1
|
Ochirbat S, Kan TC, Hsu CC, Huang TH, Chuang KH, Chen M, Cheng CC, Chang CC, Rahayu S, Chang J. The angiogenic role of the alpha 9-nicotinic acetylcholine receptor in triple-negative breast cancers. Angiogenesis 2024:10.1007/s10456-024-09944-6. [PMID: 39177676 DOI: 10.1007/s10456-024-09944-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Nicotine acts as an angiogenic factor by stimulating endogenous cholinergic pathways. Several subtypes of nicotinic acetylcholine receptors (nAChRs) have been demonstrated to be closely correlated to the formation and progression of different types of cancers. Recently, several studies have found that nicotinic acetylcholine receptors α9 (α9-nAChRs) are highly expressed in breast tumors, especially in tumors derived from patients diagnosed at advanced stages. In vitro studies have demonstrated that activation of α9-nAChRs is associated with increased proliferation and migration of breast cancer. To study the tumor-promoting role of α9-nAChRs in breast cancers, we generated a novel anti-α9-nAChR and methoxy-polyethylene glycol (mPEG) bispecific antibody (α9 BsAb) for dissecting the molecular mechanism on α9-nAChR-mediated tumor progression. Unexpectedly, we discovered the angiogenic role of α9-nAChR in nicotine-induced neovascularization of tumors. It revealed α9 BsAbs reduced nicotine-induced endothelial cell tube formation, blood vessel development in Matrigel plug assay and angiogenesis in microtube array membrane murine model (MTAMs). To unbraid the molecular mechanism of α9-nAChR in nicotine-mediated angiogenesis, the α9 BsAbs were applied and revealed the inhibitory roles in nicotine-induced production of hypoxia-inducible factor-2 alpha (HIF-2α), vascular endothelial growth factor A (VEGF-A), phosphorylated vascular endothelial growth factor receptor 2 (p-VEGFR2), vascular endothelial growth factor receptor 2 (VEGFR2) and matrix metalloproteinase-9 (MMP9) from triple-negative breast cancer cells (MDA-MB-231), suggesting α9-nAChRs played an important role in nicotine-induced angiogenesis. To confirm our results, the shRNA targeting α9-nAChRs was designed and used to silence α9-nAChR expression and then evaluated the angiogenic role of α9-nAChRs. The results showed α9 shRNA also played an inhibitory effect in blocking the nicotine-induced angiogenic signaling. Taken together, α9-nAChR played a critical role in nicotine-induced angiogenesis and this bispecific antibody (α9 BsAb) may serve as a potential therapeutic candidate for treatments of the α9 positive cancers.
Collapse
Affiliation(s)
- Sonjid Ochirbat
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Tzu-Chun Kan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Chun-Chun Hsu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, 11031, Taiwan
| | - Tzu-Hsuan Huang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
| | - Kuo-Hsiang Chuang
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, 11031, Taiwan
| | - Michael Chen
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chun-Chia Cheng
- Research Center of Radiation Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Chun-Chao Chang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, 11031, Taiwan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Sri Rahayu
- Department of Biology, Faculty of Mathematics and Natural Science, Universitas Negeri Jakarta, Jakarta, 13220, Indonesia
| | - Jungshan Chang
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
2
|
Gao XQ, Li HL, Wang M, Yang CT, Su R, Shao LH. Kaempferol inhibited invasion and metastasis of gastric cancer cells by targeting AKT/GSK3β pathway based on network pharmacology and molecular docking. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024:1-21. [PMID: 39132822 DOI: 10.1080/10286020.2024.2387756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024]
Abstract
This study aims to explore the mechanisms of the inhibitory effect of kaempferol on the invasion and metastasis of gastric cancer (GC) cells through network pharmacology prediction and experimental verification. It identifies core targets via PPI network analysis and finds that kaempferol binds to these targets well. In vitro experiments showed that kaempferol could inhibit the proliferation, colony formation, migration and invasion of GC cells. Western blotting indicated kaempferol may reduce AKT and GSK3β phosphorylation, leading to lower expression of invasion-related genes SRC, MMP9, CXCR4, KDR, and MMP2. Overall, kaempferol may prevent migration and invasion of GC cells via the AKT/GSK3β signaling pathway.
Collapse
Affiliation(s)
- Xia-Qing Gao
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Hai-Long Li
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
- Department of Geriatrics, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Meng Wang
- Department of Geriatrics, Affiliated Hongqi Hospital of Mudanjiang Medical University, Mudanjiang 157011, China
- Research Center of Traditional Chinese Medicine, Gansu Province, Lanzhou 730000, China
| | - Chun-Ting Yang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Rong Su
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Li-Hua Shao
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
| |
Collapse
|
3
|
Fu L, Zhao L, Li F, Wen F, Zhang P, Yang X, Wang Y. Pharmacological mechanism of quercetin in the treatment of colorectal cancer by network pharmacology and molecular simulation. J Biomol Struct Dyn 2024; 42:7065-7076. [PMID: 37464874 DOI: 10.1080/07391102.2023.2235589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 07/06/2023] [Indexed: 07/20/2023]
Abstract
Colorectal cancer is a serious threat to people's life due to its high incidence and high mortality. Quercetin can effectively treat colorectal carcinoma (CRC), but its exact mechanism of action is still unclear. Then quercetin-related target genes were obtained from Swiss Target Prediction database and Similarity Ensemble Approach (SEA) database, and CRC-related target genes were obtained from GeneCards database, respectively. Common target genes were obtained by FunRich software. String software was used to construct a protein-protein interaction (PPI) network. R package was used for gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Molecular docking, molecular dynamics (MD) simulation and post-dynamics simulation were used to explore the binding stability of quercetin to key targets. In total, 103 and 141 target information of quercetin were obtained from the Swiss Target Prediction database and SEA database, respectively. 1,649 CRC-related genes were obtained from GeneCards database. FunRich software was used to draw venny map and obtain 36 intersection targets of quercetin and CRC. String software was used to construct the PPI network. The core genes were AKT1, EGFR, MMP9, KDR, MET and PTK2. There were 532 items related to biological processes, 14 items related to cellular components, and 43 items related to molecular functions among the key target GO enrichment items. KEGG enrichment pathways of key targets involved cancer pathways, PI3K-Akt signal pathway, etc. The results of molecular docking, MD simulation and post-dynamics simulation showed they had a good affinity and formed a stable effect. So quercetin may play an important role in the treatment of CRC by acting on AKT1, EGFR, MMP9, KDR, MET and PTK2 to affect the development of CRC.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Le Fu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
- Chongqing University Qianjiang Hospital (Qianjiang Central Hospital of Chongqing), Chongqing, China
| | - Linan Zhao
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
- Chongqing University Qianjiang Hospital (Qianjiang Central Hospital of Chongqing), Chongqing, China
| | - Fei Li
- Chongqing University Qianjiang Hospital (Qianjiang Central Hospital of Chongqing), Chongqing, China
| | - Feng Wen
- Chongqing University Qianjiang Hospital (Qianjiang Central Hospital of Chongqing), Chongqing, China
| | - Peng Zhang
- Chongqing University Qianjiang Hospital (Qianjiang Central Hospital of Chongqing), Chongqing, China
| | - Xia Yang
- Chongqing University Qianjiang Hospital (Qianjiang Central Hospital of Chongqing), Chongqing, China
| | - Yuanqiang Wang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| |
Collapse
|
4
|
Wang H, Shi P, Shi X, Lv Y, Xie H, Zhao H. Surprising magic of CD24 beyond cancer. Front Immunol 2024; 14:1334922. [PMID: 38313430 PMCID: PMC10834733 DOI: 10.3389/fimmu.2023.1334922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/28/2023] [Indexed: 02/06/2024] Open
Abstract
CD24 has emerged as a molecule of significant interest beyond the oncological arena. Recent studies have unveiled its surprising and diverse roles in various biological processes and diseases. This review encapsulates the expanding spectrum of CD24 functions, delving into its involvement in immune regulation, cancer immune microenvironment, and its potential as a therapeutic target in autoimmune diseases and beyond. The 'magic' of CD24, once solely attributed to cancer, now inspires a new paradigm in understanding its multifunctionality in human health and disease, offering exciting prospects for medical advancements.
Collapse
Affiliation(s)
- He Wang
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Peng Shi
- Department of Emergency Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xinyu Shi
- Department of Radiology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yaqing Lv
- Department of Outpatient, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongwei Xie
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hai Zhao
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
5
|
Li D, Wang L, Jiang B, Jing Y, Li X. Improving cancer immunotherapy by preventing cancer stem cell and immune cell linking in the tumor microenvironment. Biomed Pharmacother 2024; 170:116043. [PMID: 38128186 DOI: 10.1016/j.biopha.2023.116043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/07/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Cancer stem cells are the key link between malignant tumor progression and drug resistance. This cell population has special properties that are different from those of conventional tumor cells, and the role of cancer stem cell-related exosomes in progression of tumor malignancy is becoming increasingly clear. Cancer stem cell-derived exosomes carry a variety of functional molecules involved in regulation of the microenvironment, especially with regard to immune cells, but how these exosomes exert their functions and the specific mechanisms need to be further clarified. Here, we summarize the role of cancer stem cell exosomes in regulating immune cells in detail, aiming to provide new insights for subsequent targeted drug development and clinical strategy formulation.
Collapse
Affiliation(s)
- Dongyu Li
- Department of General Surgery & VIP In-Patient Ward, the First Hospital of China Medical University, Liaoning Province 110001, China
| | - Lei Wang
- Department of Vascular and Thyroid Surgery, the First Hospital of China Medical University, Liaoning Province 110001, China
| | - Bo Jiang
- Department of Vascular and Thyroid Surgery, the First Hospital of China Medical University, Liaoning Province 110001, China
| | - Yuchen Jing
- Department of Vascular and Thyroid Surgery, the First Hospital of China Medical University, Liaoning Province 110001, China
| | - Xuan Li
- Department of Vascular and Thyroid Surgery, the First Hospital of China Medical University, Liaoning Province 110001, China.
| |
Collapse
|
6
|
Onda M, Ota A, Ito K, Ono T, Karnan S, Kato M, Kondo S, Furuhashi A, Hayashi T, Hosokawa Y, Kazaoka Y. Inhibition of VEGFR2 and EGFR signaling cooperatively suppresses the proliferation of oral squamous cell carcinoma. Cancer Med 2023; 12:16416-16430. [PMID: 37341071 PMCID: PMC10469792 DOI: 10.1002/cam4.6282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/05/2023] [Accepted: 06/12/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) is frequently overexpressed in oral squamous cell carcinoma (OSCC), and EGFR-targeting therapeutics have been widely employed to treat patients with a variety of carcinomas including OSCC. Here, we aimed to investigate alternative signaling for OSCC survival under the disruption of EGFR signaling. METHODS OSCC cell lines, namely HSC-3 and SAS, were utilized to investigate how EGFR disruption affects cell proliferation. Gene set enrichment analysis was performed to examine how EGFR disruption affects oncogenic signaling in OSCC cells. Disruption of KDR gene was performed using CRISPR/Cas9 techniques. A VEGFR inhibitor, vatalanib was used to research the impact of VEGFR inhibition on OSCC survival. RESULTS EGFR disruption significantly decreased the proliferation and oncogenic signaling including Myc and PI3K-Akt, in OSCC cells. Chemical library screening assays revealed that VEGFR inhibitors continued to inhibit the proliferation of EGFR-deficient OSCC cells. In addition, CRISPR-mediated disruption of KDR/VEGFR2 retarded OSCC cell proliferation. Furthermore, combined erlotinib-vatalanib treatment exhibited a more potent anti-proliferative effect on OSCC cells, compared to either monotherapy. The combined therapy effectively suppressed the phosphorylation levels of Akt but not p44/42. CONCLUSION VEGFR-mediated signaling would be an alternative signaling pathway for the survival of OSCC cells under the disruption of EGFR signaling. These results highlight the clinical application of VEGFR inhibitors in the development of multi-molecular-targeted therapeutics against OSCC.
Collapse
Affiliation(s)
- Maho Onda
- Department of Oral and Maxillofacial SurgeryAichi Medical University HospitalNagakuteJapan
| | - Akinobu Ota
- Department of BiochemistryAichi Medical University School of MedicineNagakuteJapan
- Department of Food and Nutritional EnvironmentCollege of Human Life and EnvironmentKinjo Gakuin UniversityNagoyaJapan
| | - Kunihiro Ito
- Department of Oral and Maxillofacial SurgeryAichi Medical University HospitalNagakuteJapan
| | - Takayuki Ono
- Department of Oral and Maxillofacial SurgeryAichi Medical University HospitalNagakuteJapan
| | - Sivasundaram Karnan
- Department of BiochemistryAichi Medical University School of MedicineNagakuteJapan
| | - Mikako Kato
- Department of Oral and Maxillofacial SurgeryAichi Medical University HospitalNagakuteJapan
| | - Sayuri Kondo
- Department of Oral and Maxillofacial SurgeryAichi Medical University HospitalNagakuteJapan
| | - Akifumi Furuhashi
- Department of Oral and Maxillofacial SurgeryAichi Medical University HospitalNagakuteJapan
| | - Tomio Hayashi
- Department of Oral and Maxillofacial SurgeryAichi Medical University HospitalNagakuteJapan
| | - Yoshitaka Hosokawa
- Department of BiochemistryAichi Medical University School of MedicineNagakuteJapan
| | - Yoshiaki Kazaoka
- Department of Oral and Maxillofacial SurgeryAichi Medical University HospitalNagakuteJapan
| |
Collapse
|
7
|
Wang Y, Yu H, Yu M, Liu H, Zhang B, Wang Y, Zhao S, Xia Q. CD24 blockade as a novel strategy for cancer treatment. Int Immunopharmacol 2023; 121:110557. [PMID: 37379708 DOI: 10.1016/j.intimp.2023.110557] [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: 10/08/2022] [Revised: 05/22/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023]
Abstract
The CD24 protein is a heat-stable protein with a small core that undergoes extensive glycosylation. It is expressed on the surface of various normal cells, including lymphocytes, epithelial cells, and inflammatory cells. CD24 exerts its function by binding to different ligands. Numerous studies have demonstrated the close association of CD24 with tumor occurrence and progression. CD24 not only facilitates tumor cell proliferation, metastasis, and immune evasion but also plays a role in tumor initiation, thus, serving as a marker on the surface of cancer stem cells (CSCs). Additionally, CD24 induces drug resistance in various tumor cells following chemotherapy. To counteract the tumor-promoting effects of CD24, several treatment strategies targeting CD24 have been explored, such as the use of CD24 monoclonal antibodies (mAb) alone, the combination of CD24 and chemotoxic drugs, or the combination of these drugs with other targeted immunotherapeutic techniques. Regardless of the approach, targeting CD24 has demonstrated significant anti-tumor effects. Therefore, the present study focuses on anti-tumor therapy and provides a comprehensive review of the structure and fundamental physiological function of CD24 and its impact on tumor development, and suggests that targeting CD24 may represent an effective strategy for treating malignant tumors.
Collapse
Affiliation(s)
- Yawen Wang
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China; Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou 450008, China; Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou 450008, China; Henan Engineering Research Center of Pathological Diagnostic Antibody, Zhengzhou 450008, China
| | - Haoran Yu
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China; Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou 450008, China; Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou 450008, China; Henan Engineering Research Center of Pathological Diagnostic Antibody, Zhengzhou 450008, China
| | - Mengyuan Yu
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China; Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou 450008, China; Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou 450008, China; Henan Engineering Research Center of Pathological Diagnostic Antibody, Zhengzhou 450008, China
| | - Hui Liu
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China
| | - Bing Zhang
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China; Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou 450008, China
| | - Yuanyuan Wang
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China; Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou 450008, China; Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou 450008, China
| | - Simin Zhao
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China; Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou 450008, China; Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou 450008, China.
| | - Qingxin Xia
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China; Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou 450008, China; Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou 450008, China; Henan Engineering Research Center of Pathological Diagnostic Antibody, Zhengzhou 450008, China.
| |
Collapse
|
8
|
Zhang Y, Wu J, Zhao C, Zhang S, Zhu J. Recent Advancement of PD-L1 Detection Technologies and Clinical Applications in the Era of Precision Cancer Therapy. J Cancer 2023; 14:850-873. [PMID: 37056391 PMCID: PMC10088895 DOI: 10.7150/jca.81899] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/14/2023] [Indexed: 04/15/2023] Open
Abstract
Programmed death-1 is a protein found on the surface of immune cells that can interact with its ligand, programmed death-ligand 1 (PD-L1), which is expressed on the plasma membrane, the surface of secreted cellular exosomes, in cell nuclei, or as a circulating soluble protein. This interaction can lead to immune escape in cancer patients. In clinical settings, PD-L1 plays an important role in tumor disease diagnosis, determining therapeutic effectiveness, and predicting patient prognosis. PD-L1 inhibitors are also essential components of tumor immunotherapy. Thus, the detection of PD-L1 levels is crucial, especially in the era of precision cancer therapy. In recent years, innovations have been made in traditional immunoassay methods and the development of new immunoassays for PD-L1 detection. This review aims to summarize recent research progress in tumor PD-L1 detection technology and highlight the clinical applications of PD-L1.
Collapse
Affiliation(s)
- Yuanfeng Zhang
- Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Juanjuan Wu
- Binzhou People's Hospital Affiliated to Shandong First Medical University, Binzhou, Shandong, 256600, China
| | - Chaobin Zhao
- Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Shuyuan Zhang
- Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Jianbo Zhu
- Binzhou People's Hospital Affiliated to Shandong First Medical University, Binzhou, Shandong, 256600, China
- ✉ Corresponding author: Pro. Jianbo Zhu, Binzhou People's Hospital Affiliated to Shandong First Medical University, 515 Yellow River Seven Road, Binzhou, Shandong, 256600, China; ,
| |
Collapse
|
9
|
Shen Q, He Y, Qian J, Wang X. Identifying tumor immunity-associated molecular features in liver hepatocellular carcinoma by multi-omics analysis. Front Mol Biosci 2022; 9:960457. [PMID: 36339710 PMCID: PMC9632276 DOI: 10.3389/fmolb.2022.960457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 10/10/2022] [Indexed: 12/01/2022] Open
Abstract
Background: Although current immunotherapies have achieved some successes for hepatocellular carcinoma (HCC) patients, their benefits are limited for most HCC patients. Therefore, the identification of biomarkers for promoting immunotherapeutic responses in HCC is urgently needed. Methods: Using the TCGA HCC cohort, we investigated correlations of various molecular features with antitumor immune signatures (CD8+ T cell infiltration and cytolytic activity) and an immunosuppressive signature (PD-L1 expression) in HCC. These molecular features included mRNAs, microRNAs (miRNAs), long non-coding RNAs (lncRNAs), proteins, and pathways. Results: We found that the mutations of several oncogenes and tumor suppressor genes significantly correlated with reduced antitumor immune signatures, including TTN, CTNNB1, RB1, ZFHX4, and TP53. It indicates that these genes’ mutations may inhibit antitumor immune responses in HCC. Four proteins (Syk, Lck, STAT5, and Caspase-7) had significant positive expression correlations with CD8+ T cell enrichment, cytolytic activity, and PD-L1 expression in HCC. It suggests that these proteins’ expression could be useful biomarkers for the response to immune checkpoint inhibitors Similiarly, we identified other types of biomarkers potentially useful for predicting the response to ICIs, including miRNAs (hsa-miR-511-5p, 150-3p, 342-3p, 181a-3p, 625-5p, 4772-3p, 155-3p, 142-5p, 142-3p, 155-5p, 625-3p, 1976, 7702), many lncRNAs, and pathways (apoptosis, cytokine-cytokine receptor interaction, Jak-STAT signaling, MAPK signaling, PI3K-AKT signaling, HIF-1 signaling, ECM receptor interaction, focal adhesion, and estrogen signaling). Further, tumor mutation burden showed no significant correlation with antitumor immunity, while tumor aneuploidy levels showed a significant negative correlation with antitumor immunity. Conclusion: The molecular features significantly associated with HCC immunity could be predictive biomarkers for immunotherapeutic responses in HCC patients. They could also be potential intervention targets for boosting antitumor immunity and immunotherapeutic responses in HCC.
Collapse
Affiliation(s)
- Qianyun Shen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yin He
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, China
| | - Jiajie Qian
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, China
- *Correspondence: Xiaosheng Wang,
| |
Collapse
|
10
|
Jinesh GG, Brohl AS. Classical epithelial-mesenchymal transition (EMT) and alternative cell death process-driven blebbishield metastatic-witch (BMW) pathways to cancer metastasis. Signal Transduct Target Ther 2022; 7:296. [PMID: 35999218 PMCID: PMC9399134 DOI: 10.1038/s41392-022-01132-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 07/14/2022] [Accepted: 07/24/2022] [Indexed: 12/13/2022] Open
Abstract
Metastasis is a pivotal event that accelerates the prognosis of cancer patients towards mortality. Therapies that aim to induce cell death in metastatic cells require a more detailed understanding of the metastasis for better mitigation. Towards this goal, we discuss the details of two distinct but overlapping pathways of metastasis: a classical reversible epithelial-to-mesenchymal transition (hybrid-EMT)-driven transport pathway and an alternative cell death process-driven blebbishield metastatic-witch (BMW) transport pathway involving reversible cell death process. The knowledge about the EMT and BMW pathways is important for the therapy of metastatic cancers as these pathways confer drug resistance coupled to immune evasion/suppression. We initially discuss the EMT pathway and compare it with the BMW pathway in the contexts of coordinated oncogenic, metabolic, immunologic, and cell biological events that drive metastasis. In particular, we discuss how the cell death environment involving apoptosis, ferroptosis, necroptosis, and NETosis in BMW or EMT pathways recruits immune cells, fuses with it, migrates, permeabilizes vasculature, and settles at distant sites to establish metastasis. Finally, we discuss the therapeutic targets that are common to both EMT and BMW pathways.
Collapse
Affiliation(s)
- Goodwin G Jinesh
- Department of Molecular Oncology, 12902 USF Magnolia Drive, H. Lee Moffitt Cancer Center & Research Institute, Tampa, 33612, FL, USA. .,Sarcoma Department, 12902 USF Magnolia Drive, H. Lee Moffitt Cancer Center & Research Institute, Tampa, 33612, FL, USA.
| | - Andrew S Brohl
- Department of Molecular Oncology, 12902 USF Magnolia Drive, H. Lee Moffitt Cancer Center & Research Institute, Tampa, 33612, FL, USA. .,Sarcoma Department, 12902 USF Magnolia Drive, H. Lee Moffitt Cancer Center & Research Institute, Tampa, 33612, FL, USA.
| |
Collapse
|
11
|
Wang F, Liu G. Influence of KDR Genetic Variation on the Effectiveness and Safety of Bevacizumab in the First-Line Treatment for Patients with Advanced Colorectal Cancer. Int J Gen Med 2022; 15:5651-5659. [PMID: 35734201 PMCID: PMC9208669 DOI: 10.2147/ijgm.s362366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/21/2022] [Indexed: 11/23/2022] Open
Abstract
Objective Bevacizumab is usually considered a first-line anti-tumor therapy, which inhibits tumor growth by downregulating the vascular endothelial growth factor (VEGF) that further silences the activity of the kinase insert region receptor (KDR) gene. In the current study, we investigated the treatment response of bevacizumab in advanced colorectal cancer (CRC) patients bearing 889 C>T mutation in the KDR gene. Methods A total of 135 advanced CRC patients were treated with bevacizumab along with chemotherapy at the seventh medical center of the People’s Liberation Army general hospital from January 2012 to June 2021 and were analyzed retrospectively. The KDR genotyping and mRNA expression analyses were performed in 57 patients. Results The KDR genotyping revealed 97 (71.85%) cases with CC genotype, 34 (25.19%) cases with CT, and 4 (2.96%) cases with TT genotype, while the minor allele frequency of 889 C>T was found as 0.16. The median progression-free survival (PFS) of the patients with CT/TT genotype and CC genotype was found to be 6.1 and 9.7 months, respectively (P = 0.009). The median overall survival (OS) of the two genotypes was 13.7 and 19.7 (P = 0.025), respectively. Multivariable Cox regression analysis of PFS, CT/TT genotype was found to be an independent factor for PFS (odds ratio (OR) = 1.88, P = 0.023). Additionally, the mRNA expression of KDR in 57 biopsies taken from patients with CT/TT genotypes was significantly higher than that of patients with CC genotype (P < 0.001). Additionally, in terms of safety, 55 patients experienced grade 2 or higher fatigue (incidence rate 40.74%) after receiving bevacizumab along with chemotherapy. Conclusion The 889 C>T mutation in KDR gene affects the KDR expression in colorectal cancer patients, thereby affecting the effectiveness of bevacizumab therapy.
Collapse
Affiliation(s)
- Fei Wang
- Department of Oncology, The Seventh Medical Center of People's Liberation Army General Hospital, Beijing, People's Republic of China
| | - Gang Liu
- Department of General Surgery, The First Medical Center of People's Liberation Army General Hospital, Beijing, People's Republic of China
| |
Collapse
|
12
|
Li X, Cheng Y, Zhu B, Geng M, Yan P, Hu M. Implication of VEGFR2 Polymorphism on the Prognosis of Anlotinib Monotherapy for Patients With Treatment-Refractory Advanced NSCLC: An Exploratory Study. Technol Cancer Res Treat 2022; 21:15330338221080993. [PMID: 35443836 PMCID: PMC9047798 DOI: 10.1177/15330338221080993] [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: 12/24/2022] Open
Abstract
Background: This study aimed to investigate the implication of
Vascular Endothelial Growth Factor Receptor 2
(VEGFR2) polymorphism on the prognosis of anlotinib
monotherapy among patients with treatment-refractory advanced nonsmall cell lung
cancer (NSCLC). Methods: Designed as a retrospective study, this
study included a total of 129 patients with treatment-refractory advanced NSCLC
who were administered with anlotinib monotherapy. The efficacy of the patients
was assessed regularly. The prognosis was performed and adverse reactions during
anlotinib administration were collected. Available and appropriate biological
specimens of the 129 patients were collected to perform VEGFR2
polymorphism analysis and VEGFR2 gene mRNA expression analysis
accordingly. Association analysis between genotype status of
VEGFR2 polymorphism and other variables was implemented in
univariate and multivariate analysis. Results: Efficacy data
indicated that the objective response rate (ORR) and disease control rate (DCR)
of the 129 patients with NSCLC who received anlotinib monotherapy was 9.3% (95%
CI: 4.9%-15.7%) and 78.3% (95%CI: 70.2%-85.1%), respectively. Additionally,
prognostic data suggested that the median progression-free survival (PFS) and
overall survival (OS) of the 129 patients with NSCLC were 4.1 months (95%CI:
2.84-5.36) and 10.1 months (95%CI: 8.58-11.62), respectively. Furthermore,
polymorphism analysis indicated that polymorphism of 4397T>C in
VEGFR2 was of clinical significance in the exploratory
analysis, which exhibited that the median PFS of patients with TC/CC and TT
genotype of 4397T>C polymorphism were 2.8 and 5.0 months, respectively
(P = .009). Additionally, patients with TT genotype
conferred a superior OS compared with those with TC/CC genotype (median OS: 11.5
vs 7.3 months, P = .016). Interestingly, mRNA expression of the
VEGFR2 gene suggested that mRNA expression of
VEGFR2 in PBMC specimens of patients with TC/CC genotype
was significantly higher than that of patients with TT genotype
(P < .001). Conclusion: Anlotinib
monotherapy exhibited potential efficacy for patients with treatment-refractory
advanced NSCLC. VEGFR2 polymorphism 4397T>C might be used as
a promising biomarker to predict the survival of patients with NSCLC who
received anlotinib administration.
Collapse
Affiliation(s)
- Xiaoyuan Li
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yang Cheng
- 66526Beijing Jishuitan Hospital, the Fourth Medical College of Peking University, Beijing, China
| | - Baorang Zhu
- The Fifth Medical Center, 104607Chinese PLA General Hospital, Beijing, China
| | - Ming Geng
- Beijing Chaoyang Integrative Medicine Emergency Medical Center, Beijing, China
| | - Peng Yan
- Center of Pulmonary and Critical Care Medicine, 104607Chinese PLA General Hospital, Beijing, China
| | - Mu Hu
- 26455Beijing Friendship Hospital of Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Uzun S, Korkmaz Y, Wuerdemann N, Arolt C, Puladi B, Siefer OG, Dönmez HG, Hufbauer M, Akgül B, Klussmann JP, Huebbers CU. Comprehensive Analysis of VEGFR2 Expression in HPV-Positive and -Negative OPSCC Reveals Differing VEGFR2 Expression Patterns. Cancers (Basel) 2021; 13:cancers13205221. [PMID: 34680369 PMCID: PMC8533978 DOI: 10.3390/cancers13205221] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 01/08/2023] Open
Abstract
VEGF signaling regulated by the vascular endothelial growth factor receptor 2 (VEGFR2) plays a decisive role in tumor angiogenesis, initiation and progression in several tumors including HNSCC. However, the impact of HPV-status on the expression of VEGFR2 in OPSCC has not yet been investigated, although HPV oncoproteins E6 and E7 induce VEGF-expression. In a series of 56 OPSCC with known HPV-status, VEGFR2 expression patterns were analyzed both in blood vessels from tumor-free and tumor-containing regions and within tumor cells by immunohistochemistry using densitometry. Differences in subcellular colocalization of VEGFR2 with endothelial, tumor and stem cell markers were determined by double-immunofluorescence imaging. Immunohistochemical results were correlated with clinicopathological data. HPV-infection induces significant downregulation of VEGFR2 in cancer cells compared to HPV-negative tumor cells (p = 0.012). However, with respect to blood vessel supply, the intensity of VEGFR2 staining differed only in HPV-positive OPSCC and was upregulated in the blood vessels of tumor-containing regions (p < 0.0001). These results may suggest different routes of VEGFR2 signaling depending on the HPV-status of the OPSCC. While in HPV-positive OPSCC, VEGFR2 might be associated with increased angiogenesis, in HPV-negative tumors, an autocrine loop might regulate tumor cell survival and invasion.
Collapse
Affiliation(s)
- Senem Uzun
- Jean-Uhrmacher-Institute for Otorhinolaryngological Research, University of Cologne, 50937 Cologne, Germany; (S.U.); (O.G.S.)
| | - Yüksel Korkmaz
- Department of Periodontology and Operative and Preventive Dentistry, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany;
| | - Nora Wuerdemann
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital of Cologne, 50937 Cologne, Germany;
- Centre for Molecular Medicine Cologne (CMMC), Faculty of Medicine, University of Cologne and University Hospital Cologne, 50931 Cologne, Germany
| | - Christoph Arolt
- Institute for Pathology, University Hospital of Cologne, 50937 Cologne, Germany;
| | - Behrus Puladi
- Department of Oral and Maxillofacial Surgery, University Hospital RWTH Aachen, 52074 Aachen, Germany;
| | - Oliver G. Siefer
- Jean-Uhrmacher-Institute for Otorhinolaryngological Research, University of Cologne, 50937 Cologne, Germany; (S.U.); (O.G.S.)
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital of Cologne, 50937 Cologne, Germany;
| | - Hanife G. Dönmez
- Institute of Virology, University of Cologne, Medical Faculty and University Hospital Cologne, 50935 Cologne, Germany; (H.G.D.); (M.H.); (B.A.)
- Department of Biology, Hacettepe University, Ankara 06800, Turkey
| | - Martin Hufbauer
- Institute of Virology, University of Cologne, Medical Faculty and University Hospital Cologne, 50935 Cologne, Germany; (H.G.D.); (M.H.); (B.A.)
| | - Baki Akgül
- Institute of Virology, University of Cologne, Medical Faculty and University Hospital Cologne, 50935 Cologne, Germany; (H.G.D.); (M.H.); (B.A.)
| | - Jens P. Klussmann
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital of Cologne, 50937 Cologne, Germany;
- Centre for Molecular Medicine Cologne (CMMC), Faculty of Medicine, University of Cologne and University Hospital Cologne, 50931 Cologne, Germany
- Correspondence: (J.P.K.); (C.U.H.)
| | - Christian U. Huebbers
- Jean-Uhrmacher-Institute for Otorhinolaryngological Research, University of Cologne, 50937 Cologne, Germany; (S.U.); (O.G.S.)
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital of Cologne, 50937 Cologne, Germany;
- Correspondence: (J.P.K.); (C.U.H.)
| |
Collapse
|
14
|
Yadollahi P, Jeon YK, Ng WL, Choi I. Current understanding of cancer-intrinsic PD-L1: regulation of expression and its protumoral activity. BMB Rep 2021. [PMID: 33298250 PMCID: PMC7851443 DOI: 10.5483/bmbrep.2021.54.1.241] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
In the last decade, we have witnessed an unprecedented clinical success in cancer immunotherapies targeting the programmed cell-death ligand 1 (PD-L1) and programmed cell-death 1 (PD-1) pathway. Besides the fact that PD-L1 plays a key role in immune regulation in tumor microenvironment, recently a plethora of reports has suggested a new perspective of non-immunological functions of PD-L1 in the regulation of cancer intrinsic activities including mesenchymal transition, glucose and lipid metabolism, stemness, and autophagy. Here we review the current understanding on the regulation of expression and intrinsic protumoral activity of cancer-intrinsic PD-L1.
Collapse
Affiliation(s)
- Pedram Yadollahi
- Innovative Therapeutic Research Institute, Inje University, Busan 47397; Department of Microbiology and Immunology, Inje University College of Medicine, Busan 47392, Korea
| | - You-Kyoung Jeon
- Innovative Therapeutic Research Institute, Inje University, Busan 47397; Department of Microbiology and Immunology, Inje University College of Medicine, Busan 47392, Korea
| | - Wooi Loon Ng
- Innovative Therapeutic Research Institute, Inje University, Busan 47397, Korea
| | - Inhak Choi
- Innovative Therapeutic Research Institute, Inje University, Busan 47397; Department of Microbiology and Immunology, Inje University College of Medicine, Busan 47392, Korea
| |
Collapse
|
15
|
Ni YH, Zhao X, Wang W. CD24, A Review of its Role in Tumor Diagnosis, Progression and Therapy. Curr Gene Ther 2021; 20:109-126. [PMID: 32576128 DOI: 10.2174/1566523220666200623170738] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/28/2020] [Accepted: 06/02/2020] [Indexed: 02/08/2023]
Abstract
CD24, is a mucin-like GPI-anchored molecules. By immunohistochemistry, it is widely detected in many solid tumors, such as breast cancers, genital system cancers, digestive system cancers, neural system cancers and so on. The functional roles of CD24 are either fulfilled by combination with ligands or participate in signal transduction, which mediate the initiation and progression of neoplasms. However, the character of CD24 remains to be intriguing because there are still opposite voices about the impact of CD24 on tumors. In preclinical studies, CD24 target therapies, including monoclonal antibodies, target silencing by RNA interference and immunotherapy, have shown us brighten futures on the anti-tumor application. Nevertheless, evidences based on clinical studies are urgently needed. Here, with expectancy to spark new ideas, we summarize the relevant studies about CD24 from a tumor perspective.
Collapse
Affiliation(s)
- Yang-Hong Ni
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041, Sichuan, China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, 610041, China
| | - Wei Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041, Sichuan, China
| |
Collapse
|
16
|
Jinesh GG, Napoli M, Smallin MT, Davis A, Ackerman HD, Raulji P, Montey N, Flores ER, Brohl AS. Mutant p53s and chromosome 19 microRNA cluster overexpression regulate cancer testis antigen expression and cellular transformation in hepatocellular carcinoma. Sci Rep 2021; 11:12673. [PMID: 34135394 PMCID: PMC8209049 DOI: 10.1038/s41598-021-91924-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/25/2021] [Indexed: 12/13/2022] Open
Abstract
A subset of hepatocellular carcinoma (HCC) overexpresses the chromosome 19 miRNA cluster (C19MC) and is associated with an undifferentiated phenotype marked by overexpression of cancer testis antigens (CTAs) including anti-apoptotic melanoma-A antigens (MAGEAs). However, the regulation of C19MC miRNA and MAGEA expression in HCCs are not understood. Here we show that, C19MC overexpression is tightly linked to a sub-set of HCCs with transcription-incompetent p53. Using next-generation and Sanger sequencing we found that, p53 in Hep3B cells is impaired by TP53-FXR2 fusion, and that overexpression of the C19MC miRNA-520G in Hep3B cells promotes the expression of MAGEA-3, 6 and 12 mRNAs. Furthermore, overexpression of p53-R175H and p53-R273H mutants promote miR-520G and MAGEA RNA expression and cellular transformation. Moreover, IFN-γ co-operates with miR-520G to promote MAGEA expression. On the other hand, metals such as nickel and zinc promote miR-526B but not miR-520G, to result in the suppression of MAGEA mRNA expression, and evoke cell death through mitochondrial membrane depolarization. Therefore our study demonstrates that a MAGEA-promoting network involving miR-520G, p53-defects and IFN-γ that govern cellular transformation and cell survival pathways, but MAGEA expression and survival are counteracted by nickel and zinc combination.
Collapse
Affiliation(s)
- Goodwin G Jinesh
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA. .,Sarcoma Department, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA.
| | - Marco Napoli
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA
| | - Marian T Smallin
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA.,Sarcoma Department, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA
| | - Andrew Davis
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA
| | - Hayley D Ackerman
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA
| | - Payal Raulji
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA
| | - Nicole Montey
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA
| | - Elsa R Flores
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA
| | - Andrew S Brohl
- Sarcoma Department, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA. .,Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA.
| |
Collapse
|
17
|
Geng N, Su J, Liu Z, Ding C, Xie S, Hu W. The Influence of KDR Genetic Variation on the Efficacy and Safety of Patients With Advanced NSCLC Receiving First-Line Bevacizumab Plus Chemotherapy Regimen. Technol Cancer Res Treat 2021; 20:15330338211019433. [PMID: 34060368 PMCID: PMC8173991 DOI: 10.1177/15330338211019433] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Objective: Angiogenesis plays an important role in the growth and metastasis of non-small cell lung cancer (NSCLC). Bevacizumab is a humanized monoclonal antibody that mainly acts on vascular endothelial growth factor A (VEGFA). Kinase insert domain receptor (KDR) is the most important target of VEGFA. The aim of present study was to investigate the influence of KDR genetic variation on the efficacy and safety of patients with advanced NSCLC receiving first-line bevacizumab plus chemotherapy regimen. Methods: A total of 169 patients with advanced NSCLC who received bevacizumab combined with chemotherapy were recruited in this study. Clinical outcome of the regimens was evaluated in the hospital. Peripheral blood and biopsy tissue specimens of patients were collected for the genotyping of KDR genetic variation and KDR mRNA expression, respectively. The association between KDR genotype status and other variables were analyzed. Univariate analysis of genotype status and prognosis was implemented using the Kaplan-Meier survival analysis method. Multivariate Cox regression analysis was performed to adjust the confounding factors. Results: Of the polymorphisms analyzed, only V297 L was of clinical significance. The prevalence of V297 L among the study population were as follows: CC genotype 123 cases (72.8%), CT genotype 41 cases (24.3%), TT genotype 5 cases (2.9%). The minimum allele frequency is 0.15. The distribution frequencies of the 3 genotypes corresponded with Hardy-Weinberg equilibrium (P = 0.489). Patients with TT and CT genotypes were merged in the subsequent comparison of clinical outcomes. The analysis of efficacy exhibited that the objective response rates (ORR) of patients with CC genotype and CT/TT genotypes were 52.8% and 47.8% (P = 0.561), respectively. Prognosis indicated that the median progression free survival (PFS) of patients with CC genotype and CT/TT genotype were 8.9 and 5.5 months, respectively (P = 0.006). The median OS of the 2 genotypes were 20.0 and 14.9 months, respectively (P = 0.021). Adjusted in multivariate Cox regression analysis of PFS, CT/TT genotypes were an independent factor for PFS [hazard ratio (HR) = 1.59, P = 0.011). Safety profile according to genotype status of V297 L failed to find significant difference. Interestingly, the expression of KDR mRNA of patients with CT/TT genotype was significantly higher than that of patients with CC genotype in the 58 cancer tissue specimens (P < 0.001). Conclusion: The clinical comes of patients with advanced NSCLC receiving first-line bevacizumab plus chemotherapy regimens might be impacted by polymorphism V297 L through mediating the mRNA expression of KDR.
Collapse
Affiliation(s)
- Nan Geng
- Department of Respiratory Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Jingwei Su
- Department of Third Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Zhikun Liu
- Department of Radiotherapy, East Hospital of The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Cuimin Ding
- Department of Respiratory Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Shaonan Xie
- Department of Second Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Wenxia Hu
- Department of Respiratory Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| |
Collapse
|
18
|
Tsuchiya H, Shiota G. Immune evasion by cancer stem cells. Regen Ther 2021; 17:20-33. [PMID: 33778133 PMCID: PMC7966825 DOI: 10.1016/j.reth.2021.02.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 02/10/2021] [Accepted: 02/21/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor immunity represents a new avenue for cancer therapy. Immune checkpoint inhibitors have successfully improved outcomes in several tumor types. In addition, currently, immune cell-based therapy is also attracting significant attention. However, the clinical efficacy of these treatments requires further improvement. The mechanisms through which cancer cells escape the immune response must be identified and clarified. Cancer stem cells (CSCs) play a central role in multiple aspects of malignant tumors. CSCs can initiate tumors in partially immunocompromised mice, whereas non-CSCs fail to form tumors, suggesting that tumor initiation is a definitive function of CSCs. However, the fact that non-CSCs also initiate tumors in more highly immunocompromised mice suggests that the immune evasion property may be a more fundamental feature of CSCs rather than a tumor-initiating property. In this review, we summarize studies that have elucidated how CSCs evade tumor immunity and create an immunosuppressive milieu with a focus on CSC-specific characteristics and functions. These profound mechanisms provide important clues for the development of novel tumor immunotherapies.
Collapse
Key Words
- ADCC, antibody-dependent cell mediated cytotoxicity
- ALDH, alcohol dehydrogenase
- AML, acute myeloid leukemia
- ARID3B, AT-rich interaction domain-containing protein 3B
- CCR7, C–C motif chemokine receptor 7
- CIK, cytokine-induced killer cell
- CMV, cytomegalovirus
- CSC, cancer stem cell
- CTL, cytotoxic T lymphocytes
- CTLA-4, cytotoxic T-cell-associated antigen-4
- Cancer stem cells
- DC, dendritic cell
- DNMT, DNA methyltransferase
- EMT, epithelial–mesenchymal transition
- ETO, fat mass and obesity associated protein
- EV, extracellular vesicle
- HNSCC, head and neck squamous cell carcinoma
- Immune checkpoints
- Immune evasion
- KDM4, lysine-specific demethylase 4C
- KIR, killer immunoglobulin-like receptor
- LAG3, lymphocyte activation gene 3
- LILR, leukocyte immunoglobulin-like receptor
- LMP, low molecular weight protein
- LOX, lysyl oxidase
- MDSC, myeloid-derived suppressor cell
- MHC, major histocompatibility complex
- MIC, MHC class I polypeptide-related sequence
- NGF, nerve growth factor
- NK cells
- NK, natural killer
- NOD, nonobese diabetic
- NSG, NOD/SCID IL-2 receptor gamma chain null
- OCT4, octamer-binding transcription factor 4
- PD-1, programmed death receptor-1
- PD-L1/2, ligands 1/2
- PI9, protease inhibitor 9
- PSME3, proteasome activator subunit 3
- SCID, severe combined immunodeficient
- SOX2, sex determining region Y-box 2
- T cells
- TAM, tumor-associated macrophage
- TAP, transporter associated with antigen processing
- TCR, T cell receptor
- Treg, regulatory T cell
- ULBP, UL16 binding protein
- uPAR, urokinase-type plasminogen activator receptor
Collapse
Affiliation(s)
- Hiroyuki Tsuchiya
- Division of Medical Genetics and Regenerative Medicine, Department of Genomic Medicine and Regenerative Therapy, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan
| | - Goshi Shiota
- Division of Medical Genetics and Regenerative Medicine, Department of Genomic Medicine and Regenerative Therapy, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan
| |
Collapse
|
19
|
Vathiotis IA, Gomatou G, Stravopodis DJ, Syrigos N. Programmed Death-Ligand 1 as a Regulator of Tumor Progression and Metastasis. Int J Mol Sci 2021; 22:ijms22105383. [PMID: 34065396 PMCID: PMC8160779 DOI: 10.3390/ijms22105383] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/18/2021] [Accepted: 05/18/2021] [Indexed: 12/21/2022] Open
Abstract
Programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) immune checkpoint has long been implicated in modeling antitumor immunity; PD-1/PD-L1 axis inhibitors exert their antitumor effects by relieving PD-L1-mediated suppression on tumor-infiltrating T lymphocytes. However, recent studies have unveiled a distinct, tumor-intrinsic, potential role for PD-L1. In this review, we focus on tumor-intrinsic PD-L1 signaling and delve into preclinical evidence linking PD-L1 protein expression with features of epithelial-to-mesenchymal transition program, cancer stemness and known oncogenic pathways. We further summarize data from studies supporting the prognostic significance of PD-L1 in different tumor types. We show that PD-L1 may indeed have oncogenic potential and act as a regulator of tumor progression and metastasis.
Collapse
Affiliation(s)
- Ioannis A. Vathiotis
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, 15772 Athens, Greece; (G.G.); (N.S.)
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
- Correspondence: ; Tel.: +30-69-4882-2683
| | - Georgia Gomatou
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, 15772 Athens, Greece; (G.G.); (N.S.)
| | - Dimitrios J. Stravopodis
- Department of Biology, School of Medicine, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Nikolaos Syrigos
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, 15772 Athens, Greece; (G.G.); (N.S.)
| |
Collapse
|
20
|
Sustained oxidative stress instigates differentiation of cancer stem cells into tumor endothelial cells: Pentose phosphate pathway, reactive oxygen species and autophagy crosstalk. Biomed Pharmacother 2021; 139:111643. [PMID: 33945913 DOI: 10.1016/j.biopha.2021.111643] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/08/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor angiogenesis plays a vital role in tumor growth and metastasis. It is proven that in tumor vasculature, endothelial cells (ECs) originate from a small population of cancer cells introduced as cancer stem cells (CSCs). Autophagy has a vital role in ECs differentiation from CSCs and tumor angiogenesis. High levels of reactive oxygen species (ROS) increased autophagy by inhibition of glucose-6-phosphate dehydrogenase (G6PD) and inactivation of the pentose phosphate pathway (PPP). Previously, we suggested that cancer cells initially increase the glycolysis rate when encountering ROS, then the metabolic balance is changed from glycolysis to PPP, following the continuation of oxidative stress. In this study, we investigate the possible role of persistent oxidative stress in the differentiation of CSCs into tumor ECs by relying on the relationship between the ROS, PPP and autophagy. Because tumor angiogenesis plays an important role in the growth and development of cancer, understanding the mechanisms involved in differentiating ECs from CSCs can help find promising treatments for cancer.
Collapse
|
21
|
Godwin I, Anto NP, Bava SV, Babu MS, Jinesh GG. Targeting K-Ras and apoptosis-driven cellular transformation in cancer. Cell Death Discov 2021; 7:80. [PMID: 33854056 PMCID: PMC8047025 DOI: 10.1038/s41420-021-00457-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/23/2021] [Accepted: 03/21/2021] [Indexed: 02/07/2023] Open
Abstract
Cellular transformation is a major event that helps cells to evade apoptosis, genomic instability checkpoints, and immune surveillance to initiate tumorigenesis and to promote progression by cancer stem cell expansion. However, the key molecular players that govern cellular transformation and ways to target cellular transformation for therapy are poorly understood to date. Here we draw key evidences from the literature on K-Ras-driven cellular transformation in the context of apoptosis to shed light on the key players that are required for cellular transformation and explain how aiming p53 could be useful to target cellular transformation. The defects in key apoptosis regulators such as p53, Bax, and Bak lead to apoptosis evasion, cellular transformation, and genomic instability to further lead to stemness, tumorigenesis, and metastasis via c-Myc-dependent transcription. Therefore enabling key apoptotic checkpoints in combination with K-Ras inhibitors will be a promising therapeutic target in cancer therapy.
Collapse
Affiliation(s)
- Isha Godwin
- Saveetha Medical College, Thandalam, Chennai, Tamil Nadu, 602105, India.
| | - Nikhil Ponnoor Anto
- Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Smitha V Bava
- Department of Biotechnology, University of Calicut, Malappuram, Kerala, 673635, India
| | - Mani Shankar Babu
- Department of Botany, University College, Thiruvananthapuram, Kerala, 695 034, India
| | - Goodwin G Jinesh
- Departments of Molecular Oncology, and Sarcoma, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
22
|
Bai M, Li ZG, Ba Y. Influence of KDR Genetic Variation on the Efficacy and Safety of Patients with Chemotherapy Refractory Metastatic CRC Who Received Apatinib Treatment. Int J Gen Med 2021; 14:1041-1055. [PMID: 33790633 PMCID: PMC8006973 DOI: 10.2147/ijgm.s300968] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/22/2021] [Indexed: 12/20/2022] Open
Abstract
Background The aim of the present study was to investigate the influence of kinase insert domain containing receptor (KDR) genetic variation on the efficacy of treatment and safety of patients with chemotherapy-refractory metastatic colorectal cancer (CRC) receiving apatinib. Methods A total of 108 patients with chemotherapy refractory metastatic CRC who were treated with apatinib participated in this study retrospectively. Efficacy of the patients' treatment was evaluated. Prognosis was carried out and safety profile was documented, respectively. Blood specimens and peripheral blood mononuclear cells (PBMC) of the patients were obtained for the analysis of genetic variation and KDR gene mRNA expression, respectively. The association between genotype status and clinical outcomes was presented. Results Objective response rate (ORR) and disease control rate (DCR) of the 108 patients with metastatic CRC receiving apatinib treatment were 5.6% and 69.4%, respectively. Survival analysis results exhibited that the median progression-free survival (PFS) and overall survival (OS) of the 108 patients with metastatic CRC was 3.6 months (95% confidence interval (CI): 3.03-4.17 months) and 8.9 months (95% CI: 7.57-10.23 months), respectively. Subsequently, the analysis of KDR genetic variation indicated that rs2071559 was of clinical significance. The minor allele frequency of rs2071559 was 0.22 and the genotype status corresponded with Hardy-Weinberg equilibrium (P=0.949). Prognosis analysis in a dominant inheritance manner through the combination of patients with TC and CC genotype showed that the median PFS of patients with TT genotype and TC/CC genotype was 4.1 and 3.0 months, respectively (P=0.012). Furthermore, the median OS of patients with the two genotypes was 10.5 and 6.1 months, respectively (P=0.007). Additionally, multivariate Cox regression analysis of OS showed that TC/CC genotype was an independent factor for OS (Hazard ratio (HR)=0.65, P=0.021). Interestingly, mRNA expression analysis suggested that the mRNA expression of KDR in PBMC differed significantly according to rs2071559 genotype status (P<0.001). Conclusion Apatinib demonstrated a potentially superior clinical outcome for patients with chemotherapy-refractory metastatic CRC. KDR polymorphism rs2071559 could be used as a potential biomarker for the prognosis evaluation of patients with CRC receiving apatinib therapy.
Collapse
Affiliation(s)
- Ming Bai
- Department of Gastrointestinal Oncology, Affiliated Tumor Hospital of Tianjin Medical University, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, People's Republic of China
| | - Zhi-Guo Li
- Department of Minimally Invasive Digestive Surgery, Shanxi Cancer Hospital, Taiyuan, People's Republic of China
| | - Yi Ba
- Department of Gastrointestinal Oncology, Affiliated Tumor Hospital of Tianjin Medical University, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, People's Republic of China
| |
Collapse
|
23
|
Asha K, Sharma-Walia N. Targeting Host Cellular Factors as a Strategy of Therapeutic Intervention for Herpesvirus Infections. Front Cell Infect Microbiol 2021; 11:603309. [PMID: 33816328 PMCID: PMC8017445 DOI: 10.3389/fcimb.2021.603309] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 01/12/2021] [Indexed: 12/13/2022] Open
Abstract
Herpesviruses utilize various host factors to establish latent infection, survival, and spread disease in the host. These factors include host cellular machinery, host proteins, gene expression, multiple transcription factors, cellular signal pathways, immune cell activation, transcription factors, cytokines, angiogenesis, invasion, and factors promoting metastasis. The knowledge and understanding of host genes, protein products, and biochemical pathways lead to discovering safe and effective antivirals to prevent viral reactivation and spread infection. Here, we focus on the contribution of pro-inflammatory, anti-inflammatory, and resolution lipid metabolites of the arachidonic acid (AA) pathway in the lifecycle of herpesvirus infections. We discuss how various herpesviruses utilize these lipid pathways to their advantage and how we target them to combat herpesvirus infection. We also summarize recent development in anti-herpesvirus therapeutics and new strategies proposed or under clinical trials. These anti-herpesvirus therapeutics include inhibitors blocking viral life cycle events, engineered anticancer agents, epigenome influencing factors, immunomodulators, and therapeutic compounds from natural extracts.
Collapse
Affiliation(s)
| | - Neelam Sharma-Walia
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| |
Collapse
|
24
|
Grillo E, Corsini M, Ravelli C, Zammataro L, Bacci M, Morandi A, Monti E, Presta M, Mitola S. Expression of activated VEGFR2 by R1051Q mutation alters the energy metabolism of Sk-Mel-31 melanoma cells by increasing glutamine dependence. Cancer Lett 2021; 507:80-88. [PMID: 33744390 DOI: 10.1016/j.canlet.2021.03.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/03/2021] [Accepted: 03/05/2021] [Indexed: 12/12/2022]
Abstract
Vascular endothelial growth factor receptor 2 (VEGFR2) activating mutations are emerging as important oncogenic driver events. Understanding the biological implications of such mutations may help to pinpoint novel therapeutic targets. Here we show that activated VEGFR2 via the pro-oncogenic R1051Q mutation induces relevant metabolic changes in melanoma cells. The expression of VEGFR2R1051Q leads to higher energy metabolism and ATP production compared to control cells expressing VEGFR2WT. Furthermore, activated VEGFR2R1051Q augments the dependence on glutamine (Gln) of melanoma cells, thus increasing Gln uptake and their sensitivity to Gln deprivation and to inhibitors of glutaminase, the enzyme initiating Gln metabolism by cells. Overall, these results highlight Gln addiction as a metabolic vulnerability of tumors harboring the activating VEGFR2R1051Q mutation and suggest novel therapeutic approaches for those patients harboring activating mutations of VEGFR2.
Collapse
Affiliation(s)
- Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy.
| | - Michela Corsini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy
| | - Luca Zammataro
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Marina Bacci
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, 50134, Italy
| | - Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, 50134, Italy
| | - Eugenio Monti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy
| | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy.
| |
Collapse
|
25
|
Geng N, Ding CM, Liu ZK, Song S, Hu WX. Influence of VEGFR2 gene polymorphism on the clinical outcomes of apatinib for patients with chemotherapy-refractory extensive-stage SCLC: a real-world retrospective study. Int J Clin Oncol 2021; 26:670-683. [PMID: 33392882 DOI: 10.1007/s10147-020-01849-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/25/2020] [Indexed: 01/07/2023]
Abstract
PURPOSE Great individual differences were observed regarding the efficacy of apatinib clinically. The aim of present study was to investigate the influence of vascular endothelial growth factor receptor2 (VEGFR2) gene polymorphism on the clinical outcomes of apatinib for patients with chemotherapy-refractory extensive-stage small cell lung cancer (ES-SCLC). METHODS A total of 128 patients with chemotherapy-refractory ES-SCLC who were treated with apatinib at an initial dosage of 250 or 500 mg were included in this study. The change of target lesions was assessed. Overall response rate (ORR) was evaluated. Prognosis was carried out and safety profile was documented. Additionally, peripheral blood and biopsy cancer tissue specimens of the patients with SCLC were collected for the analysis of polymorphism and VEGFR2 gene mRNA expression, respectively. The association between genotype status and baseline characteristics was performed. Univariate analysis of genotype status and prognosis was carried out using Kaplan-Meier survival analysis and multivariate analysis were adjusted by Cox regression analysis. RESULTS Efficacy of apatinib included partial response (PR) in 15 patients, stable disease (SD) in 86 patients, progressive disease (PD) in 27 patients. Therefore, ORR of the 128 patients with ES-SCLC was 11.7%, and disease control rate (DCR) was 78.9%. Prognosis suggested that the median progression-free survival (PFS) and overall survival (OS) of the 128 patients with ES-SCLC was 4.2 months and 8.2 months, respectively. The polymorphism analysis focusing on VEGFR2 gene indicated that one single nucleotide polymorphism 889C>T was of clinical significance. Prevalence of 889C>T among the 128 patients with SCLC were as follows: CC genotype 87 cases (68.0%), CT genotype 38 cases (29.7%) and TT genotype 3 cases (2.3%), the minor allele frequency of 889C>T was 0.17, which was in accordance with Hardy-Weinberg Equilibrium (P = 0.628). Patients with CT and TT genotypes were merged in the subsequent analysis. Prognosis analysis exhibited that the median PFS of patients with CT/TT genotype and CC genotype was 3.3 and 5.0 months, respectively (P = 0.02). Furthermore, the median OS of patients was 5.5 and 9.0 months, respectively (P = 0.008). Additionally, multivariate Cox regression analysis of OS demonstrated that CT/TT genotype was an independent factor for OS [Hazard ratio (HR) = 0.64, P = 0.019]. However, the safety profile according to genotype status of 889C>T failed to show significant difference. Interestingly, mRNA expression analysis suggested that the mRNA expression of VEGFR2 in cancer tissues were significantly different according to CC and CT/TT genotypes (P < 0.001). CONCLUSION The administration with apatinib for patients with chemotherapy-refractory ES-SCLC was of potential clinical significance. The clinical outcomes of patients with ES-SCLC who were treated with apatinib could be impacted by VEGFR2 889C>T polymorphism through mediating the VEGFR2 mRNA expression.
Collapse
Affiliation(s)
- Nan Geng
- Department of Respiratory Medicine, The Fourth Hospital of Hebei Medical University, 12 Jian-Kang Road, Shijiazhuang, 050012, People's Republic of China
| | - Cui-Min Ding
- Department of Respiratory Medicine, The Fourth Hospital of Hebei Medical University, 12 Jian-Kang Road, Shijiazhuang, 050012, People's Republic of China
| | - Zhi-Kun Liu
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050012, People's Republic of China
| | - Shan Song
- Department of Respiratory Medicine, The Fourth Hospital of Hebei Medical University, 12 Jian-Kang Road, Shijiazhuang, 050012, People's Republic of China
| | - Wen-Xia Hu
- Department of Respiratory Medicine, The Fourth Hospital of Hebei Medical University, 12 Jian-Kang Road, Shijiazhuang, 050012, People's Republic of China.
| |
Collapse
|
26
|
Regulation of MYO18B mRNA by a network of C19MC miRNA-520G, IFN-γ, CEBPB, p53 and bFGF in hepatocellular carcinoma. Sci Rep 2020; 10:12371. [PMID: 32704163 PMCID: PMC7378193 DOI: 10.1038/s41598-020-69179-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023] Open
Abstract
MYO18B has been proposed to contribute to the progression of hepatocellular carcinoma (HCC). However, the signals that govern MYO18B transcription are not known. Here we show that, a network of C19MC miRNA-520G, IFN-γ, CEBPB and p53 transcriptional-defects promote MYO18B mRNA expression in HCCs. IFN-γ by itself suppresses MYO18B transcription, but promotes it when miRNA-520G is stably overexpressed. Similarly, CEBPB-liver-enriched activator protein (LAP) isoform overexpression suppresses MYO18B transcription but promotes transcription when the cells are treated with IFN-γ. Furthermore, miR-520G together with mutant-p53 promotes MYO18B transcription. Conversely, bFGF suppresses MYO18B mRNA irrespective of CEBPB, miR-520G overexpression or IFN-γ treatment. Finally high MYO18B expression reflects poor prognosis while high MYL5 or MYO1B expression reflects better survival of HCC patients. Thus, we identified a network of positive and negative regulators of MYO18B mRNA expression which reflects the survival of HCC patients.
Collapse
|
27
|
Lv J, Guo T, Qu X, Che X, Li C, Wang S, Gong J, Wu P, Liu Y, Liu Y, Xu L. PD-L1 Under Regulation of miR-429 Influences the Sensitivity of Gastric Cancer Cells to TRAIL by Binding of EGFR. Front Oncol 2020; 10:1067. [PMID: 32775300 PMCID: PMC7387728 DOI: 10.3389/fonc.2020.01067] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 05/28/2020] [Indexed: 01/16/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has received extensive attention as a cancer therapeutic due to its high propensity for tumor targeting with minimal toxicity to healthy tissue. Gastric cancer (GCa) cells show high levels of TRAIL resistance. Epidermal growth factor receptor (EGFR) antagonizes TRAIL-induced apoptosis, but the mechanisms of these effects remain unclear. Our past research confirmed TRAIL-resistant (BGC823 and SGC7901) and TRAIL-sensitive cells (HGC27 and MKN45). miR-429 associated with TRAIL sensitivity was screened using microRNA arrays. The transfection of mimics and inhibitors confirmed that miR-429 negatively correlated with GCa TRAIL resistance. The target gene of miR-429 was identified as PD-L1, which positively correlated with TRAIL resistance through gene silencing and recovery experiments. Using co-immunoprecipitation (co-IP) and proximity ligation assay, we demonstrated that the pro-survival effects of PD-L1 are mediated through the binding and activation of EGFR. Cell viability experiments demonstrated that PD-L1 is key to the maintenance of cell viability in TRAIL-treated cells. This indicated that PD-L1 binds to and participates in EGFR activation through miR-429 regulation to antagonize TRAIL-induced apoptosis. This provides a new theoretical basis for the combination of the EGFR monoclonal antibodies including cetuximab, PD-L1 inhibitors, and human recombinant TRAIL in gastric cancer therapy and can filter patients who are currently sensitive to TRAIL treatment.
Collapse
Affiliation(s)
- Jinqi Lv
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Tianshu Guo
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Xiaofang Che
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Ce Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Shuo Wang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Jing Gong
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Peihong Wu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Yang Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Ling Xu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| |
Collapse
|
28
|
Yan Z, Gu YY, Hu XD, Zhao Q, Kang HL, Wang M, Duan W, Guan Y. Clinical outcomes and safety of apatinib monotherapy in the treatment of patients with advanced epithelial ovarian carcinoma who progressed after standard regimens and the analysis of the VEGFR2 polymorphism. Oncol Lett 2020; 20:3035-3045. [PMID: 32782621 DOI: 10.3892/ol.2020.11857] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 06/05/2020] [Indexed: 12/30/2022] Open
Abstract
The aims of the present study were to investigate the clinical outcomes and safety of apatinib monotherapy in the treatment of patients with advanced epithelial ovarian carcinoma (EOC) who have progressed after standard regimens, and to analyze the vascular endothelial growth factor receptor 2 (VEGFR2) rs2071559 polymorphism. A total of 118 patients with advanced EOC who received apatinib treatment were included in the study. Tumor response was evaluated using progression-free survival (PFS) and overall survival (OS) time, and safety data were documented. Additionally, peripheral blood and peripheral blood mononuclear cell (PBMC) specimens from the patients with EOC were collected to perform the genotyping of genetic polymorphism and assess the mRNA expression of VEGFR2, respectively. The objective response rate across the 118 patients with advanced EOC was 38.98%, the disease control rate was 63.56%, the median PFS time was 4.65 months and the median OS time was 15.10 months. Regarding the polymorphism analysis, the prevalence of rs2071559 in VEGFR2 among the 118 patients with advanced EOC was recorded as the TT genotype in 72 cases (61.02%), TC genotype in 41 cases (34.75%) and CC genotype in 5 cases (4.23%), and the minor allele frequency of rs2071559 was 0.22. The distribution of the three genotypes was in accordance with the Hardy-Weinberg equilibrium (P=0.781). TC and CC genotypes were merged in the subsequent analysis. The prognosis analyses suggested that the median PFS time of patients with the TC/CC genotype and the TT genotype was 3.10 and 5.40 months, respectively (P=0.015). Moreover, the median OS time of the two genotypes was 12.60 and 17.50 months, respectively (P=0.009). However, no association was noted between genotype status of the polymorphism and adverse reactions. Additionally, the mRNA expression analysis indicated that the mRNA expression levels of VEGFR2 in PBMC specimens were significantly different between TT and TC/CC genotypes (P<0.001). The present study suggested that the clinical outcomes of patients with advanced EOC, who progressed after standard regimens and received apatinib treatment, might be influenced by the VEGFR2 rs2071559 polymorphism.
Collapse
Affiliation(s)
- Zhen Yan
- Department of Gynecological Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, P.R. China
| | - Yuan-Yuan Gu
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Xiao-Di Hu
- Department of Gynecological Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, P.R. China
| | - Qun Zhao
- Department of Gynecological Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, P.R. China
| | - Hai-Li Kang
- Department of Gynecological Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, P.R. China
| | - Miao Wang
- Department of Gynecological Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, P.R. China
| | - Wei Duan
- Department of Gynecological Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, P.R. China
| | - Yin Guan
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| |
Collapse
|
29
|
PD-L1 promotes tumor growth and progression by activating WIP and β-catenin signaling pathways and predicts poor prognosis in lung cancer. Cell Death Dis 2020; 11:506. [PMID: 32632098 PMCID: PMC7338457 DOI: 10.1038/s41419-020-2701-z] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/10/2020] [Accepted: 06/14/2020] [Indexed: 12/13/2022]
Abstract
PD-L1 is overexpressed in tumor cells and contributes to cancer immunoevasion. However, the role of the tumor cell-intrinsic PD-L1 in cancers remains unknown. Here we show that PD-L1 regulates lung cancer growth and progression by targeting the WIP and β-catenin signaling. Overexpression of PD-L1 promotes tumor cell growth, migration and invasion in lung cancer cells, whereas PD-L1 knockdown has the opposite effects. We have also identified WIP as a new downstream target of PD-L1 in lung cancer. PD-L1 positively modulates the expression of WIP. Knockdown of WIP also inhibits cell viability and colony formation, whereas PD-L1 overexpression can reverse this inhibition effects. In addition, PD-L1 can upregulate β-catenin by inhibiting its degradation through PI3K/Akt signaling pathway. Moreover, we show that in lung cancer cells β-catenin can bind to the WIP promoter and activate its transcription, which can be promoted by PD-L1 overexpression. The in vivo experiments in a human lung cancer mouse model have also confirmed the PD-L1-mediated promotion of tumor growth and progression through activating the WIP and β-catenin pathways. Furthermore, we demonstrate that PD-L1 expression is positively correlated with WIP in tumor tissues of human adenocarcinoma patients and the high expression of PD-L1 and WIP predicts poor prognosis. Collectively, our results provide new insights into understanding the pro-tumorigenic role of PD-L1 and its regulatory mechanism on WIP in lung cancer, and suggest that the PD-L1/Akt/β-catenin/WIP signaling axis may be a potential therapeutic target for lung cancers.
Collapse
|
30
|
Perry JM, Tao F, Roy A, Lin T, He XC, Chen S, Lu X, Nemechek J, Ruan L, Yu X, Dukes D, Moran A, Pace J, Schroeder K, Zhao M, Venkatraman A, Qian P, Li Z, Hembree M, Paulson A, He Z, Xu D, Tran TH, Deshmukh P, Nguyen CT, Kasi RM, Ryan R, Broward M, Ding S, Guest E, August K, Gamis AS, Godwin A, Sittampalam GS, Weir SJ, Li L. Overcoming Wnt-β-catenin dependent anticancer therapy resistance in leukaemia stem cells. Nat Cell Biol 2020; 22:689-700. [PMID: 32313104 PMCID: PMC8010717 DOI: 10.1038/s41556-020-0507-y] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 03/12/2020] [Indexed: 02/07/2023]
Abstract
Leukaemia stem cells (LSCs) underlie cancer therapy resistance but targeting these cells remains difficult. The Wnt-β-catenin and PI3K-Akt pathways cooperate to promote tumorigenesis and resistance to therapy. In a mouse model in which both pathways are activated in stem and progenitor cells, LSCs expanded under chemotherapy-induced stress. Since Akt can activate β-catenin, inhibiting this interaction might target therapy-resistant LSCs. High-throughput screening identified doxorubicin (DXR) as an inhibitor of the Akt-β-catenin interaction at low doses. Here we repurposed DXR as a targeted inhibitor rather than a broadly cytotoxic chemotherapy. Targeted DXR reduced Akt-activated β-catenin levels in chemoresistant LSCs and reduced LSC tumorigenic activity. Mechanistically, β-catenin binds multiple immune-checkpoint gene loci, and targeted DXR treatment inhibited expression of multiple immune checkpoints specifically in LSCs, including PD-L1, TIM3 and CD24. Overall, LSCs exhibit distinct properties of immune resistance that are reduced by inhibiting Akt-activated β-catenin. These findings suggest a strategy for overcoming cancer therapy resistance and immune escape.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/pharmacology
- Apoptosis
- Cell Proliferation
- Doxorubicin/pharmacology
- Drug Resistance, Neoplasm
- Female
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Male
- Mice
- Mice, Knockout
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- PTEN Phosphohydrolase/physiology
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Tumor Cells, Cultured
- Wnt Proteins/physiology
- Xenograft Model Antitumor Assays
- beta Catenin/physiology
Collapse
Affiliation(s)
- John M Perry
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Children's Mercy Kansas City, Kansas City, MO, USA
- University of Kansas Medical Center, Kansas City, KS, USA
- University of Missouri Kansas City School of Medicine, Kansas City, MO, USA
| | - Fang Tao
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Children's Mercy Kansas City, Kansas City, MO, USA
| | - Anuradha Roy
- High Throughput Screening Laboratory, University of Kansas, Lawrence, KS, USA
| | - Tara Lin
- University of Kansas Medical Center, Kansas City, KS, USA
| | - Xi C He
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Shiyuan Chen
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Xiuling Lu
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | | | - Linhao Ruan
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Center for Cell Dynamics, Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Xiazhen Yu
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Debra Dukes
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Andrea Moran
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | | | - Meng Zhao
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Key Laboratory of Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
| | | | - Pengxu Qian
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Center of Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Zhenrui Li
- Stowers Institute for Medical Research, Kansas City, MO, USA
- St. Jude, Memphis, TN, USA
| | - Mark Hembree
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Ariel Paulson
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Zhiquan He
- Department of Electrical Engineering and Computer Science and C.S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Dong Xu
- Department of Electrical Engineering and Computer Science and C.S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Thanh-Huyen Tran
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, US
| | - Prashant Deshmukh
- Polymer Program, Institute of Materials Science, University of Connecticut, Storrs, CT, USA
| | - Chi Thanh Nguyen
- Department of Chemistry, University of Connecticut, Storrs, CT, USA
| | - Rajeswari M Kasi
- Polymer Program, Institute of Materials Science, University of Connecticut, Storrs, CT, USA
- Department of Chemistry, University of Connecticut, Storrs, CT, USA
| | - Robin Ryan
- Children's Mercy Kansas City, Kansas City, MO, USA
| | | | - Sheng Ding
- School of Pharmaceutical Science, Tsinghua University, Beijing, China
| | - Erin Guest
- Children's Mercy Kansas City, Kansas City, MO, USA
| | - Keith August
- Children's Mercy Kansas City, Kansas City, MO, USA
| | - Alan S Gamis
- Children's Mercy Kansas City, Kansas City, MO, USA
| | - Andrew Godwin
- University of Kansas Medical Center, Kansas City, KS, USA
| | - G Sitta Sittampalam
- University of Kansas Medical Center, Kansas City, KS, USA
- Therapeutics for Rare and Neglected Diseases, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Scott J Weir
- Department of Cancer Biology, The Institute for Advancing Medical Innovation and University of Kansas Cancer Center, Kansas City, Kansas, USA
| | - Linheng Li
- Stowers Institute for Medical Research, Kansas City, MO, USA.
- Department of Pathology and Laboratory Medicine and Division of Medical Oncology, Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
31
|
Liu JY, Zhu BR, Wang YD, Sun X. The efficacy and safety of Apatinib mesylate in the treatment of metastatic osteosarcoma patients who progressed after standard therapy and the VEGFR2 gene polymorphism analysis. Int J Clin Oncol 2020; 25:1195-1205. [PMID: 32215805 DOI: 10.1007/s10147-020-01644-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 02/28/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the efficacy and safety of Apatinib mesylate in the treatment of metastatic osteosarcoma patients who progressed after standard therapy and the VEGFR2 gene polymorphism analysis. METHODS Designed as a retrospective study, a total of 105 metastatic osteosarcoma patients who progressed after standard therapy were included in this study. The metastatic osteosarcoma patients received 500-750 mg Apatinib mesylate according to body surface area until disease progression or unacceptable toxicity with 28 days one cycle. Overall response was evaluated after two cycles Apatinib treatment, then progression-free survival (PFS) and overall survival (OS) were evaluated, and safety data were recorded. Additionally. peripheral blood and peripheral blood mononuclear cell (PBMC) specimens in the osteosarcoma patients were collected for the genotyping of VEGFR2 genetic variation and mRNA expression, respectively. Analysis on the association between genotype and baseline characteristics and VEGFR2 gene mRNA expression was analyzed. The univariate analysis of genotypes and prognosis was carried out by Kaplan-Meier survival analysis, and multivariate analysis was adjusted by Cox regression analysis. RESULTS The objective response rate (ORR) of the 105 metastatic osteosarcoma patients was 37.14%, disease control rate (DCR) was 77.14%, median PFS was 4.1 months, and median OS was 9.0 months. Regarding the VEGFR2 gene polymorphisms analysis, only - 906 T > C was of clinical significance. The prevalence of - 906 T > C in VEGFR2 among the study population was as follows: TT genotype 62 cases (59.05%), TC genotype 36 cases (34.29%) and CC genotype 7 cases (6.66%), minor allele frequency of - 906 T > C was 0.24. Compared with patients with TC/CC genotype, patients with TT genotype showed longer median PFS (5.0 versus 3.1 months, P = 0.011) and median OS (9.8 versus 7.6 months, P = 0.032). There was no correlation between the polymorphism and adverse reactions. Additionally, the mRNA expression in 69 randomly selected sample indicated that the mRNA expression of VEGFR2 of the patients with CC/TC genotypes were significantly higher than those of the TT genotype patients (P < 0.001). CONCLUSION Apatinib was safe and effective in the treatment of metastatic osteosarcoma patients who progressed after standard therapy. The clinical outcomes of Apatinib may be influenced by the polymorphism - 906 T > C of VEGFR2 through mediating the mRNA expression of VEGFR2.
Collapse
Affiliation(s)
- Jia-Yong Liu
- Department of Bone and Soft Tissue Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, P. R. China.
| | - Bao-Rang Zhu
- Department of Tumor Minimally Invasive Treatment, The Fifth Medical Centre, Chinese PLA General Hospital (Former 307th Hospital of the PLA), Beijing, 100071, P. R. China
| | - Yu-Dong Wang
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, P. R. China
| | - Xin Sun
- Department of Orthopedic Oncology, Peking University People's Hospital, Beijing, 100044, P. R. China.
| |
Collapse
|
32
|
Song ZZ, Zhao LF, Zuo J, Fan ZS, Wang L, Wang YD. Clinical Outcomes and Safety of Apatinib Mesylate in the Treatment of Advanced Non-Squamous Non-Small Cell Lung Cancer in Patients Who Progressed After Standard Therapy and Analysis of the KDR Gene Polymorphism. Onco Targets Ther 2020; 13:603-613. [PMID: 32021302 PMCID: PMC6982468 DOI: 10.2147/ott.s222985] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 12/17/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose This study investigated the clinical outcomes and safety of apatinib mesylate in the treatment of advanced non-squamous non-small cell lung cancer (NSCLC) in patients who progressed after standard therapy, and analyzed the kinase insert domain receptor (KDR) gene polymorphism. Methods A total of 135 patients with advanced non-squamous NSCLC who received apatinib mesylate were included. Objective response rates were evaluated. Subsequently, progression-free survival (PFS) and overall survival (OS) were assessed and safety data were recorded. Additionally, peripheral blood and biopsy cancer tissue specimens were collected from the patients with NSCLC for the genotyping of the genetic polymorphism and mRNA expression of the KDR gene, respectively. Analysis on the association between genotypes and prognosis was conducted. Results The objective response rate of the 135 patients with NSCLC was 18.52%, disease control rate was 65.19%, median PFS was 3.95 months, and median OS was 10.05 months. Regarding the KDR gene polymorphism analysis, the distribution of the 4397T>C polymorphism genotypes was in accordance with the Hardy–Weinberg Equilibrium (P=0.868). Moreover, the prognosis analysis indicated that the median PFS of patients with the CC/TC and TT genotypes was 2.80 and 4.80 months, respectively (P=0.002). Furthermore, the median OS of patients with the two genotypes was 9.10 and 10.56 months, respectively (P=0.041). The multivariate Cox regression analysis showed that the TC/CC genotypes were an independent factor for PFS (odds ratio: 1.72, P=0.009). There was no correlation between the polymorphism and adverse reactions. Additionally, the mRNA expression analysis suggested that the mRNA levels of KDR in cancer tissues were significantly different between the TT and TC/CC genotypes (P<0.001). Conclusion The clinical outcomes of treatment with apatinib mesylate for advanced non-squamous NSCLC in patients who progressed after standard therapy may be influenced by the KDR 4397T>C polymorphism through mediation of the mRNA expression of KDR.
Collapse
Affiliation(s)
- Zi-Zheng Song
- Department of Medical Oncology, The Affiliated Hospital of Hebei University, Baoding, People's Republic of China
| | - Li-Fen Zhao
- Department of Respiratory and Critical Care Medicine, The Shanxi Dayi Hospital, Taiyuan, People's Republic of China
| | - Jing Zuo
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Zhi-Song Fan
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Long Wang
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Yu-Dong Wang
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| |
Collapse
|
33
|
Dormant tumor cells interact with memory CD8 + T cells in RET transgenic mouse melanoma model. Cancer Lett 2020; 474:74-81. [PMID: 31962142 DOI: 10.1016/j.canlet.2020.01.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/21/2019] [Accepted: 01/15/2020] [Indexed: 12/18/2022]
Abstract
Melanoma is an aggressive form of skin-cancer. Melanoma cells are characterized by their plasticity, resulting in therapy resistance. Using RET transgenic mouse melanoma model, we characterized dormant tumor cells accumulated in the bone marrow (BM) and investigated their interaction with effector memory CD8+ T cells. We found that cells expressing melanoma-associated antigen tyrosinase related protein (TRP)-2 and stemness marker CD133 represented less than 1.5% of all melanoma cells in primary skin lesions and metastatic lymph nodes. The majority of these cells were negative for the proliferation marker Ki67. In the BM, CD133+TRP-2+ melanoma cells displayed an aberrant expression of p16, p27, Ki67 and PCNA proteins, suggesting their dormant phenotype. Moreover, these cells were characterized by an elevated expression of various molecules characterized stemness, metastatic, angiogenic and immunosuppressive properties such as CD271, CD34, HIF-1α, CXCR3, CXCR4, VEGR2, PD-L1, CTLA-4, CD39 and CCR4 as compared to their CD133- counterparts. Disseminated BM dormant TRP-2+ tumor cells were found to be co-localized with memory CD8+ T cells. Our data suggest that these dormant melanoma cells in the BM could play an important role in the maintenance of memory T cells in the BM.
Collapse
|
34
|
Romano S, Tufano M, D'Arrigo P, Vigorito V, Russo S, Romano MF. Cell stemness, epithelial-to-mesenchymal transition, and immunoevasion: Intertwined aspects in cancer metastasis. Semin Cancer Biol 2019; 60:181-190. [PMID: 31422157 DOI: 10.1016/j.semcancer.2019.08.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 12/22/2022]
Abstract
Recent advances in tumor immunology, fostered by dramatic outcomes with cancer immunotherapy, have opened new scenarios in cancer metastasis. The cancer stemness/mesenchymal phenotype and an excess of immune suppressive signals are emerging as Intertwined aspects of human tumors. This review examines recent studies that explored the mechanistic links between cancer cell stemness and immunoevasion, and the evidence points to these key events in cancer metastasis as two sides of the same coin. This review also covers the mechanisms involved in tumor expression of programmed cell death ligand 1 (PD-L1), a major factor exploited by human neoplasias to suppress immune control. We highlight the convergence of mesenchymal traits and PD-L1 expression and examine the functions of this immune inhibitory molecule, which confers cancer cell resistance and aggressiveness.
Collapse
Affiliation(s)
- Simona Romano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Italy
| | - Martina Tufano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Italy
| | - Paolo D'Arrigo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Italy
| | - Vincenza Vigorito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Italy
| | - Salvatore Russo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Italy
| | - Maria Fiammetta Romano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Italy.
| |
Collapse
|
35
|
Badrinath N, Yoo SY. Recent Advances in Cancer Stem Cell-Targeted Immunotherapy. Cancers (Basel) 2019; 11:cancers11030310. [PMID: 30841635 PMCID: PMC6468501 DOI: 10.3390/cancers11030310] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 02/22/2019] [Accepted: 03/01/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs) are one of the reasons for the relapse of cancer cells and metastasis. They have drug resistance against most chemotherapeutic agents. CSCs are also responsible for tumor cell heterogeneity and cause minimal residual disease. In order to achieve complete regression of tumors, CSCs have to be targeted. Recent advances in immunotherapies have shown promising outcomes in curing cancer, which are also applicable to target CSCs. CSCs express immune markers and exhibit specific immune characteristics in various cancers, which can be used in immunotherapies to target CSCs in the tumor microenvironment. Recently, various strategies have been used to target CSCs. Adaptive T-cells, dendritic cell (DC)-based vaccines, oncolytic viruses, immune checkpoint inhibitors, and combination therapies are now being used to target CSCs. Here, we discuss the feasibility of these immunological approaches and the recent trends in immunotherapies to target CSCs.
Collapse
Affiliation(s)
- Narayanasamy Badrinath
- Biomedical Sciences, School of Medicine, Pusan National University, Yangsan 50612, Korea.
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea.
| | - So Young Yoo
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea.
- BIO-IT Foundry Technology Institute, Pusan National University, Busan 46241, Korea.
| |
Collapse
|
36
|
Lian L, Li XL, Xu MD, Li XM, Wu MY, Zhang Y, Tao M, Li W, Shen XM, Zhou C, Jiang M. VEGFR2 promotes tumorigenesis and metastasis in a pro-angiogenic-independent way in gastric cancer. BMC Cancer 2019; 19:183. [PMID: 30819137 PMCID: PMC6393973 DOI: 10.1186/s12885-019-5322-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 01/25/2019] [Indexed: 02/08/2023] Open
Abstract
Background VEGF/VEGFR2 pathway is the central therapeutic target in anti-angiogenic treatment in multiple cancers. However, little work has been carried out concerning the pro-malignancy functions of VEGFR2 that are independent of its pro-angiogenesis effects in gastric cancer. Here, we demonstrated that VEGFR2 up-regulation in gastric cancer tissues was a prognostic marker for poor disease-free survival and overall survival of gastric cancer patients. Methods Immunohistochemistry was used to detect VEGFR2 and VTN expressions in specimens. Kaplan–Meier curves were constructed for survival analysis. Stably knockdown cell lines and overexpression cell lines were constructed by small interfering RNA and plasmids transfection. Real-time PCR and Western blot were used to confirm the expressions of target genes at both RNA and protein levels. Cell proliferation was measured by using Cell Counting Kit-8 and xenograft models. Microarray and bioinformatic analysis were also performed to identify the relationship between Vitronectin (VTN) and VEGFR2. Results When overexpressed in gastric cancer cells, VEGFR2 increased cellular proliferation and invasion in vitro and tumor formation in xenograft models. By using integrating microarray and bioinformatic analysis, we identifiedVTN as a downstream of VEGFR2 pathway. In gain- and loss-of function analysis in gastric cancer cells, VTN was further verified in consistent with VEGFR2 in expression levels and in regulating cell growth and motility in vitro and in vivo. Moreover, in gastric cancer samples, VTN was as also revealed as a poor prognostic factor. Conclusions Our present findings defined a novel activity for VEGFR2 in promoting tumorogenicity, motility and indicating a poor survival in gastric cancer beyond its known pro-angiogenic effects. Implications Our present findings defined a novel activity for VEGFR2 in promoting tumorogenicity, motility and indicating a poor survival in gastric cancer beyond its known pro-angiogenic effects, which may provide a new and valuable target for design of therapies for intervention and a new cognitive perspective for the anti-angiogenesis therapies. Electronic supplementary material The online version of this article (10.1186/s12885-019-5322-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lian Lian
- Department of Oncology, Suzhou Xiangcheng People's Hospital, Suzhou, 215131, China.,Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Xiang-Li Li
- Department of General Surgery, Suzhou Xiangcheng People's Hospital, Suzhou, 215131, China.,Comprehensive Cancer Center, Suzhou Xiangcheng People's Hospital, Suzhou, 215131, China
| | - Meng-Dan Xu
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Xian-Min Li
- Department of Oncology, Suzhou Xiangcheng People's Hospital, Suzhou, 215131, China
| | - Meng-Yao Wu
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yan Zhang
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Min Tao
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Wei Li
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Comprehensive Cancer Center, Suzhou Xiangcheng People's Hospital, Suzhou, 215131, China
| | - Xiao-Ming Shen
- Department of Oncology, Suzhou Xiangcheng People's Hospital, Suzhou, 215131, China.
| | - Chong Zhou
- Department of Radiation Oncology, Xuzhou Central Hospital, Xuzhou, 221009, China.
| | - Min Jiang
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
37
|
Chen M, Sharma A, Lin Y, Wu Y, He Q, Gu Y, Xu ZP, Monteiro M, Gu W. Insluin and epithelial growth factor (EGF) promote programmed death ligand 1(PD-L1) production and transport in colon cancer stem cells. BMC Cancer 2019; 19:153. [PMID: 30770752 PMCID: PMC6377751 DOI: 10.1186/s12885-019-5364-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/11/2019] [Indexed: 02/06/2023] Open
Abstract
Background Programmed cell death ligand 1 (PD-L1) is an important immune-inhibitory protein expressed on cancer cells to mediate cancer escape through interaction with PD-1 expressed on activated T lymphocytes (T cells). Previously, we reported that colon and breast cancer stem cells (CSCs) expressed much higher levels of PD-L1 than their parental cells, suggesting they will be more resistant to immune attack. Methods We investigated the underlining mechanism of PD-L1 increase in colon CSCs, with a special focus on the effect of insulin and epithelial growth factor (EGF), the two fundamental components to sustain the metabolism and stemness in the culture of CSCs. Results We found that insulin increased the total and surface PD-L1 levels through PI3K/Akt/mTOR pathway as the increase could be inhibited by the dual inhibitor of the pathway, BEZ235. EGF didn’t affect the total PD-L1 levels of CSCs but increased the cell surface protein levels by flow cytometry analysis, indicating EGF promotes the transport of PD-L1 to the cell surface. Blocking cell surface PD-L1 with a specific antibody resulted in a significant reduction of tumour sphere formation but didn’t interfere with the sphere growth, suggesting that cell surface PD-L1 may act as an adhering molecule for CSCs. Conclusions Apart from the essential roles in metabolism and stemness, insulin and EGF involve in up-regulation of PD-L1 expression in colon CSCs, therefore the inhibition of insulin and EGF/EGFR pathways can be considered for cancer immunotherapy or combined with PD-1/PD-L1 antibody-based cancer immunotherapy to eliminate CSCs.
Collapse
Affiliation(s)
- Mingshui Chen
- Australian Institute for Bioengineering and Nanotechnology (Building 75), The University of Queensland, Cooper Rd., St Lucia, Brisbane, QLD, 4072, Australia.,Laboratory of Immuno-Oncology, Department of Medical Oncology, Fujian Provincial Cancer Hospital &Institute, Fuzhou, 350014, China.,Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, China
| | - Aditi Sharma
- Australian Institute for Bioengineering and Nanotechnology (Building 75), The University of Queensland, Cooper Rd., St Lucia, Brisbane, QLD, 4072, Australia
| | - Yanling Lin
- Australian Institute for Bioengineering and Nanotechnology (Building 75), The University of Queensland, Cooper Rd., St Lucia, Brisbane, QLD, 4072, Australia
| | - Yanheng Wu
- Australian Institute for Bioengineering and Nanotechnology (Building 75), The University of Queensland, Cooper Rd., St Lucia, Brisbane, QLD, 4072, Australia
| | - Qi He
- Australian Institute for Bioengineering and Nanotechnology (Building 75), The University of Queensland, Cooper Rd., St Lucia, Brisbane, QLD, 4072, Australia
| | - Yushu Gu
- Australian Institute for Bioengineering and Nanotechnology (Building 75), The University of Queensland, Cooper Rd., St Lucia, Brisbane, QLD, 4072, Australia
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology (Building 75), The University of Queensland, Cooper Rd., St Lucia, Brisbane, QLD, 4072, Australia
| | - Michael Monteiro
- Australian Institute for Bioengineering and Nanotechnology (Building 75), The University of Queensland, Cooper Rd., St Lucia, Brisbane, QLD, 4072, Australia
| | - Wenyi Gu
- Australian Institute for Bioengineering and Nanotechnology (Building 75), The University of Queensland, Cooper Rd., St Lucia, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
38
|
Zhu S, Lv X, Zhang X, Li T, Zang G, Yang N, Wang X, Wu J, Chen W, Liu YJ, Chen J. An effective dendritic cell-based vaccine containing glioma stem-like cell lysate and CpG adjuvant for an orthotopic mouse model of glioma. Int J Cancer 2019; 144:2867-2879. [PMID: 30565657 DOI: 10.1002/ijc.32008] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 10/23/2018] [Accepted: 11/08/2018] [Indexed: 12/29/2022]
Abstract
Owing to the limited therapeutic efficacy of glioma vaccines, new strategies are required to improve cancer vaccines. Our study aimed to assess the therapeutic efficacy of a glioma vaccine called STDENVANT. This vaccine, comprising glioma stem-like cell (GSC) lysate, dendritic cells (DCs), and Toll-like receptor (TLR) 9 agonist CpG motif-containing oligodeoxynucleotides (CpG ODNs), was assessed using a GL261-C57BL/6 orthotopic mouse model of glioma. STDENVANT markedly improved survival and tumor regression by enhancing anti-tumor immune function. Moreover, STDENVANT upregulated programmed death 1 (PD-1) and its ligand PD-L1 on effector T cells, DCs, and glioma tissues, resulting in the accumulation of regulatory T (Treg) cells in the brain and lymph nodes. Combinatorial administration of anti-PD-L1 antibody and STDENVANT conferred a greater survival advantage and decreased the Treg cell population in the brain. The present results indicate that PD-L1 blockade can promote tumor regression via STDENVANT in a mouse model of glioma, and combinatorial administration of anti-PD-L1 antibody and STDENVANT increases the therapeutic anti-tumor efficacy of treatment.
Collapse
Affiliation(s)
- Shan Zhu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xinping Lv
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xuhao Zhang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Tete Li
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Guoxia Zang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Ning Yang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xue Wang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jing Wu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Wei Chen
- ADC Biomedical Research Institute, Saint Paul, MN
| | - Yong-Jun Liu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China.,Sanofi R&D, Cambridge, MA
| | - Jingtao Chen
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
39
|
Chromosome 19 miRNA cluster and CEBPB expression specifically mark and potentially drive triple negative breast cancers. PLoS One 2018; 13:e0206008. [PMID: 30335837 PMCID: PMC6193703 DOI: 10.1371/journal.pone.0206008] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/04/2018] [Indexed: 12/31/2022] Open
Abstract
Triple negative breast cancers (TNBCs) are known to express low PGR, ESR1, and ERBB2, and high KRT5, KRT14, and KRT17. However, the reasons behind the increased expressions of KRT5, KRT14, KRT17 and decreased expressions of PGR, ESR1, and ERBB2 in TNBCs are not fully understood. Here we show that, expression of chromosome 19 miRNA cluster (C19MC) specifically marks human TNBCs. Low REST and high CEBPB correlate with expression of C19MC, KRT5, KRT14, and KRT17 and enhancers of these genes/cluster are regulated by CEBPB and REST binding sites. The C19MC miRNAs in turn can potentially target REST to offer a positive feedback loop, and might target PGR, ESR1, ERBB2, GATA3, SCUBE2, TFF3 mRNAs to contribute towards TNBC phenotype. Thus our study demonstrates that C19MC miRNA expression marks TNBCs and that C19MC miRNAs and CEBPB might together determine the TNBC marker expression pattern.
Collapse
|
40
|
Dong P, Xiong Y, Yue J, Hanley SJB, Watari H. Tumor-Intrinsic PD-L1 Signaling in Cancer Initiation, Development and Treatment: Beyond Immune Evasion. Front Oncol 2018; 8:386. [PMID: 30283733 PMCID: PMC6156376 DOI: 10.3389/fonc.2018.00386] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 08/28/2018] [Indexed: 12/20/2022] Open
Abstract
Although the role of PD-L1 in suppressing the anti-tumor immune response is extensively documented, recent discoveries indicate a distinct tumor-intrinsic role for PD-L1 in modulating epithelial-to-mesenchymal transition (EMT), cancer stem cell (CSC)-like phenotype, metastasis and resistance to therapy. In this review, we will focus on the newly discovered functions of PD-L1 in the regulation of cancer development, describe underlying molecular mechanisms responsible for PD-L1 upregulation and discuss current insights into novel components of PD-L1 signaling. Furthermore, we summarize our current understanding of the link between PD-L1 signaling and the EMT program as well as the CSC state. Tumor cell-intrinsic PD-L1 clearly contributes to cancer stemness, EMT, tumor invasion and chemoresistance in multiple tumor types. Conversely, activation of OCT4 signaling and upregulation of EMT inducer ZEB1 induce PD-L1 expression in cancer cells, thereby suggesting a possible immune evasion mechanism employed by cancer stem cells during metastasis. Our meta-analysis demonstrated that PD-L1 is co-amplified along with MYC, SOX2, N-cadherin and SNAI1 in the TCGA endometrial and ovarian cancer datasets. Further identification of immune-independent PD-L1 functions and characterization of crucial signaling events upstream or downstream of PD-L1 in diverse cancer types and specific cancer subtypes, would provide additional targets and new therapeutic approaches.
Collapse
Affiliation(s)
- Peixin Dong
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan
| | - Ying Xiong
- Department of Gynecology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Junming Yue
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Sharon J B Hanley
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hidemichi Watari
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
41
|
Essaadi A, Nollet M, Moyon A, Stalin J, Simoncini S, Balasse L, Bertaud A, Bachelier R, Leroyer AS, Sarlon G, Guillet B, Dignat-George F, Bardin N, Blot-Chabaud M. Stem cell properties of peripheral blood endothelial progenitors are stimulated by soluble CD146 via miR-21: potential use in autologous cell therapy. Sci Rep 2018; 8:9387. [PMID: 29925894 PMCID: PMC6010456 DOI: 10.1038/s41598-018-27715-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 05/21/2018] [Indexed: 12/27/2022] Open
Abstract
Cell-based therapies constitute a real hope for the treatment of ischaemic diseases. One of the sources of endothelial progenitors for autologous cell therapy is Endothelial Colony Forming Cells (ECFC) that can be isolated from peripheral blood. However, their use is limited by their low number in the bloodstream and the loss of their stem cell phenotype associated with the acquisition of a senescent phenotype in culture. We hypothesized that adding soluble CD146, a novel endothelial growth factor with angiogenic properties, during the isolation and growth procedures could improve their number and therapeutic potential. Soluble CD146 increased the number of isolated peripheral blood ECFC colonies and lowered their onset time. It prevented cellular senescence, induced a partial mesenchymal phenotype and maintained a stem cell phenotype by stimulating the expression of embryonic transcription factors. These different effects were mediated through the induction of mature miR-21. When injected in an animal model of hindlimb ischaemia, sCD146-primed ECFC isolated from 40 ml of blood from patients with peripheral arterial disease were able to generate new blood vessels and restore blood flow. Treatment with sCD146 could thus constitute a promising strategy to improve the use of autologous cells for the treatment of ischaemic diseases.
Collapse
Affiliation(s)
- Amel Essaadi
- Aix Marseille Univ, INSERM 1263, INRA 1260, C2VN, Marseille, France
| | - Marie Nollet
- Aix Marseille Univ, INSERM 1263, INRA 1260, C2VN, Marseille, France
| | - Anaïs Moyon
- Aix Marseille Univ, INSERM 1263, INRA 1260, C2VN, Marseille, France.,CERIMED (European Center of Research in Medical Imaging), Aix-Marseille University, Marseille, France
| | - Jimmy Stalin
- Aix Marseille Univ, INSERM 1263, INRA 1260, C2VN, Marseille, France
| | | | - Laure Balasse
- CERIMED (European Center of Research in Medical Imaging), Aix-Marseille University, Marseille, France
| | | | | | | | - Gabrielle Sarlon
- Service of Vascular Surgery, La Timone Hospital, Marseille, France
| | - Benjamin Guillet
- Aix Marseille Univ, INSERM 1263, INRA 1260, C2VN, Marseille, France.,CERIMED (European Center of Research in Medical Imaging), Aix-Marseille University, Marseille, France
| | | | - Nathalie Bardin
- Aix Marseille Univ, INSERM 1263, INRA 1260, C2VN, Marseille, France
| | | |
Collapse
|
42
|
Wang Q, Feng F, Wang F, Liu Z, Liu S, Xu G, Zheng G, Guo M, Lian X, Zhang H. PD-L1 Expression On tumor Cells Was Associated With Unfavorable Prognosis In Esophageal Squamous Cell Carcinoma. J Cancer 2018; 9:2224-2231. [PMID: 29937943 PMCID: PMC6010673 DOI: 10.7150/jca.24493] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/26/2018] [Indexed: 12/28/2022] Open
Abstract
Background: Evidence about the association between programmed cell death ligand 1 (PD-L1) expression and prognosis of esophageal squamous cell carcinoma (ESCC) were limited and controversial. Thus, the present study aims to investigate the prognostic value of tumor immune microenvironment (TIM) based on PD-L1 expression and CD8+ T cell infiltration in ESCC tissues. Methods: From September 2008 to March 2010, a total of 146 ESCC patients received radical esophagectomy were retrospectively analyzed in our present study. PD-L1 expression and CD8+ T cell infiltration were evaluated through immunohistochemistry. The clinicopathological characteristics and survival were analyzed. Results: There were 111 male and 35 female. The median age was 59.1 years (37-78 years). The positive rate of PD-L1 expression was 61.7%. The rate of high CD8+ T cell infiltration was 33%. No significant differences were found between clinicopathological features and PD-L1 expression or CD8+ T cell infiltration. PD-L1 expression was significantly associated with poor overall survival (P=0.010). However, CD8+ T cell infiltration was not a prognostic risk factor. Type of TIM was significantly associated with the prognosis of ESCC patients (P=0.021). Conclusions: PD-L1 expression was an independent risk factor for the prognosis of ESCC patients. Immunotherapy may achieve promising outcomes in ESCC patients with type I TIM.
Collapse
Affiliation(s)
- Qiao Wang
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University,127 West Changle Road, 710032, Xi'an, Shaanxi, China.,Department of General Surgery, No. 91 Central Hospital of PLA, 454000, Jiaozuo, Henan, China
| | - Fan Feng
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University,127 West Changle Road, 710032, Xi'an, Shaanxi, China
| | - Fei Wang
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University,127 West Changle Road, 710032, Xi'an, Shaanxi, China.,Department of General Surgery, No. 534 Hospital of PLA, 471000, Luoyang, Henan, China
| | - Zhen Liu
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University,127 West Changle Road, 710032, Xi'an, Shaanxi, China
| | - Shushang Liu
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University,127 West Changle Road, 710032, Xi'an, Shaanxi, China
| | - Guanghui Xu
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University,127 West Changle Road, 710032, Xi'an, Shaanxi, China
| | - Gaozan Zheng
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University,127 West Changle Road, 710032, Xi'an, Shaanxi, China
| | - Man Guo
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University,127 West Changle Road, 710032, Xi'an, Shaanxi, China
| | - Xiao Lian
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University,127 West Changle Road, 710032, Xi'an, Shaanxi, China
| | - Hongwei Zhang
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University,127 West Changle Road, 710032, Xi'an, Shaanxi, China
| |
Collapse
|
43
|
Zhang M, Lu L, Ying M, Ruan H, Wang X, Wang H, Chai Z, Wang S, Zhan C, Pan J, Lu W. Enhanced Glioblastoma Targeting Ability of Carfilzomib Enabled by a DA7R-Modified Lipid Nanodisk. Mol Pharm 2018; 15:2437-2447. [PMID: 29734808 DOI: 10.1021/acs.molpharmaceut.8b00270] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The robust proliferation of tumors relies on a rich neovasculature for nutrient supplies. Therefore, a basic strategy of tumor targeting therapy should include not only killing regular cancer cells but also blocking tumor neovasculature. D-peptide DA7R, which was previously reported to specifically bind vascular endothelial growth factor receptor 2 (VEGFR2) and neuropilin-1 (NRP-1), could achieve the goal of multitarget recognition. Accordingly, the main purposes of this work were to establish a carfilzomib-loaded lipid nanodisk modified with multifunctional peptide DA7R (DA7R-ND/CFZ) and to evaluate its anti-glioblastoma efficacy in vitro and in vivo. It is testified that the DA7R peptide-conjugated lipid nanodisk can be specifically taken up by U87MG cells and HUVECs. Furthermore, DA7R-ND demonstrated a more enhanced penetration than that of the nonmodified formulation on the tumor spheroid model in vitro and more tumor region accumulation in vivo on the subcutaneous and intracranial tumor-bearing nude mice model. DA7R-ND was shown to co-localize with tumor neovasculature in vivo. When loaded with proteasome inhibitor carfilzomib, the DA7R-decorated nanodisk could remarkably suppress tumor proliferation, extend survival time of nude mice bearing an intracranial tumor, and inhibit neovasculature formation with an efficacy higher than that of the nonmodified nanodisk in vitro and in vivo. The present study verified that the heptapeptide DA7R-conjugated nanodisk is a promising nanocarrier for glioblastoma targeting therapy.
Collapse
Affiliation(s)
- Mingfei Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, and Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, & State Key Laboratory of Medical Neurobiology, and Collaborative Innovation Center for Brain Science , Fudan University , Shanghai 200032 , China
| | - Linwei Lu
- Department of Integrative Medicine, Huashan Hospital , Fudan University, & Institute of Integrative Medicine of Fudan University , Shanghai 200041 , China
| | - Man Ying
- Department of Pharmaceutics, School of Pharmacy, Fudan University, and Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, & State Key Laboratory of Medical Neurobiology, and Collaborative Innovation Center for Brain Science , Fudan University , Shanghai 200032 , China
| | - Huitong Ruan
- Department of Pharmaceutics, School of Pharmacy, Fudan University, and Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, & State Key Laboratory of Medical Neurobiology, and Collaborative Innovation Center for Brain Science , Fudan University , Shanghai 200032 , China
| | - Xiaoyi Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, and Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, & State Key Laboratory of Medical Neurobiology, and Collaborative Innovation Center for Brain Science , Fudan University , Shanghai 200032 , China
| | - Huan Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, and Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, & State Key Laboratory of Medical Neurobiology, and Collaborative Innovation Center for Brain Science , Fudan University , Shanghai 200032 , China
| | - Zhilan Chai
- Department of Pharmaceutics, School of Pharmacy, Fudan University, and Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, & State Key Laboratory of Medical Neurobiology, and Collaborative Innovation Center for Brain Science , Fudan University , Shanghai 200032 , China
| | - Songli Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, and Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, & State Key Laboratory of Medical Neurobiology, and Collaborative Innovation Center for Brain Science , Fudan University , Shanghai 200032 , China
| | - Changyou Zhan
- School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers , Fudan University , Shanghai 200032 , P.R. China
| | - Jun Pan
- Department of Pharmaceutics, School of Pharmacy, Fudan University, and Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, & State Key Laboratory of Medical Neurobiology, and Collaborative Innovation Center for Brain Science , Fudan University , Shanghai 200032 , China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University, and Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, & State Key Laboratory of Medical Neurobiology, and Collaborative Innovation Center for Brain Science , Fudan University , Shanghai 200032 , China.,Minhang Branch, Zhongshan Hospital and Institute of Fudan-Minghang Academic Health System , Minghang Hospital, Fudan University , Shanghai 201199 , China
| |
Collapse
|
44
|
Hur W, Yoon SK. Molecular Pathogenesis of Radiation-Induced Cell Toxicity in Stem Cells. Int J Mol Sci 2017; 18:ijms18122749. [PMID: 29258244 PMCID: PMC5751348 DOI: 10.3390/ijms18122749] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 12/16/2017] [Accepted: 12/17/2017] [Indexed: 12/26/2022] Open
Abstract
Radiation therapy is an effective cancer therapy, but damage to normal tissues surrounding the tumor due to radiotherapy causes severe complications. The importance of the therapeutic area between tumor suppression and normal tissue injury has long been highlighted in radiation therapy. Recent advances in stem cell biology have shown that stem cell (SC) responses to genotoxic stresses of ionizing radiation can improve the therapeutic effect of radiation by repairing damaged cells. In contrast, cancer stem cells (CSCs), a small subpopulation of cells within tumors, are generally resistant to chemotherapy and radiotherapy and cause tumor recurrence. Although the underlying mechanisms are not clearly understood in detail, efforts are still underway to identify SC treatment or CSC resistant pathogenesis of DNA damage agents such as radiation therapy. In response to radiation, CSCs differ from normal SCs in their biological properties due to severe deregulation of the self-renewal ability in CSCs. Differences of cleavage mode, cell cycle characteristics, replication potential, and activation/inactivation of DNA damage treatment and cancer-specific molecular pathways between normal SCs and CSCs confer a malignant phenotype upon CSCs. However, further studies are needed to identify normal SC and CSC-specific targets. In this review, we summarize the current advances in research regarding how normal SCs and CSCs respond to ionizing radiation, with a special emphasis on cell toxicity, radiosensitivity, signaling networks, DNA damage response (DDR) and DNA repair. In addition, we discuss strategies to develop new diagnostic and therapeutic techniques for predicting responses to cancer treatment and overcoming radiation-related toxicity.
Collapse
Affiliation(s)
- Wonhee Hur
- The Catholic University Liver Research Center & WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Seung Kew Yoon
- The Catholic University Liver Research Center & WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
- Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| |
Collapse
|
45
|
Molecular genetics and cellular events of K-Ras-driven tumorigenesis. Oncogene 2017; 37:839-846. [PMID: 29059163 PMCID: PMC5817384 DOI: 10.1038/onc.2017.377] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 08/11/2017] [Accepted: 09/08/2017] [Indexed: 02/06/2023]
Abstract
Cellular transformation and the accumulation of genomic instability are the two key events required for tumorigenesis. K-Ras (Kirsten-rat sarcoma viral oncogene homolog) is a prominent oncogene that has been proven to drive tumorigenesis. K-Ras also modulates numerous genetic regulatory mechanisms and forms a large tumorigenesis network. In this review, we track the genetic aspects of K-Ras signaling networks and assemble the sequence of cellular events that constitute the tumorigenesis process, such as regulation of K-Ras expression (which is influenced by miRNA, small nucleolar RNA and lncRNA), activation of K-Ras (mutations), generation of reactive oxygen species (ROS), induction of DNA damage and apoptosis, induction of DNA damage repair pathways and ROS detoxification systems, cellular transformation after apoptosis by the blebbishield emergency program and the accumulation of genomic/chromosomal instability that leads to tumorigenesis.
Collapse
|