and manageable.
Collapse
Affiliation(s)
- Xuetian Gao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, 250117, Shandong Province, China
| | - Ling Peng
- Department of Respiratory Disease, Zhejiang Provincial People's Hospital, Hangzhou, 310000, Zhejiang Province, China
| | - Li Zhang
- Department of Oncology, Yunyang County People's Hospital, Yunyang, 404599, Chongqing, China
| | - Kai Huang
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
| | - Cuihua Yi
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
| | - Bei Li
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
| | - Xue Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, 250117, Shandong Province, China
| | - Jisheng Li
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China.
| |
Collapse
|
20
|
Gao Y, Mi J, Liu Z, Song Q. Leisure Sedentary Behavior and Risk of Lung Cancer: A Two-Sample Mendelian Randomization Study and Mediation Analysis. Front Genet 2021; 12:763626. [PMID: 34777480 PMCID: PMC8582637 DOI: 10.3389/fgene.2021.763626] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/18/2021] [Indexed: 12/20/2022] Open
Abstract
Leisure sedentary behavior, especially television watching, has been previously reported as associated with the risk of lung cancer in observational studies. This study aims to evaluate the causal association with two-sample Mendelian randomization (MR) analysis. Single nucleotide polymorphisms associated with leisure television watching, computer use, and driving were extracted from genome-wide association studies. Summary-level results of lung cancer overall and histological types were obtained from International Lung Cancer Consortium (ILCCO). In univariable MR using inverse-variance-weighted method, we observed causal effects of television watching on lung cancer [OR, 1.89, 95% confidence interval (CI), 1.41, 2.54; p = 2.33 × 10-5], and squamous cell lung cancer (OR, 2.37, 95% CI, 1.58, 3.55; p = 3.02 × 10-5), but not on lung adenocarcinoma (OR, 1.40, 95% CI, 0.94, 2.09; p = 0.100). No causal effects of computer use and driving on lung cancer were observed. Television watching significantly increased the exposure to several common risk factors of lung cancer. The associations of television watching with lung cancer and squamous cell lung cancer were compromised after adjusting for smoking quantity with multivariable MR. Our mediation analyses estimated indirect effects of television watching on lung cancer (beta, 0.31, 95% CI, 0.13, 0.52; p = 6.64 × 10-4) and squamous cell lung cancer (beta, 0.33, 95% CI, 0.14, 0.53, p = 4.76 × 10-4) mediated by smoking quantity. Our findings indicate that television watching is positively correlated with the risk of lung cancer, potentially mediated through affecting smoking quantity.
Collapse
Affiliation(s)
- Yan Gao
- Department of Oncology, Cancer Center, Remin Hospital of Wuhan University, Wuhan, China
| | - Jiarui Mi
- Master Program of Biomedicine, Karolinska Institutet, Solna, Sweden
| | - Zhengye Liu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qibin Song
- Department of Oncology, Cancer Center, Remin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
21
|
Šutić M, Vukić A, Baranašić J, Försti A, Džubur F, Samaržija M, Jakopović M, Brčić L, Knežević J. Diagnostic, Predictive, and Prognostic Biomarkers in Non-Small Cell Lung Cancer (NSCLC) Management. J Pers Med 2021; 11:1102. [PMID: 34834454 PMCID: PMC8624402 DOI: 10.3390/jpm11111102] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 12/25/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Despite growing efforts for its early detection by screening populations at risk, the majority of lung cancer patients are still diagnosed in an advanced stage. The management of lung cancer has dramatically improved in the last decade and is no longer based on the "one-fits-all" paradigm or the general histological classification of non-small cell versus small cell lung cancer. Emerging options of targeted therapies and immunotherapies have shifted the management of lung cancer to a more personalized treatment approach, significantly influencing the clinical course and outcome of the disease. Molecular biomarkers have emerged as valuable tools in the prognosis and prediction of therapy response. In this review, we discuss the relevant biomarkers used in the clinical management of lung tumors, from diagnosis to prognosis. We also discuss promising new biomarkers, focusing on non-small cell lung cancer as the most abundant type of lung cancer.
Collapse
Affiliation(s)
- Maja Šutić
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (M.Š.); (A.V.); (J.B.)
| | - Ana Vukić
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (M.Š.); (A.V.); (J.B.)
| | - Jurica Baranašić
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (M.Š.); (A.V.); (J.B.)
| | - Asta Försti
- Hopp Children’s Cancer Center (KiTZ), 69120 Heidelberg, Germany;
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Feđa Džubur
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; (F.D.); (M.S.); (M.J.)
- Clinical Department for Respiratory Diseases Jordanovac, University Hospital Centre Zagreb, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Miroslav Samaržija
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; (F.D.); (M.S.); (M.J.)
- Clinical Department for Respiratory Diseases Jordanovac, University Hospital Centre Zagreb, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Marko Jakopović
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; (F.D.); (M.S.); (M.J.)
- Clinical Department for Respiratory Diseases Jordanovac, University Hospital Centre Zagreb, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Luka Brčić
- Diagnostic and Research Institute of Pathology, Medical University of Graz, 8010 Graz, Austria;
| | - Jelena Knežević
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (M.Š.); (A.V.); (J.B.)
- Faculties for Dental Medicine and Health, University of Osijek, 31000 Osijek, Croatia
| |
Collapse
|
22
|
The Value of Preoperative Plasma VEGF Levels in Urothelial Carcinoma of the Bladder Treated with Radical Cystectomy. Eur Urol Focus 2021; 8:972-979. [PMID: 34454852 DOI: 10.1016/j.euf.2021.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/15/2021] [Accepted: 08/12/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Elevated preoperative plasma levels of the angiogenesis-related marker VEGF have been associated with worse oncological outcomes in various malignancies. OBJECTIVE To investigate the predictive/prognostic role of VEGF in patients with urothelial carcinoma of the bladder (UCB) treated with radical cystectomy (RC). DESIGN, SETTING, AND PARTICIPANTS VEGF plasma levels were measured preoperatively in 1036 patients with UCB who underwent RC. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The correlation between plasma VEGF levels and pathological and survival outcomes was assessed using logistic regression and Cox regression analyses. Discrimination was assessed using the concordance index (C index). The clinical net benefit was evaluated using decision curve analysis (DCA). RESULTS AND LIMITATIONS Patients with higher pretreatment plasma VEGF levels had poorer recurrence-free survival (RFS), cancer-specific survival (CSS), and overall survival (OS) according to log-rank tests (all p < 0.001). Higher VEGF levels were not independently associated with higher risk of lymph node metastasis, ≥pT3 disease, or non-organ-confined disease (all p > 0.05). Preoperative plasma VEGF levels were independently associated with RFS, CSS, and OS in preoperative and postoperative multivariable models. However, in all cases the C index increased by <0.02 and there was no improvement in net benefit on DCA. A limitation is that none of the patients received current elements of standard of care such as neoadjuvant chemotherapy. CONCLUSIONS Elevated plasma VEGF levels were associated with features of biologically and clinically aggressive disease such as worse survival outcomes among patients with UCB treated with RC. However, VEGF appears to have relatively limited incremental additive value in clinical use. Further study of VEGF for UCB prognostication is warranted before routine use in clinical algorithms. PATIENT SUMMARY Currently available models for predicting outcomes in bladder cancer are less than optimal. A protein called vascular endothelial growth factor (VEGF), which is a marker of the formation of blood vessels (angiogenesis), may have a role in predicting survival outcomes in bladder cancer. TAKE HOME MESSAGE Elevated plasma VEGF levels are associated with worse survival outcomes for patients with urothelial carcinoma of the bladder (UCB) treated with radical cystectomy. VEGF could be used as a part of a biomarker panel to enhance tools currently used for risk stratification for patients with UCB.
Collapse
|
23
|
Jantus-Lewintre E, Massutí Sureda B, González Larriba JL, Rodríguez-Abreu D, Juan O, Blasco A, Dómine M, Provencio Pulla M, Garde J, Álvarez R, Maestu I, Pérez de Carrión R, Artal Á, Rolfo C, de Castro J, Guillot M, Oramas J, de Las Peñas R, Ferrera L, Martínez N, Serra Ò, Rosell R, Camps C. Prospective Exploratory Analysis of Angiogenic Biomarkers in Peripheral Blood in Advanced NSCLC Patients Treated With Bevacizumab Plus Chemotherapy: The ANGIOMET Study. Front Oncol 2021; 11:695038. [PMID: 34381717 PMCID: PMC8350788 DOI: 10.3389/fonc.2021.695038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/21/2021] [Indexed: 12/24/2022] Open
Abstract
Finding angiogenic prognostic markers in advanced non-small-cell lung cancer is still an unmet medical need. We explored a set of genetic variants in the VEGF-pathway as potential biomarkers to predict clinical outcomes of patients with non-small-cell lung cancer treated with chemotherapy plus bevacizumab. We prospectively analyzed the relationship between VEGF-pathway components with both pathological and prognostic variables in response to chemotherapy plus bevacizumab in 168 patients with non-squamous non-small-cell lung cancer. Circulating levels of VEGF and VEGFR2 and expression of specific endothelial surface markers and single-nucleotide polymorphisms in VEGF-pathway genes were analyzed. The primary clinical endpoint was progression-free survival. Secondary endpoints included overall survival and objective tumor response. VEGFR-1 rs9582036 variants AA/AC were associated with increased progression-free survival (p = 0.012 and p = 0.035, respectively), and with improved overall survival (p = 0.019) with respect to CC allele. Patients with VEGF-A rs3025039 harboring allele TT had also reduced mortality risk (p = 0.049) compared with the CC allele. The VEGF-A rs833061 variant was found to be related with response to treatment, with 61.1% of patients harboring the CC allele achieving partial treatment response. High pre-treatment circulating levels of VEGF-A were associated with shorter progression-free survival (p = 0.036). In conclusion, in this prospective study, genetic variants in VEGFR-1 and VEGF-A and plasma levels of VEGF-A were associated with clinical benefit, progression-free survival, or overall survival in a cohort of advanced non-squamous non-small-cell lung cancer patients receiving chemotherapy plus antiangiogenic therapy.
Collapse
Affiliation(s)
- Eloisa Jantus-Lewintre
- Departamento de Biotecnología, Universitat Politècnica de València, Unidad Mixta TRIAL, Fundación para la Investigación del Hospital General Universitario de Valencia/Centro de Investigación Príncipe Felipe, CIBERONC, Valencia, Spain
| | | | | | - Delvys Rodríguez-Abreu
- Complejo Hospitalario Universitario Insular Materno-Infantil de Gran Canaria, Universidad de las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Oscar Juan
- Servicio de Oncología Médica, Hospital Politécnico y Universitario La Fe, Valencia, Spain
| | - Ana Blasco
- Consorcio Hospital General Universitario de Valencia, CIBERONC, Valencia, Spain
| | - Manuel Dómine
- Servicio de Oncología Médica, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | | | - Javier Garde
- Departamento de Oncología Médica, Hospital Arnau de Vilanova, Valencia, Spain
| | - Rosa Álvarez
- Departamento de Oncología Médica, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Inmaculada Maestu
- Departamento de Oncología Médica, Hospital Universitari Doctor Peset, Valencia, Spain
| | | | - Ángel Artal
- Servicio de Oncología Médica, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Christian Rolfo
- Experimental Therapeutics Program, Greenbaum Comprehensive Cancer Center, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Javier de Castro
- Servicio de Oncología Médica, Hospital Universitario La Paz, Madrid, Spain
| | - Mónica Guillot
- Servicio de Oncología Médica, Hospital Son Espases, Palma de Mallorca, Spain
| | - Juana Oramas
- Departamento de Oncología Médica, Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | - Ramón de Las Peñas
- Departamento de Oncología Médica, Hospital Provincial de Castelló, Castellón, Spain
| | - Lioba Ferrera
- Servicio de Oncología Médica, Hospital Universitario Nuestra Señora de la Candelaria, Santa Cruz de Tenerife, Spain
| | - Natividad Martínez
- Departamento de Oncología, Hospital General Universitario de Elche, Elche, Spain
| | - Òlbia Serra
- Departamento de Oncología Médica, Hospital General de l' Hospitalet, L'Hospitalet de Llobregat, Spain
| | - Rafael Rosell
- Programa de la Biología del Cáncer y Medicina de Precisión, Institut de Recerca Germans Trias i Pujol, Badalona, Spain/Instituto Oncológico Dr. Rosell, Barcelona, Spain
| | - Carlos Camps
- Departamento de Medicina, Universitat Politècnica de València, Unidad Mixta TRIAL, Fundación para la Investigación del Hospital General Universitario de Valencia/Centro de Investigación Príncipe Felipe, CIBERONC, Valencia, Spain
| |
Collapse
|
24
|
Maleki P, Mowla SJ, Taheri M, Ghafouri-Fard S, Raheb J. The role of long intergenic non-coding RNA for kinase activation (LINK-A) as an oncogene in non-small cell lung carcinoma. Sci Rep 2021; 11:4210. [PMID: 33602983 PMCID: PMC7892821 DOI: 10.1038/s41598-021-82892-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 01/20/2021] [Indexed: 12/24/2022] Open
Abstract
The oncogenic role of long intergenic non-coding RNA for kinase activation (LINK-A) has been appraised in triple-negative breast cancer. However, the molecular function of LINK-A is still unclear in most cancers including lung cancer. The present study aimed to evaluate the impact of down-regulation of LINK-A in A549 and Calu-3 cell lines as cellular models of non-small cell lung carcinoma (NSCLC). We used the RNA interference system to knock down LINK-A. LINK-A expression was significantly reduced by siRNA transfection in A549 and Calu-3 cell lines. LINK-A down-regulation significantly reduced cell viability, colony-forming ability and cell migration, as measured by MTT, colony formation and invasion assays. Finally, cell cycle analysis and Annexin-V/7AAD staining indicated that apoptosis was influenced by LINK-A silencing. Taken together, LINK-A can be proposed as an oncogene in NSCLC.
Collapse
Affiliation(s)
- Parichehr Maleki
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Seyed Javad Mowla
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jamshid Raheb
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran.
| |
Collapse
|
25
|
Reck M, Luft A, Bondarenko I, Shevnia S, Trukhin D, Kovalenko NV, Vacharadze K, Andrea F, Hontsa A, Choi J, Shin D. A phase III, randomized, double-blind, multicenter study to compare the efficacy, safety, pharmacokinetics, and immunogenicity between SB8 (proposed bevacizumab biosimilar) and reference bevacizumab in patients with metastatic or recurrent nonsquamous non-small cell lung cancer. Lung Cancer 2020; 146:12-18. [DOI: 10.1016/j.lungcan.2020.05.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/20/2020] [Accepted: 05/23/2020] [Indexed: 10/24/2022]
|
26
|
Clinical relevance of tumor microenvironment: immune cells, vessels, and mouse models. Hum Cell 2020; 33:930-937. [PMID: 32507979 DOI: 10.1007/s13577-020-00380-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/20/2020] [Indexed: 12/19/2022]
Abstract
The tumor microenvironment (TME) plays a crucial role in tumor progression, therapeutic response, and patient outcomes. TME includes immune cells, blood and lymphatic vessels, and so on. There are anti-cancer and pro-cancer immune cells. In general, infiltration of anti-cancer immune cells, such as cytotoxic T cells (CTLs), is associated with a favorable patient prognosis. In contrast, infiltration of pro-cancer immune cells, such as regulatory T cells (Tregs), tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), is associated with a worse prognosis. However, some immune cells, which play an ambivalent role in cancer immunity, have demonstrated contradictory impacts on patient prognosis. Blood and lymphatic vessels play crucial roles in TME not only as delivery and draining systems of fluid and molecules, but also allowing cancer cells access to systematic circulation to metastasize. Angiogenesis promotes cancer aggressiveness and is associated with a worse prognosis. Its targeted therapy shows a benefit in some cancers, however, because the target can vary by caner type, a benefit of anti-angiogenesis therapy is limited in the current standard of care. Lymphangiogenesis plays a role in lymph node metastasis, thus, it is associated with a poor prognosis in some cancers. To study TME, the mouse model is one of the most commonly used tools. The choice of appropriate mouse model depends on the hypothesis being tested and the scientific question being asked. Here, we review recent studies that investigated the clinical relevance of TME components and introduce mouse models to study TME.
Collapse
|
27
|
Qin S, Yi M, Jiao D, Li A, Wu K. Distinct Roles of VEGFA and ANGPT2 in Lung Adenocarcinoma and Squamous Cell Carcinoma. J Cancer 2020; 11:153-167. [PMID: 31892982 PMCID: PMC6930396 DOI: 10.7150/jca.34693] [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: 03/07/2019] [Accepted: 09/26/2019] [Indexed: 01/03/2023] Open
Abstract
Background: Vascular endothelial growth factor A (VEGFA) and angiopoietin 2 (ANGPT2) are key mediators in angiogenesis. The expression and clinical significance of VEGFA and ANGPT2 have been investigated in lung cancer, but the results are controversial. The specific roles of VEGFA and ANGPT2 in adenocarcinoma (ADC) and squamous cell carcinoma (SQC) are still not fully understood. To characterize it, we conducted the current study. Materials and Methods: The relationships between clinic-pathological characteristics and the protein expressions of VEGFA and ANGPT2 were analyzed on tissue microarrays by immunohistochemistry (IHC) staining. Then public databases were used to evaluate the association of VEGFA and ANGPT2 mRNA expressions with clinic-pathological parameters and prognosis. Cobalt chloride (CoCl2) was adopted to mimic a hypoxic microenvironment and western blot was used to detect the expression of hypoxia inducible factor-1α (HIF-1α), VEGFA and ANGPT2 in lung cancer cell lines. Results: IHC staining revealed that the expressions of VEGFA and ANGPT2 were enriched in lung cancer tissues compared with normal tissues. Additionally, both VEGFA and ANGPT2 protein levels were significantly associated with the tumor size and lymph node metastasis only in ADC, not SQC. More importantly, increased VEGFA and ANGPT2 protein levels were negatively correlated with overall survival (OS) of ADC individuals. Meta-analyses of 22 gene expression omnibus (GEO) databases of lung cancer implicated that patients with higher VEGFA and ANGPT2 mRNA expressions tended to have advanced stage in ADC rather than SQC. Kaplan-Meier plot analyses further verified that high levels of VEGFA and ANGPT2 mRNA were associated with poor survival only in ADC. Moreover, the combination of VEGFA and ANGPT2 could more precisely predict prognosis in ADC. In hypoxia-mimicking conditions, induced expression of HIF-1α unregulated VEGFA and ANGPT2 proteins abundance. Conclusion: Our results showed hypoxia upregulated the protein levels of VEGFA and ANGPT2 in lung cancer cell lines, and the roles of VEGFA and ANGPT2 were distinct in ADC and SQC. Combined detections of VEGFA and ANGPT2 may be valuable prognostic biomarkers for ADC and double block of VEGFA and ANGPT2 may improve therapeutic outcome.
Collapse
Affiliation(s)
- Shuang Qin
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dechao Jiao
- Department of Interventional Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Anping Li
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| |
Collapse
|
28
|
Boutsikou E, Porpodis K, Chatzipavlidou V, Hardavella G, Gerasimou G, Domvri K, Papadopoulos N, Avramidou V, Spyratos D, Kontakiotis T, Zarogoulidis K. Predictive Value of 99MTC-hynic-toc Scintigraphy in Lung Neuroendocrine Tumor Diagnosis. Technol Cancer Res Treat 2019; 18:1533033819842586. [PMID: 31079574 PMCID: PMC6535698 DOI: 10.1177/1533033819842586] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: Νeuroendocrine tumors of the lungs are rare arising in the thymus and gastro-entero-pancreatic tract and belonging to foregut of neuroendocrine tumors. The aim of the present prospective study was to estimate the potential impact of single-photon emission computed tomography somatostatin receptor scintigraphy using 99mTc-Tektrotyd on diagnosis, treatment response, and prognosis in patients with neuroendocrine tumors of the lungs. Methods: Thirty-six patients with neuroendocrine tumors of the lungs were evaluated by using 99mTc-HYNIC-TOC scintigraphy. The scintigraphic results were compared to liver tissue uptake (Krenning score). Likewise, the functional imaging results were compared with biochemical indices including chromogranin A, neuroendocrine-specific enolase, and insulin-like growth factor 1 at the time of diagnosis (baseline) and disease progression. Results: The number of somatostatin receptors, expressed with Krenning score, did not show any correlation with the survival of patients both at baseline (P = .08) and at disease progression (P = .24), and scintigraphy results did not relate significantly to progression-free survival. Comparing the results of 99mTc-HYNIC-TOC scintigraphy according to the response of patients in the initial treatment, a statistically significant negative correlation was observed both in the first and in the second scintigraphy with patients’ response (P = .001 and P < .001, respectively). The concentrations of biochemical markers were in accordance with scintigraphy results in the diagnosis. Conclusion: This study indicates that 99mTc-HYNIC-TOC scintigraphy appears to be a reliable, noninvasive technique for detection of primary neuroendocrine tumors and their locoregional or distant metastases, although it cannot be used as a neuroendocrine tumors of the lungs predictive technique.
Collapse
Affiliation(s)
- Efimia Boutsikou
- 1 Pulmonary Department, Oncology Unit, Aristotle University of Thessaloniki, G. Papanikolaou Hospital, Thessaloniki, Macedonia, Greece
| | - Konstantinos Porpodis
- 1 Pulmonary Department, Oncology Unit, Aristotle University of Thessaloniki, G. Papanikolaou Hospital, Thessaloniki, Macedonia, Greece
| | - Vasiliki Chatzipavlidou
- 2 Nuclear Medicine Department, Anticancer Hospital Theagenio, Thessaloniki, Macedonia, Greece
| | - Georgia Hardavella
- 3 Department of Respiratory Medicine, King's College Hospital, London, United Kingdom
| | - George Gerasimou
- 4 2nd Clinical Laboratory of Nuclear Medicine, AHEPA University Hospital, Thessaloniki, Macedonia, Greece
| | - Kalliopi Domvri
- 1 Pulmonary Department, Oncology Unit, Aristotle University of Thessaloniki, G. Papanikolaou Hospital, Thessaloniki, Macedonia, Greece
| | - Nikitas Papadopoulos
- 2 Nuclear Medicine Department, Anticancer Hospital Theagenio, Thessaloniki, Macedonia, Greece
| | - Vasiliki Avramidou
- 5 3rd Paediatric Department, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Dionisis Spyratos
- 1 Pulmonary Department, Oncology Unit, Aristotle University of Thessaloniki, G. Papanikolaou Hospital, Thessaloniki, Macedonia, Greece
| | - Theodoros Kontakiotis
- 1 Pulmonary Department, Oncology Unit, Aristotle University of Thessaloniki, G. Papanikolaou Hospital, Thessaloniki, Macedonia, Greece
| | - Konstantinos Zarogoulidis
- 1 Pulmonary Department, Oncology Unit, Aristotle University of Thessaloniki, G. Papanikolaou Hospital, Thessaloniki, Macedonia, Greece
| |
Collapse
|
29
|
Ghimessy AK, Gellert A, Schlegl E, Hegedus B, Raso E, Barbai T, Timar J, Ostoros G, Megyesfalvi Z, Gieszer B, Moldvay J, Renyi-Vamos F, Lohinai Z, Hoda MA, Klikovits T, Klepetko W, Laszlo V, Dome B. KRAS Mutations Predict Response and Outcome in Advanced Lung Adenocarcinoma Patients Receiving First-Line Bevacizumab and Platinum-Based Chemotherapy. Cancers (Basel) 2019; 11:E1514. [PMID: 31600989 PMCID: PMC6827133 DOI: 10.3390/cancers11101514] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/23/2019] [Accepted: 10/03/2019] [Indexed: 01/09/2023] Open
Abstract
Bevacizumab, combined with platinum-based chemotherapy, has been widely used in the treatment of advanced-stage lung adenocarcinoma (LADC). Although KRAS (V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog) mutation is the most common genetic alteration in human LADC and its role in promoting angiogenesis has been well established, its prognostic and predictive role in the above setting remains unclear. The association between KRAS exon 2 mutational status and clinicopathological variables including progression-free survival and overall survival (PFS and OS, respectively) was retrospectively analyzed in 501 Caucasian stage IIIB-IV LADC patients receiving first-line platinum-based chemotherapy (CHT) with or without bevacizumab (BEV). EGFR (epidermal growth factor receptor)-mutant cases were excluded. Of 247 BEV/CHT and 254 CHT patients, 95 (38.5%) and 75 (29.5%) had mutations in KRAS, respectively. KRAS mutation was associated with smoking (p = 0.008) and female gender (p = 0.002) in the BEV/CHT group. We found no difference in OS between patients with KRAS-mutant versus KRAS wild-type tumors in the CHT-alone group (p = 0.6771). Notably, patients with KRAS-mutant tumors demonstrated significantly shorter PFS (p = 0.0255) and OS (p = 0.0186) in response to BEV/CHT compared to KRAS wild-type patients. KRAS mutation was an independent predictor of shorter PFS (hazard ratio, 0.597; p = 0.011) and OS (hazard ratio, 0.645; p = 0.012) in the BEV/CHT group. G12D KRAS-mutant patients receiving BEV/CHT showed significantly shorter PFS (3.7 months versus 8.27 months in the G12/13x group; p = 0.0032) and OS (7.2 months versus 16.1 months in the G12/13x group; p = 0.0144). In this single-center, retrospective study, KRAS-mutant LADC patients receiving BEV/CHT treatment exhibited inferior PFS and OS compared to those with KRAS wild-type advanced LADC. G12D mutations may define a subset of KRAS-mutant LADC patients unsuitable for antiangiogenic therapy with BEV.
Collapse
Affiliation(s)
- Aron Kristof Ghimessy
- Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, 1122 Budapest, Hungary.
| | - Aron Gellert
- Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, 1122 Budapest, Hungary.
| | - Erzsebet Schlegl
- Department of Tumor Biology, National Koranyi Institute of Pulmonology-Semmelweis University, 1122 Budapest, Hungary.
| | - Balazs Hegedus
- Department of Thoracic Surgery, Ruhrlandklinik, University Duisburg-Essen, 45239 Essen, Germany.
- nd Department of Pathology, Semmelweis University, 1091 Budapest, Hungary.
- Tumor Progression Research Group, Hungarian Academy of Sciences-Semmelweis University, 1091 Budapest, Hungary.
| | - Erzsebet Raso
- nd Department of Pathology, Semmelweis University, 1091 Budapest, Hungary.
- Tumor Progression Research Group, Hungarian Academy of Sciences-Semmelweis University, 1091 Budapest, Hungary.
| | - Tamas Barbai
- nd Department of Pathology, Semmelweis University, 1091 Budapest, Hungary.
- Tumor Progression Research Group, Hungarian Academy of Sciences-Semmelweis University, 1091 Budapest, Hungary.
| | - Jozsef Timar
- nd Department of Pathology, Semmelweis University, 1091 Budapest, Hungary.
- Tumor Progression Research Group, Hungarian Academy of Sciences-Semmelweis University, 1091 Budapest, Hungary.
| | - Gyula Ostoros
- th Department of Pulmonology, National Koranyi Institute of Pulmonology, 1122 Budapest, Hungary.
| | - Zsolt Megyesfalvi
- Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, 1122 Budapest, Hungary.
- Department of Tumor Biology, National Koranyi Institute of Pulmonology-Semmelweis University, 1122 Budapest, Hungary.
| | - Balazs Gieszer
- Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, 1122 Budapest, Hungary.
| | - Judit Moldvay
- Department of Tumor Biology, National Koranyi Institute of Pulmonology-Semmelweis University, 1122 Budapest, Hungary.
- nd Department of Pathology, Semmelweis University, 1091 Budapest, Hungary.
- MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, 1091 Budapest, Hungary.
| | - Ferenc Renyi-Vamos
- Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, 1122 Budapest, Hungary.
| | - Zoltan Lohinai
- Department of Tumor Biology, National Koranyi Institute of Pulmonology-Semmelweis University, 1122 Budapest, Hungary.
| | - Mir Alireza Hoda
- Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Centre Vienna, Medical University Vienna, A-1090 Vienna, Austria.
| | - Thomas Klikovits
- Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Centre Vienna, Medical University Vienna, A-1090 Vienna, Austria.
| | - Walter Klepetko
- Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Centre Vienna, Medical University Vienna, A-1090 Vienna, Austria.
| | - Viktoria Laszlo
- Department of Tumor Biology, National Koranyi Institute of Pulmonology-Semmelweis University, 1122 Budapest, Hungary.
- Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Centre Vienna, Medical University Vienna, A-1090 Vienna, Austria.
| | - Balazs Dome
- Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, 1122 Budapest, Hungary.
- Department of Tumor Biology, National Koranyi Institute of Pulmonology-Semmelweis University, 1122 Budapest, Hungary.
- Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Centre Vienna, Medical University Vienna, A-1090 Vienna, Austria.
| |
Collapse
|
30
|
Xu Y, Huang Z, Lu H, Yu X, Li Y, Li W, Chen J, Chen M, Gong L, Chen K, Qin J, Xu X, Jin Y, Zhao J, Shi X, Han N, Xie F, Zhang P, Xu W, Fan Y. Apatinib in patients with extensive-stage small-cell lung cancer after second-line or third-line chemotherapy: a phase II, single-arm, multicentre, prospective study. Br J Cancer 2019; 121:640-646. [PMID: 31523058 PMCID: PMC6889407 DOI: 10.1038/s41416-019-0583-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 08/23/2019] [Accepted: 08/30/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Small-cell lung cancer (SCLC) remains an aggressive cancer with short-term survival due to limited therapeutic options. Apatinib is a small-molecule tyrosine kinase inhibitor that selectively inhibits vascular endothelial growth factor receptor-2. This study aimed to investigate the efficacy and safety of apatinib in patients with extensive-stage (EC) SCLC who had progressed after two or three previous therapies. METHODS Eligible patients were histologically confirmed ES-SCLC after two or three previous treatments, including a platinum-based regimen. Patients received apatinib at an initial dose of 500 mg once daily. The primary endpoint was the objective response rate. RESULTS Forty patients were enrolled. At the data cut-off time (November 15, 2018), the median follow-up was 7.4 months; no patients remained on treatment, and five were still in follow-up. An objective response was achieved in 7 of 40 patients (17.5%) in the intention-to-treat population, and 7 of 38 patients (18.4%) in the per-protocol population. The median progression-free survival and overall survival were 3.0 months and 5·8 months, respectively. The most commonly observed grade 3 or greater treatment-related adverse events were hypertension, hand-foot syndrome, increased L-gamma-glutamyltransferase. CONCLUSIONS Apatinib exhibited efficacy and an acceptable safety profile in previously heavily-treated ES-SCLC patients. Further exploration of apatinib in phase III trials is warranted. TRIAL REGISTRATION NCT02945852.
Collapse
Affiliation(s)
- Yanjun Xu
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, China
| | - Zhiyu Huang
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, China
| | - Hongyang Lu
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, China
| | - Xinming Yu
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, China
| | - Yuping Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenfeng Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jun Chen
- Department of Radiotherapy and Chemotherapy, Yinzhou People's Hospital, Ningbo, China
| | - Ming Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Lei Gong
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, China
| | - Kaiyan Chen
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, China
| | - Jin Qin
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, China
| | - Xiaoling Xu
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, China
| | - Ying Jin
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, China
| | - Jun Zhao
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, China
| | - Xun Shi
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, China
| | - Na Han
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, China
| | - Fajun Xie
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, China
| | - Peng Zhang
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, China
| | - Weizhen Xu
- Good Clinical Practice Center of Zhejiang Cancer Hospital, Hangzhou, China
| | - Yun Fan
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, China. .,Key laboratory on Diagnosis and Treatment Technology on Thoracic Cancer, Zhejiang Cancer Hospital (Zhejiang Cancer Research Institute), Hangzhou, China.
| |
Collapse
|
31
|
Li N, Liu T, Li H, Zhang L, Chu L, Meng Q, Qiao Q, Han W, Zhang J, Guo M, Zhao J. ILF2 promotes anchorage independence through direct regulation of PTEN. Oncol Lett 2019; 18:1689-1696. [PMID: 31423236 PMCID: PMC6614677 DOI: 10.3892/ol.2019.10510] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 05/09/2019] [Indexed: 12/19/2022] Open
Abstract
Anoikis is a specific form of programmed cell death induced by loss of contact between cells and extracellular matrices or other cells. Only tumor cells that are resistant to anoikis can survive in the state of detachment from the primary tissue during the early stages of metastasis. The ability to resist anoikis is crucial for cancer cell metastasis. ILF2 is a proto-oncogene previously studied in glioma, NSCLC, esophageal cancer and pancreatic ductal carcinoma. The results from the present study revealed that the transcription factor interleukin enhancer-binding factor 2 (ILF2) was highly expressed in non-small cell lung cancer (NSCLC) cell lines compared with in normal cell lines. ChIP and luciferase reporter gene assays demonstrated that ILF2 inhibited the expression level of the tumor suppressor gene phosphatase and tensin homolog (PTEN) by directly binding to its upstream regulatory region. Furthermore, the results from the detection of cell adhesion and apoptosis in cell suspension culture demonstrated that this mechanism enabled NSCLC cells to reduce adherence to the matrix and to survive in this abnormal state. These results suggested that ILF2 may promote the anchorage-independence of NSCLC cells through the suppression of PTEN.
Collapse
Affiliation(s)
- Na Li
- Pathology Department, Xingtai Medical College, Xingtai, Hebei 054000, P.R. China
| | - Tao Liu
- Department of Internal Medicine, Civil Administration General Hospital of Hebei, Xingtai, Hebei 054000, P.R. China
| | - Hui Li
- Pathology Department, Xingtai Medical College, Xingtai, Hebei 054000, P.R. China
| | - Lifang Zhang
- Department of Internal Medicine, Civil Administration General Hospital of Hebei, Xingtai, Hebei 054000, P.R. China
| | - Liping Chu
- Department of Internal Medicine, Civil Administration General Hospital of Hebei, Xingtai, Hebei 054000, P.R. China
| | - Qingge Meng
- Department of Internal Medicine, Xingtai Medical College, The Third Affiliated Hospital of Xingtai Medical College, Xingtai, Hebei 054000, P.R. China
| | - Qinzeng Qiao
- Department of Internal Medicine, Civil Administration General Hospital of Hebei, Xingtai, Hebei 054000, P.R. China
| | - Weikun Han
- Department of Internal Medicine, Civil Administration General Hospital of Hebei, Xingtai, Hebei 054000, P.R. China
| | - Junhui Zhang
- Pathology Department, Xingtai Medical College, Xingtai, Hebei 054000, P.R. China
| | - Minying Guo
- Pathology Department, Xingtai Medical College, Xingtai, Hebei 054000, P.R. China
| | - Jia Zhao
- Department of Internal Medicine, The Third Affiliated Hospital of Xingtai Medical College, Xingtai, Hebei 054000, P.R. China
| |
Collapse
|
32
|
Prognostic effect of VEGF gene variants in metastatic non-small-cell lung cancer patients. Angiogenesis 2019; 22:433-440. [PMID: 30977010 DOI: 10.1007/s10456-019-09668-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/08/2019] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Clinical and pathological characteristics are still considered prognostic markers in metastatic non-small-cell lung cancer (NSCLC) patients but they cannot explain all interindividual variability. Tumoral angiogenesis mediated by the vascular endothelial growth factor (VEGF) is critical for the progression and metastasis of the disease. We aimed to investigate the prognostic role of genetic variants within the VEGF pathway in patients with metastatic NSCLC. MATERIALS AND METHODS We prospectively included 170 patients with metastatic NSCLC treated with first-line platinum-based chemotherapy. A comprehensive panel of single-nucleotide polymorphisms (SNPs) in genes belonging to the VEGF pathway (VEGFA, VEGFR1/FLT1, VEGFR2/KDR, GRB2, ITGAV, KISS1, KRAS, PRKCE, HIF1α, MAP2K4, MAP2K6, and MAPK11) were genotyped in blood DNA samples. SNPs were evaluated for association with overall survival (OS) and progression-free survival (PFS). RESULTS In multivariate analyses adjusted for patient characteristics, we found that VEGFA rs2010963 and VEGFR2 rs2071559 were significantly associated with OS [Hazard Ratio (HR) 0.7 (0.5-0.9); p = 0.026 and HR 1.5 (1.1-2.3); p = 0.025, respectively]. Additionally, ITGAV rs35251833 and MAPK11 rs2076139 were significantly associated with PFS [HR 2.5 (1.4-4.3; p = 0.002 and HR 0.6 (0.5-0.9); p = 0.013, respectively]. CONCLUSION Our findings reinforce the potential clinical value of germline variants in VEGFA and VEGFR2 and show for the first time variants in ITGAV and MAPK11 as promising prognostic markers in metastatic NSCLC patients receiving platinum-based chemotherapy.
Collapse
|
33
|
Li H, Zeng J, Jin X, Yu X, Zhou G, Hong W. Apatinib for chemotherapy-refractory extensive-stage SCLC: a retrospective study. Cancer Chemother Pharmacol 2019; 83:1083-1090. [PMID: 30937519 DOI: 10.1007/s00280-019-03823-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 03/19/2019] [Indexed: 10/27/2022]
Abstract
PURPOSE There is no standard treatment strategy for patients with extensive-stage small cell lung cancer (SCLC) who have failed two or more prior chemotherapeutic regimens. In this study, we retrospectively evaluated the efficacy and safety of apatinib in patients with extensive-stage SCLC after failure of more than second-line chemotherapy. METHODS A study group comprised of 22 patients with extensive-stage SCLC after failure of more than two prior chemotherapeutic regimens was given apatinib orally at an initial dose of 500 mg daily until disease progression or unacceptable toxicity. This study was analyzed according to the National Cancer Institute Common Toxicity Criteria for adverse events (AEs) and Response Evaluation Criteria in Solid Tumors (RECIST) for response assessment. RESULTS Between August 30, 2015, and May 26, 2017, 22 patients were enrolled for evaluating the efficacy and safety of apatinib. Among them, 12/22 (54.5%) underwent dose reduction during treatment. Up to July 31, 2018, the median progression-free survival rate was 135.0 days [95% confidence interval (CI) 63.8-206.2]. According to the RECIST criteria, the disease control rate (DCR) was 86.4%, 19/22 [comprised of partial response (PR) 18.2%, 4/22; and stable disease (SD) 68.2%, 15/22 patients]. The most frequent AEs were hand-foot syndrome (45.5%, 10/22), secondary hypertension (45.5%, 10/22) and fatigue (40.9%, 9/22). The primary grade 3 or 4 toxicities were hypertension (22.7%, 5/22), hand-foot syndrome (13.6%, 3/22), and proteinuria (9.1%, 2/22). CONCLUSIONS Apatinib exhibits modest activity and acceptable toxicity for patients with heavily pretreated extensive-stage SCLC.
Collapse
Affiliation(s)
- Hui Li
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Jian Zeng
- Department of Department of Thoracic Surgery, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Xiangyu Jin
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Xinmin Yu
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Guoming Zhou
- Department of Laboratory Medicines, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Wei Hong
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China.
| |
Collapse
|
34
|
Dubois C, Rocks N, Blacher S, Primac I, Gallez A, García-Caballero M, Gérard C, Brouchet L, Noël A, Lenfant F, Cataldo D, Pequeux C. Lymph/angiogenesis contributes to sex differences in lung cancer through oestrogen receptor alpha signalling. Endocr Relat Cancer 2019; 26:201-216. [PMID: 30444717 DOI: 10.1530/erc-18-0328] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 11/14/2018] [Indexed: 12/23/2022]
Abstract
Oestrogen signalling pathways are emerging targets for lung cancer therapy. Unravelling the contribution of oestrogens in lung cancer development is a pre-requisite to support the development of sex-based treatments and identify patients who could potentially benefit from anti-oestrogen treatments. In this study, we highlight the contribution of lymphatic and blood endothelia in the sex-dependent modulation of lung cancer. The orthotopic graft of syngeneic lung cancer cells into immunocompetent mice showed that lung tumours grow faster in female mice than in males. Moreover, oestradiol (E2) promoted tumour development, increased lymph/angiogenesis and VEGFA and bFGF levels in lung tumours of females through an oestrogen receptor (ER) alpha-dependent pathway. Furthermore, while treatment with ERb antagonist was inefficient, ERa antagonist (MPP) and tamoxifen decreased lung tumour volumes, altered blood and lymphatic vasculature and reduced VEGFA and bFGF levels in females, but not in males. Finally, the quantification of lymphatic and blood vasculature of lung adenocarcinoma biopsies from patients aged between 35 and 55 years revealed more extensive lymphangiogenesis and angiogenesis in tumour samples issued from women than from men. In conclusion, our findings highlight an E2/ERa-dependent modulation of lymphatic and blood vascular components of lung tumour microenvironment. Our study has potential clinical implication in a personalised medicine perspective by pointing to the importance of oestrogen status or supplementation on lung cancer development that should be considered to adapt therapeutic strategies.
Collapse
Affiliation(s)
- Charline Dubois
- Laboratory of Tumour and Development Biology, GIGA-Cancer, University of Liège, CHU-B23, Liège, Belgium
| | - Natacha Rocks
- Laboratory of Tumour and Development Biology, GIGA-Cancer, University of Liège, CHU-B23, Liège, Belgium
| | - Silvia Blacher
- Laboratory of Tumour and Development Biology, GIGA-Cancer, University of Liège, CHU-B23, Liège, Belgium
| | - Irina Primac
- Laboratory of Tumour and Development Biology, GIGA-Cancer, University of Liège, CHU-B23, Liège, Belgium
| | - Anne Gallez
- Laboratory of Tumour and Development Biology, GIGA-Cancer, University of Liège, CHU-B23, Liège, Belgium
| | - Melissa García-Caballero
- Laboratory of Tumour and Development Biology, GIGA-Cancer, University of Liège, CHU-B23, Liège, Belgium
| | - Céline Gérard
- Laboratory of Tumour and Development Biology, GIGA-Cancer, University of Liège, CHU-B23, Liège, Belgium
| | - Laurent Brouchet
- Thoracic Surgery Department, University Hospital CHU Toulouse, Toulouse, France
| | - Agnès Noël
- Laboratory of Tumour and Development Biology, GIGA-Cancer, University of Liège, CHU-B23, Liège, Belgium
| | - Françoise Lenfant
- INSERM UMR1048, Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, University of Toulouse III Paul Sabatier, UPS, Toulouse, France
| | - Didier Cataldo
- Laboratory of Tumour and Development Biology, GIGA-Cancer, University of Liège, CHU-B23, Liège, Belgium
| | - Christel Pequeux
- Laboratory of Tumour and Development Biology, GIGA-Cancer, University of Liège, CHU-B23, Liège, Belgium
| |
Collapse
|
35
|
The novel PI3K inhibitor S1 synergizes with sorafenib in non-small cell lung cancer cells involving the Akt-S6 signaling. Invest New Drugs 2018; 37:828-836. [PMID: 30456603 DOI: 10.1007/s10637-018-0698-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/12/2018] [Indexed: 12/19/2022]
Abstract
Non-small cell lung cancer (NSCLC) has been the major cause of cancer-related deaths worldwide. Targeted therapy has been available as an additive strategy for NSCLC patients, but the inevitable resistance to mono-targeted agents has largely hampered its usage in the clinic. We have previously designed and synthesized a novel small molecule compound S1, 2-methoxy-3-phenylsulfonamino-5-(quinazolin-6-yl) benzamides and demonstrated its inhibition of PI3K and mTOR as well as the anti-tumor potential. In the present study, we have identified that S1 alone or combined with the multi-kinase inhibitor sorafenib can inhibit the in vitro cell proliferation of NSCLC cells (A549, NCI-H157 and 95D cells) and tumor growth in the A549 xenograft model. S1 alone produced inhibitory effects on the colony formation, cell migration and invasion and angiogenesis, with more pronounced inhibition when used with sorafenib. We further revealed that S1 mainly inhibited the Akt/S6 phosphorylation while sorafenib mostly decreased the phosphorylation of ERK. Together, the novel PI3K/mTOR inhibitor S1 per se exhibits strong anti-tumor effects in NSCLC cells and A549 xenograft, effects possibly via its inhibition of cell proliferation, invasion and migration and angiogenesis. The combination of S1 and sorafenib exerts potentiated anti-tumor effects, in which the underlying mechanisms may involve their differential modulation of the phosphorylation of Akt and S6 in the PI3K/Akt/mTOR cascades and ERK phosphorylation in the Raf/MEK/ERK pathways. The combination of S1 and sorafenib could be used as an additive approach in treating NSCLC in the clinic.
Collapse
|
36
|
Zhao Z, Xia G, Li N, Su R, Chen X, Zhong L. Autophagy Inhibition Promotes Bevacizumab-induced Apoptosis and Proliferation Inhibition in Colorectal Cancer Cells. J Cancer 2018; 9:3407-3416. [PMID: 30271503 PMCID: PMC6160673 DOI: 10.7150/jca.24201] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 07/02/2018] [Indexed: 12/21/2022] Open
Abstract
Aim: Anti-VEGF therapy plays an important role in the treatment of malignant tumors, especially metastatic malignant tumors. However, resistance and an inefficient response to anti-VEGF therapy exist. The current study aimed to investigate whether autophagy plays a part in the anti-tumor effect of bevacizumab in colorectal cancer cells. Methods: VEGF-A expression was measured by immunohistochemical methods. Cell viability and cell apoptosis were detected using 3-(4,5-dimethylthiazol-2-yl)-3,5-diphenylformazan (MTT) and flow cytometry. Autophagy was assessed by a western blot, fluorescence microscopy and transmission electron microscopy. HIF-1α was measured using a western blot. A xenograft tumor model of colorectal cancer was constructed to determine the efficacy of the treatment of bevacizumab and chloroquine. Results: VEGF-A protein was upregulated in colorectal cancer tissue. Anti-VEGF (bevacizumab) inhibited cell viability and induced apoptosis. Moreover, bevacizumab induced autophagy. The inhibition of autophagy by chloroquine or by small interfering RNA promoted bevacizumab-induced apoptosis and proliferation inhibition. Further study showed that bevacizumab treatment significantly augmented HIF-1α. Furthermore, cells pretreated with YC-1, a HIF-1α inhibitor, displayed significantly attenuated bevacizumab-induced autophagy. Finally, a combinatory treatment of bevacizumab and chloroquine synergistically inhibited tumor growth in a xenograft tumor model of colorectal cancer cells. Conclusions: Our results showed that the inhibition of autophagy promoted the anti-tumor effect of bevacizumab and may offer a promising therapeutic strategy for colorectal cancer.
Collapse
Affiliation(s)
- Zhi Zhao
- Department of Gastrointestinal and Hernia Surgery, People's Hospital of Guilin, Guilin, China, 541002
| | - Guanggai Xia
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China, 200233
| | - Ni Li
- Health Management Center, People's Hospital of Guilin, Guilin, China, 541002
| | - Ruping Su
- Department of Gastrointestinal and Hernia Surgery, People's Hospital of Guilin, Guilin, China, 541002
| | - Xiao Chen
- Department of Gastrointestinal and Hernia Surgery, People's Hospital of Guilin, Guilin, China, 541002
| | - Li Zhong
- Department of Gastrointestinal and Hernia Surgery, People's Hospital of Guilin, Guilin, China, 541002
| |
Collapse
|
37
|
Ayala-Domínguez L, Brandan ME. Quantification of tumor angiogenesis with contrast-enhanced x-ray imaging in preclinical studies: a review. Biomed Phys Eng Express 2018; 4. [DOI: 10.1088/2057-1976/aadc2d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/22/2018] [Indexed: 01/01/2023]
|
38
|
Wang L, Dou X, Liu T, Lu W, Ma Y, Yang Y. Tumor size and lymph node metastasis are prognostic markers of small cell lung cancer in a Chinese population. Medicine (Baltimore) 2018; 97:e11712. [PMID: 30075575 PMCID: PMC6081096 DOI: 10.1097/md.0000000000011712] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 07/05/2018] [Indexed: 12/31/2022] Open
Abstract
Small cell lung cancer (SCLC) is a high-grade neuroendocrine tumor characterized by rapid growth, early metastatic spread, and poor prognosis. This study aimed to explore the prognosis factors of survival in Chinese SCLC patients.A total of 78 patients with stage IIIA SCLC (mean age: 53.9 years, 65 males and 13 females) were enrolled in this retrospective study. At least of 5 years follow-up was performed.The survival time of these patients ranged from 1 month to 66 months with a median survival time of 11 months. Kaplan-Meier method with log-rank test was performed and showed that survival time in patients with tumor size ≤4 cm (median: 16 months) was significantly longer (P < .001) than that in patients with tumor size > 4 cm (median: 8 months); the median survival time of the patients with single lymph node metastasis was significantly longer than that in patients with multiple lymph node metastasis (P = .043). Combined multiple lymph node metastasis and tumor size >4 cm presented the worst survival outcome than others. Multivariate analysis by Cox Hazard model shows that the lymph node metastasis and tumors size were prognostic factors independent of age, sex, smoke, surgery, and treatment regimen (P < .05).Results showed that larger tumor size and multiple lymph node metastasis were associated with the poor survival in SCLC.
Collapse
Affiliation(s)
- Liang Wang
- Key Laboratory of Carcinogensis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute
| | - Xuejun Dou
- Department of Thoracic Surgery, Aerospace Center Hospital
| | - Tao Liu
- Department of Thoracic Surgery, Beijing Chest Hospital, Beijing, China
| | - Weiqiang Lu
- Department of Thoracic Surgery, Aerospace Center Hospital
| | - Yunlei Ma
- Department of Thoracic Surgery, Aerospace Center Hospital
| | - Yue Yang
- Key Laboratory of Carcinogensis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute
| |
Collapse
|
39
|
The EGFR Inhibitor Gefitinib Enhanced the Response of Human Oral Squamous Cell Carcinoma to Cisplatin In Vitro. Drugs R D 2018; 17:545-555. [PMID: 28828595 PMCID: PMC5694417 DOI: 10.1007/s40268-017-0204-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Introduction The epidermal growth factor receptor (EGFR) is highly expressed in a variety of solid tumors including oral cavity squamous cell carcinoma (OSCC) and has been implicated in the resistance of these tumors to cisplatin. This study was performed to determine if the EGFR tyrosine kinase inhibitor gefitinib could enhance the cytotoxic effect of cisplatin on OSCC cells in vitro. Methods The expression of EGFR and the phosphorylation of its downstream signaling to ERK, and AKT pathway were detected by Western blotting. Cell proliferation and survival were determined by AlamarBlue and colony formation assay respectively. Cells apoptosis were determined by Western blotting for cleaved PARP protein and by flowcytometry of cells stained with Annexin V and PI. Results Cal27, OSC19, and SCC25 cells treated with gefitinib 1 μM demonstrated reduced phosphorylation of EGFR, AKT, and ERK proteins with very limited inhibition of proliferation. Cisplatin inhibited proliferation of the same cell lines in a dose-dependent manner. The concentration producing 50% inhibition (IC50) for cisplatin decreased in the presence of gefitinib 1 μM, and a combination of cisplatin 5 µM and gefitinib 1 µM caused synergistic growth inhibition and synergistic reduction in cell survival. The growth inhibitory effect of the combination was associated with reduced ERK and AKT activation, increased poly ADP ribose polymerase (PARP) cleavage, and increased apoptosis. Conclusion Thus, in OSCC cells in vitro, inhibition of EGFR activity with gefitinib enhances the apoptotic effect of cisplatin. This has potential implications for enhancement of cisplatin effectiveness in tumors that over-express the EGFR.
Collapse
|
40
|
Sécher T, Guilleminault L, Reckamp K, Amanam I, Plantier L, Heuzé-Vourc'h N. Therapeutic antibodies: A new era in the treatment of respiratory diseases? Pharmacol Ther 2018; 189:149-172. [PMID: 29730443 DOI: 10.1016/j.pharmthera.2018.05.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Respiratory diseases affect millions of people worldwide, and account for significant levels of disability and mortality. The treatment of lung cancer and asthma with therapeutic antibodies (Abs) is a breakthrough that opens up new paradigms for the management of respiratory diseases. Antibodies are becoming increasingly important in respiratory medicine; dozens of Abs have received marketing approval, and many more are currently in clinical development. Most of these Abs target asthma, lung cancer and respiratory infections, while very few target chronic obstructive pulmonary disease - one of the most common non-communicable causes of death - and idiopathic pulmonary fibrosis. Here, we review Abs approved for or in clinical development for the treatment of respiratory diseases. We notably highlight their molecular mechanisms, strengths, and likely future trends.
Collapse
Affiliation(s)
- T Sécher
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université François Rabelais de Tours, F-37032 Tours, France
| | - L Guilleminault
- Pôle des Voies respiratoires, Hôpital Larrey, CHU de Toulouse, F-31059 Toulouse, France; STROMALab, Université de Toulouse, CNRS ERL 5311, EFS, INP-ENVT, Inserm, UPS, F-31013 Toulouse, France
| | - K Reckamp
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - I Amanam
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - L Plantier
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université François Rabelais de Tours, F-37032 Tours, France; CHRU de Tours, Service de Pneumologie, F-37000 Tours, France
| | - N Heuzé-Vourc'h
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université François Rabelais de Tours, F-37032 Tours, France.
| |
Collapse
|
41
|
Guo S, Martin MG, Tian C, Cui J, Wang L, Wu S, Gu W. Evaluation of Detection Methods and Values of Circulating Vascular Endothelial Growth Factor in Lung Cancer. J Cancer 2018; 9:1287-1300. [PMID: 29675110 PMCID: PMC5907677 DOI: 10.7150/jca.22020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 01/19/2018] [Indexed: 01/08/2023] Open
Abstract
Lung cancer is the deadliest cancer in the world. Angiogenesis plays a crucial role of the incidence, progression, and metastasis in lung cancer. Angiogenesis inhibitors are used to treat non-small cell lung cancer (NSCLC) patients, and the molecular biomarkers are also being assessed to predict treatment response/therapeutic response and patients' prognosis. Vascular endothelial growth factor (VEGF) is a signal protein produced by cells that stimulates angiogenesis. Due to its predictive values of prognosis on NSCLC, a large number of methods have been developed and evaluated to detect VEGF levels in a variety of studies. In this article, we review the detection methods designed to measure the VEGF levels in different body fluids and prognosticate the value of VEGF in treatment, diagnosis and survival in lung cancer.
Collapse
Affiliation(s)
- Sumin Guo
- Department of Oncology, Hebei Chest Hospital, Lung Cancer Control and Prevention Center of Hebei Province, Shijiazhuang, Hebei, 050041, China.,Department of Orthopaedic Surgery- Campbell Clinic and Pathology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Michael G Martin
- West Cancer Center, University of Tennessee Health Science Center, Memphis, Tennessee, 38163, USA
| | - Cheng Tian
- Department of Orthopaedic Surgery- Campbell Clinic and Pathology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Jinglin Cui
- Department of Orthopaedic Surgery- Campbell Clinic and Pathology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Center of Integrative Research, The First Hospital of Qiqihaer City, Qiqihaer, Heilongjiang, 161005, PR China
| | - Lishi Wang
- Department of Basic Medicine (Basic Medical Research), Inner Mongolia Medical University, Inner Mongolia, 010110, PR China
| | - Shucai Wu
- Department of Oncology, Hebei Chest Hospital, Lung Cancer Control and Prevention Center of Hebei Province, Shijiazhuang, Hebei, 050041, China
| | - Weikuan Gu
- Department of Orthopaedic Surgery- Campbell Clinic and Pathology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Research Service, Veterans Affairs Medical Center, 1030 Jefferson Avenue, Memphis TN 38104, USA
| |
Collapse
|
42
|
Aguilar-Company J, Fernández-Ruiz M, García-Campelo R, Garrido-Castro AC, Ruiz-Camps I. ESCMID Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the safety of targeted and biological therapies: an infectious diseases perspective (Cell surface receptors and associated signaling pathways). Clin Microbiol Infect 2018; 24 Suppl 2:S41-S52. [PMID: 29426804 DOI: 10.1016/j.cmi.2017.12.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 12/18/2017] [Accepted: 12/30/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND The present review is part of the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Infections in Compromised Hosts (ESGICH) consensus document on the safety of targeted and biologic therapies. AIMS To review, from an infectious diseases perspective, the safety profile of therapies targeting cell surface receptors and associated signaling pathways among cancer patients and to suggest preventive recommendations. SOURCES Computer-based Medline searches with MeSH terms pertaining to each agent or therapeutic family. CONTENT Vascular endothelial growth factor (VEGF)-targeted agents (bevacizumab and aflibercept) are associated with a meaningful increase in the risk of infection, likely due to drug-induced neutropaenia, although no clear benefit is expected from the universal use of anti-infective prophylaxis. VEGF tyrosine kinase inhibitors (i.e. sorafenib or sunitinib) do not seem to significantly affect host's susceptibility to infection, and universal anti-infective prophylaxis is not recommended either. Anti-epidermal growth factor receptor (EGFR) monoclonal antibodies (cetuximab or panitumumab) induce neutropaenia and secondary skin and soft tissue infection in cases of severe papulopustular rash. Systemic antibiotics (doxycycline or minocycline) should be administered to prevent the latter complication, whereas no recommendation can be established on the benefit from antiviral, antifungal or anti-Pneumocystis prophylaxis. A lower risk of infection is reported for anti-ErbB2/HER2 monoclonal antibodies (trastuzumab and pertuzumab) and ErbB receptor tyrosine kinase inhibitors (including dual-EGFR/ErbB2 inhibitors such as lapatinib or neratinib) compared to conventional chemotherapy, presumably as a result of the decreased occurrence of drug-induced neutropaenia. IMPLICATIONS With the exception of VEGF-targeted agents, the overall risk of infection associated with the reviewed therapies seems to be low.
Collapse
Affiliation(s)
- J Aguilar-Company
- Departments of Infectious Diseases and Oncology, University Hospital Vall d'Hebron, Autonomous University of Barcelona, Barcelona, Spain
| | - M Fernández-Ruiz
- Unit of Infectious Diseases, Hospital Universitario '12 de Octubre', Instituto de Investigación Hospital '12 de Octubre' (i + 12), School of Medicine, Universidad Complutense, Madrid, Spain; Spanish Network for Research in Infectious Diseases (REIPI RD16/0016), Instituto de Salud Carlos III, Madrid, Spain
| | - R García-Campelo
- Department of Medical Oncology, Complejo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - A C Garrido-Castro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - I Ruiz-Camps
- Department of Infectious Diseases, University Hospital Vall d'Hebron, Autonomous University of Barcelona, Barcelona, Spain; Spanish Network for Research in Infectious Diseases (REIPI RD16/0016), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
43
|
Katas H, Thian Sian T, Abdul Ghaf M. Topical Temperature-sensitive Gel Containing DsiRNA-chitosan Nanoparticles for Potential Treatment of Skin Cancer. ACTA ACUST UNITED AC 2017. [DOI: 10.3923/tmr.2017.1.13] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
44
|
Lin CY, Cho CF, Bai ST, Liu JP, Kuo TT, Wang LJ, Lin YS, Lin CC, Lai LC, Lu TP, Hsieh CY, Chu CN, Cheng DC, Sher YP. ADAM9 promotes lung cancer progression through vascular remodeling by VEGFA, ANGPT2, and PLAT. Sci Rep 2017; 7:15108. [PMID: 29118335 PMCID: PMC5678093 DOI: 10.1038/s41598-017-15159-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 10/23/2017] [Indexed: 12/27/2022] Open
Abstract
Lung cancer has a very high prevalence of brain metastasis, which results in a poor clinical outcome. Up-regulation of a disintegrin and metalloproteinase 9 (ADAM9) in lung cancer cells is correlated with metastasis to the brain. However, the molecular mechanism underlying this correlation remains to be elucidated. Since angiogenesis is an essential step for brain metastasis, microarray experiments were used to explore ADAM9-regulated genes that function in vascular remodeling. The results showed that the expression levels of vascular endothelial growth factor A (VEGFA), angiopoietin-2 (ANGPT2), and tissue plasminogen activator (PLAT) were suppressed in ADAM9-silenced cells, which in turn leads to decreases in angiogenesis, vascular remodeling, and tumor growth in vivo. Furthermore, simultaneous high expression of ADAM9 and VEGFA or of ADAM9 and ANGPT2 was correlated with poor prognosis in a clinical dataset. These findings suggest that ADAM9 promotes tumorigenesis through vascular remodeling, particularly by increasing the function of VEGFA, ANGPT2, and PLAT.
Collapse
Affiliation(s)
- Chen-Yuan Lin
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, 404, Taiwan.,Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan.,Division of Hematology and Oncology, China Medical University Hospital, Taichung, 404, Taiwan
| | - Chia-Fong Cho
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Shih-Ting Bai
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Jing-Pei Liu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan
| | - Ting-Ting Kuo
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Li-Ju Wang
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Yu-Sen Lin
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, 404, Taiwan.,Division of Thoracic Surgery, China Medical University Hospital, Taichung, 404, Taiwan
| | - Ching-Chan Lin
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, 404, Taiwan.,Division of Hematology and Oncology, China Medical University Hospital, Taichung, 404, Taiwan
| | - Liang-Chuan Lai
- Graduate Institute of Physiology, National Taiwan University, Taipei, 106, Taiwan
| | - Tzu-Pin Lu
- Department of Public Health, National Taiwan University, Taipei, 106, Taiwan
| | - Chih-Ying Hsieh
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Chin-Nan Chu
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, 404, Taiwan.,Department of Radiation Oncology, China Medical University Hospital, Taichung, 404, Taiwan
| | - Da-Chuan Cheng
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, 404, Taiwan.,Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, 404, Taiwan
| | - Yuh-Pyng Sher
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, 404, Taiwan. .,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan. .,Center for Molecular Medicine, China Medical University Hospital, Taichung, 404, Taiwan.
| |
Collapse
|
45
|
Upregulation of vascular endothelial growth factor (VEGF), its role in progression and prognosis of non-small cell lung carcinoma. Cancer Genet 2017; 216-217:67-73. [PMID: 29025597 DOI: 10.1016/j.cancergen.2017.07.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 07/22/2017] [Accepted: 07/23/2017] [Indexed: 11/20/2022]
Abstract
Elevated VEGF mRNA (-ΔCT) was significantly associated with adenocarcinoma histology (vs squamous) and advanced NSCLC clinical stages in a univariable analysis; however, this association did not remain significant in the multivariable analysis. Of interest, a Kaplan-Meier analysis showed that NSCLC patients with higher VEGF mRNA (-ΔCT ≥10) had a significantly poorer overall survival and shorter postoperative relapse time in adenocarcinoma and in stage III/IV than those with VEGF mRNA of -ΔCT <10 (P < 0.001). The multivariable analysis confirmed that patients with higher VEGF mRNA levels, as well as with adenocarcinoma and advanced stages, were independent predictors of a poorer survival. However, only the histology of adenocarcinoma remained a significant prognostic factor of a shorter postoperative relapse in the multivariable model. Quantity of VEGF mRNA can be used as a prognosis factor to predict shorter overall survival in patients with NSCLC.
Collapse
|
46
|
Isolating the Role of Bevacizumab in Elderly Patients With Previously Untreated Nonsquamous Non-Small Cell Lung Cancer: Secondary Analyses of the ECOG 4599 and PointBreak Trials. Am J Clin Oncol 2017; 39:441-7. [PMID: 25628268 PMCID: PMC5016219 DOI: 10.1097/coc.0000000000000163] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Supplemental Digital Content is available in the text. Patient-level data from 2 phase III studies in patients with previously untreated, advanced-stage, nonsquamous non–small cell lung cancer (NSCLC) were pooled to examine outcomes with bevacizumab and chemotherapy based on age.
Collapse
|
47
|
Koinis F, Agelaki S, Karavassilis V, Kentepozidis N, Samantas E, Peroukidis S, Katsaounis P, Hartabilas E, Varthalitis II, Messaritakis I, Fountzilas G, Georgoulias V, Kotsakis A. Second-line pazopanib in patients with relapsed and refractory small-cell lung cancer: a multicentre phase II study of the Hellenic Oncology Research Group. Br J Cancer 2017; 117:8-14. [PMID: 28510571 PMCID: PMC5520202 DOI: 10.1038/bjc.2017.137] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 04/19/2017] [Accepted: 04/24/2017] [Indexed: 01/15/2023] Open
Abstract
Background: Pazopanib is a tyrosine kinase inhibitor with antiangiogenic activity. Vascular endothelial growth factor expression is increased in SCLC and is correlated with poor prognosis. The efficacy and tolerance of second-line pazopanib in SCLC was evaluated. Patients and methods: Patients with platinum-sensitive (cohort A; n=39) and -resistant/refractory (cohort B; n=19) SCLC were enrolled in a multicentre phase II study. The primary end point was the progression-free survival rate (PFS-R) at week 8 in each cohort. Pazopanib (800 mg per day per os) was administered until progressive disease (PD). Circulating tumour cells (CTCs) were enumerated using the Cellsearch assay. Results: All patients were evaluable for response and toxicity. In the intention-to-treat analysis, eight (13.8%) patients achieved partial response (PR) (95% confidence interval (CI): 5.0–22.7), 20 (34.5%) stable disease (SD) and 30 (51.7%) PD. Accrual in cohort B was halted because the hard-stop rule was met; in cohort A, the PFS-R was 59% (95% CI: 43.5–74.4; PR=7, SD=16). Nine (23.1%) patients received pazopanib for >6 months and 3 of them for >12 months. One pazopanib cycle resulted to a significant decrease to the number of patients with ⩾5 CTCs/7.5 ml of blood (20%) compared with baseline (50%). The median PFS and OS for all patients was 2.5 months (95% CI: 1.9–3.1 months) and 6.0 months (95% CI: 3.8–8.2 months), respectively (cohort A: PFS=3.7 months and OS=8.0 months). No unexpected toxicity was observed. Conclusions: Second-line treatment with pazopanib in platinum-sensitive SCLC is well tolerated and resulted in promising objective responses and disease control; CTC enumeration might serve as a reliable surrogate biomarker of response.
Collapse
Affiliation(s)
- F Koinis
- Hellenic Oncology Research Group (HORG), 55, Lombardou Street, Athens 11474, Greece
| | - S Agelaki
- Hellenic Oncology Research Group (HORG), 55, Lombardou Street, Athens 11474, Greece
| | - V Karavassilis
- Hellenic Oncology Research Group (HORG), 55, Lombardou Street, Athens 11474, Greece
| | - N Kentepozidis
- Hellenic Oncology Research Group (HORG), 55, Lombardou Street, Athens 11474, Greece
| | - E Samantas
- Hellenic Oncology Research Group (HORG), 55, Lombardou Street, Athens 11474, Greece
| | - S Peroukidis
- Hellenic Oncology Research Group (HORG), 55, Lombardou Street, Athens 11474, Greece
| | - P Katsaounis
- Hellenic Oncology Research Group (HORG), 55, Lombardou Street, Athens 11474, Greece
| | - E Hartabilas
- Hellenic Oncology Research Group (HORG), 55, Lombardou Street, Athens 11474, Greece
| | - I I Varthalitis
- Hellenic Oncology Research Group (HORG), 55, Lombardou Street, Athens 11474, Greece
| | - I Messaritakis
- Hellenic Oncology Research Group (HORG), 55, Lombardou Street, Athens 11474, Greece
| | - G Fountzilas
- Hellenic Oncology Research Group (HORG), 55, Lombardou Street, Athens 11474, Greece
| | - V Georgoulias
- Hellenic Oncology Research Group (HORG), 55, Lombardou Street, Athens 11474, Greece
| | - A Kotsakis
- Hellenic Oncology Research Group (HORG), 55, Lombardou Street, Athens 11474, Greece
| |
Collapse
|
48
|
Leal-Egaña A, Letort G, Martiel JL, Christ A, Vignaud T, Roelants C, Filhol O, Théry M. The size-speed-force relationship governs migratory cell response to tumorigenic factors. Mol Biol Cell 2017; 28:1612-1621. [PMID: 28428257 PMCID: PMC5469605 DOI: 10.1091/mbc.e16-10-0694] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 03/28/2017] [Accepted: 04/10/2017] [Indexed: 12/18/2022] Open
Abstract
Normal and transformed motile cells follow a common trend in which size and contractile forces are negatively correlated with cell speed. However, tumorigenic factors amplify the preexisting population heterogeneity and lead some cells to exhibit biomechanical properties that are more extreme than those observed with normal cells. Tumor development progresses through a complex path of biomechanical changes leading first to cell growth and contraction and then cell deadhesion, scattering, and invasion. Tumorigenic factors may act specifically on one of these steps or have a wider spectrum of actions, leading to a variety of effects and thus sometimes to apparent contradictory outcomes. Here we used micropatterned lines of collagen type I/fibronectin on deformable surfaces to standardize cell behavior and measure simultaneously cell size, speed of motion and magnitude of the associated traction forces at the level of a single cell. We analyzed and compared the normal human breast cell line MCF10A in control conditions and in response to various tumorigenic factors. In all conditions, a wide range of biomechanical properties was identified. Despite this heterogeneity, normal and transformed motile cells followed a common trend whereby size and contractile forces were negatively correlated with cell speed. Some tumorigenic factors, such as activation of ErbB2 or loss of the βsubunit of casein kinase 2, shifted the whole population toward a faster speed and lower contractility state. Treatment with transforming growth factor β induced some cells to adopt opposing behaviors such as extremely high versus extremely low contractility. Thus tumor transformation amplified preexisting population heterogeneity and led some cells to exhibit biomechanical properties that were more extreme than those observed with normal cells.
Collapse
Affiliation(s)
- Aldo Leal-Egaña
- CytoMorpho Lab, LPCV, Biosciences and Biotechnology Institute of Grenoble, UMR5168, CEA, CNRS, INRA, Université Grenoble-Alpes, 38054 Grenoble, France
| | - Gaelle Letort
- CytoMorpho Lab, LPCV, Biosciences and Biotechnology Institute of Grenoble, UMR5168, CEA, CNRS, INRA, Université Grenoble-Alpes, 38054 Grenoble, France
| | - Jean-Louis Martiel
- CytoMorpho Lab, LPCV, Biosciences and Biotechnology Institute of Grenoble, UMR5168, CEA, CNRS, INRA, Université Grenoble-Alpes, 38054 Grenoble, France
| | - Andreas Christ
- CytoMorpho Lab, LPCV, Biosciences and Biotechnology Institute of Grenoble, UMR5168, CEA, CNRS, INRA, Université Grenoble-Alpes, 38054 Grenoble, France
| | - Timothée Vignaud
- CytoMorpho Lab, LPCV, Biosciences and Biotechnology Institute of Grenoble, UMR5168, CEA, CNRS, INRA, Université Grenoble-Alpes, 38054 Grenoble, France
| | - Caroline Roelants
- Biologie du Cancer et de l'Infection, Biosciences and Biotechnology Institute of Grenoble, UMRS1036, CEA, INSERM, CNRS, Université Grenoble-Alpes, 38054 Grenoble, France
| | - Odile Filhol
- Biologie du Cancer et de l'Infection, Biosciences and Biotechnology Institute of Grenoble, UMRS1036, CEA, INSERM, CNRS, Université Grenoble-Alpes, 38054 Grenoble, France
| | - Manuel Théry
- CytoMorpho Lab, LPCV, Biosciences and Biotechnology Institute of Grenoble, UMR5168, CEA, CNRS, INRA, Université Grenoble-Alpes, 38054 Grenoble, France .,CytoMorpho Lab, A2T, Hopital Saint Louis, Institut Universitaire d'Hematologie, UMRS1160, CEA, INSERM, AP-HP, Université Paris Diderot, 75010 Paris, France
| |
Collapse
|
49
|
Vascular endothelial growth factor A amplification in colorectal cancer is associated with reduced M1 and M2 macrophages and diminished PD-1-expressing lymphocytes. PLoS One 2017; 12:e0175563. [PMID: 28403223 PMCID: PMC5389821 DOI: 10.1371/journal.pone.0175563] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 03/28/2017] [Indexed: 01/22/2023] Open
Abstract
VEGFA is an angiogenic factor secreted by tumors, in particular those with VEGFA amplification, as well as by macrophages and lymphocytes in the tumor microenvironment. Here we sought to define the presence of M1/M2 macrophages, PD-1-positive lymphocytes and PD-L1 tumoral and stromal expression in colorectal cancers harboring VEGFA amplification or chromosome 6 polysomy. 38 CRCs of which 13 harbored VEGFA amplification, 6 with Chr6 polysomy and 19 with neutral VEGFA copy number were assessed by immunohistochemistry for CD68 (marker for M1/M2 macrophages), CD163 (M2 macrophages), programmed death 1(PD-1)- tumor infiltrating and stromal lymphocytes as well as tumoral and stromal PD-1 ligand (PD-L1) expression. CRCs with VEGFA amplification or Chr6 polysomy were associated with decreased M1/M2 macrophages, reduced PD-1-expressing lymphocyte infiltration, as well as reduced stromal expression of PD-L1 at the tumor front. Compared to intermediate-grade CRCs, high-grade CRCs were associated with increased M1/M2 macrophages and increased tumoral expression of PD-L1. Our results suggest that VEGFA amplification or Chr6 polysomy is associated with an altered tumor immune microenvironment.
Collapse
|
50
|
Usmanij EA, Natroshvili T, Timmer-Bonte JN, Oyen WJ, van der Drift MA, Bussink J, Geus-Oei LFD. The Predictive Value of Early In-Treatment 18F-FDG PET/CT Response to Chemotherapy in Combination with Bevacizumab in Advanced Nonsquamous Non–Small Cell Lung Cancer. J Nucl Med 2017; 58:1243-1248. [DOI: 10.2967/jnumed.116.185314] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 01/31/2017] [Indexed: 01/25/2023] Open
|