1
|
Ma L, Zhang J, Dai Z, Liao P, Guan J, Luo Z. Top 100 most-cited articles on apoptosis of non-small cell lung cancer over the past two decades: a bibliometrics analysis. Front Immunol 2025; 15:1512349. [PMID: 39872524 PMCID: PMC11770037 DOI: 10.3389/fimmu.2024.1512349] [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: 10/16/2024] [Accepted: 12/09/2024] [Indexed: 01/30/2025] Open
Abstract
Background Recently there has been an increasing number of studies have explored apoptosis mechanisms in lung cancer (LC). However, no researchers have conducted a bibliometric analysis of the most cited articles in this field. Objective To examine the top 100 most influential and cited publications on apoptosis in non-small cell lung cancer (NSCLC) from 2004 to 2023, summarizing research trends and key focus areas. Methods This study utilized the Web of Science Core Database (WOSCC) to research NSCLC apoptosis from 2004 to 2023, using keyword selection and manual screening for article searches. Bibliometrix package of R software 4.3.1 was used to generate distribution statistics for the top ten institutions, journals and authors. Citespace6.2. R6 was used to create the visualization maps for keyword co-occurrence and clustering. VOSviewer1.6.19 was used to conduct cluster analysis of publishing countries (regions), with data exported to SCImago Graphica for geographic visualization and cooperation analysis. VOSviewer1.6.19 was used to produced co-citation maps of institutions, journals, authors, and references. Results From 2004 to 2023, 13316 articles were retrieved, and the top 100 most cited were chosen. These were authored by 934 individuals from 269 institutions across 18 countries and appeared in 45 journals. Citations ranged from 150 to 1,389, with a median of 209.5. The most influential articles appeared in 2005 and 2007 (n=13). The leading countries (regions), institutions, journals and authors were identified as the United States (n=60), Harvard University (n=64), CANCER RESEARCH (n=15), SUN M and YANG JS (n=6). The top five keywords were "expression", "activation", "apoptosis", "pathway" and "gefitinib". This study indicates that enhancing apoptosis through circular RNA regulation and targeting the Nrf2 signaling pathway could become a key research focus in recent years. Conclusion Apoptosis has been the subject of extensive research over many years, particularly in relation to its role in the pathogenesis, diagnosis, and treatment of NSCLC. This study aims to identify highly influential articles and forecast emerging research trends, thereby offering insights into novel therapeutic targets and strategies to overcome drug resistance. The findings are intended to serve as a valuable reference for scholars engaged in this field of study.
Collapse
Affiliation(s)
- Leshi Ma
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jing Zhang
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zi Dai
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pei Liao
- Department of Oncology, Chongqing Hospital, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Chongqing, China
| | - Jieshan Guan
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Oncology, Shenshan Hospital, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Shanwei, China
| | - Zhijie Luo
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
2
|
Phookphan P, Racha S, Yokoya M, Ei ZZ, Hotta D, Zou H, Chanvorachote P. A New Renieramycin T Right-Half Analog as a Small Molecule Degrader of STAT3. Mar Drugs 2024; 22:370. [PMID: 39195486 DOI: 10.3390/md22080370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024] Open
Abstract
Constitutive activation of STAT3 contributes to tumor development and metastasis, making it a promising target for cancer therapy. (1R,4R,5S)-10-hydroxy-9-methoxy-8,11-dimethyl-3-(naphthalen-2-ylmethyl)-1,2,3,4,5,6-hexahydro-1,5-epiminobenzo[d]azocine-4-carbonitrile, DH_31, a new derivative of the marine natural product Renieramycin T, showed potent activity against H292 and H460 cells, with IC50 values of 5.54 ± 1.04 µM and 2.9 ± 0.58 µM, respectively. Structure-activity relationship (SAR) analysis suggests that adding a naphthalene ring with methyl linkers to ring C and a hydroxyl group to ring E enhances the cytotoxic effect of DH_31. At 1-2.5 µM, DH_31 significantly inhibited EMT phenotypes such as migration, and sensitized cells to anoikis. Consistent with the upregulation of ZO1 and the downregulation of Snail, Slug, N-cadherin, and Vimentin at both mRNA and protein levels, in silico prediction identified STAT3 as a target, validated by protein analysis showing that DH_31 significantly decreases STAT3 levels through ubiquitin-proteasomal degradation. Immunofluorescence and Western blot analysis confirmed that DH_31 significantly decreased STAT3 and EMT markers. Additionally, molecular docking suggests a covalent interaction between the cyano group of DH_31 and Cys-468 in the DNA-binding domain of STAT3 (binding affinity = -7.630 kcal/mol), leading to destabilization thereafter. In conclusion, DH_31, a novel RT derivative, demonstrates potential as a STAT3-targeting drug that significantly contribute to understanding of the development of new targeted therapy.
Collapse
Affiliation(s)
- Preeyaphan Phookphan
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Satapat Racha
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Interdisciplinary Program in Pharmacology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
| | - Masashi Yokoya
- Department of Pharmaceutical Chemistry, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose, Tokyo 204-8588, Japan
| | - Zin Zin Ei
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Daiki Hotta
- Department of Pharmaceutical Chemistry, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose, Tokyo 204-8588, Japan
| | - Hongbin Zou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Pithi Chanvorachote
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| |
Collapse
|
3
|
Abe Y, Sano T, Otsuka N, Ogawa M, Tanaka N. PRMT5-mediated methylation of STAT3 is required for lung cancer stem cell maintenance and tumour growth. Commun Biol 2024; 7:593. [PMID: 38760429 PMCID: PMC11101626 DOI: 10.1038/s42003-024-06290-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 05/03/2024] [Indexed: 05/19/2024] Open
Abstract
STAT3 is constitutively activated in many cancer types, including lung cancer, and can induce cancer cell proliferation and cancer stem cell (CSC) maintenance. STAT3 is activated by tyrosine kinases, such as JAK and SRC, but the mechanism by which STAT3 maintains its activated state in cancer cells remains unclear. Here, we show that PRMT5 directly methylates STAT3 and enhances its activated tyrosine phosphorylation in non-small cell lung cancer (NSCLC) cells. PRMT5 expression is also induced by STAT3, suggesting the presence of a positive feedback loop in cancer cells. Furthermore, methylation of STAT3 at arginine 609 by PRMT5 is important for its transcriptional activity and support of tumour growth and CSC maintenance. Indeed, NSCLC cells expressing the STAT3 mutant which R609 was replaced to alanine (R609K) show significantly impaired tumour growth in nude mice. Overall, our study reveals a mechanism by which STAT3 remains activated in NSCLC and provides a new target for cancer therapeutic approaches.
Collapse
Affiliation(s)
- Yoshinori Abe
- Laboratory of Molecular Analysis, Nippon Medical School, Tokyo, Japan
- Department of Molecular Oncology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Takumi Sano
- Department of Molecular Oncology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Naoki Otsuka
- Department of Molecular Oncology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Masashi Ogawa
- Department of Molecular Oncology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Nobuyuki Tanaka
- Department of Molecular Oncology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan.
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan.
| |
Collapse
|
4
|
Hu Y, Dong Z, Liu K. Unraveling the complexity of STAT3 in cancer: molecular understanding and drug discovery. J Exp Clin Cancer Res 2024; 43:23. [PMID: 38245798 PMCID: PMC10799433 DOI: 10.1186/s13046-024-02949-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/08/2024] [Indexed: 01/22/2024] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a transcriptional factor involved in almost all cancer hallmark features including tumor proliferation, metastasis, angiogenesis, immunosuppression, tumor inflammation, metabolism reprogramming, drug resistance, cancer stemness. Therefore, STAT3 has become a promising therapeutic target in a wide range of cancers. This review focuses on the up-to-date knowledge of STAT3 signaling in cancer. We summarize both the positive and negative modulators of STAT3 together with the cancer hallmarks involving activities regulated by STAT3 and highlight its extremely sophisticated regulation on immunosuppression in tumor microenvironment and metabolic reprogramming. Direct and indirect inhibitors of STAT3 in preclinical and clinical studies also have been summarized and discussed. Additionally, we highlight and propose new strategies of targeting STAT3 and STAT3-based combinations with established chemotherapy, targeted therapy, immunotherapy and combination therapy. These efforts may provide new perspectives for STAT3-based target therapy in cancer.
Collapse
Affiliation(s)
- Yamei Hu
- Tianjian Laboratory for Advanced Biomedical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
- Medical Research Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zigang Dong
- Tianjian Laboratory for Advanced Biomedical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China.
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China.
| | - Kangdong Liu
- Tianjian Laboratory for Advanced Biomedical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan, China.
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, China.
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China.
| |
Collapse
|
5
|
James A, Akash K, Sharma A, Bhattacharyya S, Sriamornsak P, Nagraik R, Kumar D. Himalayan flora: targeting various molecular pathways in lung cancer. Med Oncol 2023; 40:314. [PMID: 37787816 DOI: 10.1007/s12032-023-02171-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/21/2023] [Indexed: 10/04/2023]
Abstract
The fatal amplification of lung cancer across the globe and the limitations of current treatment strategies emphasize the necessity for substitute therapeutics. The incorporation of phyto-derived components in chemo treatment holds promise in addressing those challenges. Despite the significant progressions in lung cancer therapeutics, the complexities of molecular mechanism and pathways underlying this disease remain inadequately understood, necessitating novel biomarker targeting. The Himalayas, abundant in diverse plant varieties with established chemotherapeutic potential, presents a promising avenue for investigating potential cures for lung carcinoma. The vast diversity of phytocompounds herein can be explored for targeting the disease. This review delves into the multifaceted targets of lung cancer and explores the established phytochemicals with their specific molecular targets. It emphasizes comprehending the intricate pathways that govern effective therapeutic interventions for lung cancer. Through this exploration of Himalayan flora, this review seeks to illuminate potential breakthroughs in lung cancer management using natural compounds. The amalgamation of Himalayan plant-derived compounds with cautiously designed combined therapeutic approaches such as nanocarrier-mediated drug delivery and synergistic therapy offers an opportunity to redefine the boundaries of lung cancer treatment by reducing the drug resistance and side effects and enabling an effective targeted delivery of drugs. Furthermore, additional studies are obligatory to understand the possible derivation of natural compounds used in current lung cancer treatment from plant species within the Himalayan region.
Collapse
Affiliation(s)
- Abija James
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - K Akash
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Avinash Sharma
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Sanjib Bhattacharyya
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, 400715, Chongqing, People's Republic of China
- Department of Sciences, Nirma University, Ahmedabad, Gujarat, 382481, India
| | | | - Rupak Nagraik
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India.
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India.
| |
Collapse
|
6
|
Alam M, Hasan GM, Eldin SM, Adnan M, Riaz MB, Islam A, Khan I, Hassan MI. Investigating regulated signaling pathways in therapeutic targeting of non-small cell lung carcinoma. Biomed Pharmacother 2023; 161:114452. [PMID: 36878052 DOI: 10.1016/j.biopha.2023.114452] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/19/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Non-small cell lung carcinoma (NSCLC) is the most common malignancy worldwide. The signaling cascades are stimulated via genetic modifications in upstream signaling molecules, which affect apoptotic, proliferative, and differentiation pathways. Dysregulation of these signaling cascades causes cancer-initiating cell proliferation, cancer development, and drug resistance. Numerous efforts in the treatment of NSCLC have been undertaken in the past few decades, enhancing our understanding of the mechanisms of cancer development and moving forward to develop effective therapeutic approaches. Modifications of transcription factors and connected pathways are utilized to develop new treatment options for NSCLC. Developing designed inhibitors targeting specific cellular signaling pathways in tumor progression has been recommended for the therapeutic management of NSCLC. This comprehensive review provided deeper mechanistic insights into the molecular mechanism of action of various signaling molecules and their targeting in the clinical management of NSCLC.
Collapse
Affiliation(s)
- Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Sayed M Eldin
- Center of Research, Faculty of Engineering, Future University in Egypt, New Cairo 11835, Egypt
| | - Mohd Adnan
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Muhammad Bilal Riaz
- Faculty of Applied Physics and Mathematics, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdnask, Poland; Department of Computer Science and Mathematics, Lebanese American University, Byblos, Lebanon
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Ilyas Khan
- Department of Mathematics, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
7
|
Papavassiliou KA, Marinos G, Papavassiliou AG. Combining STAT3-Targeting Agents with Immune Checkpoint Inhibitors in NSCLC. Cancers (Basel) 2023; 15:386. [PMID: 36672335 PMCID: PMC9857288 DOI: 10.3390/cancers15020386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/31/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Despite recent therapeutic advances, non-small cell lung cancer (NSCLC) remains the leading cause of cancer-related death. Signal transducer and activator of transcription 3 (STAT3) is a transcription factor (TF) with multiple tumor-promoting effects in NSCLC, including proliferation, anti-apoptosis, angiogenesis, invasion, metastasis, immunosuppression, and drug resistance. Recent studies suggest that STAT3 activation contributes to resistance to immune checkpoint inhibitors. Thus, STAT3 represents an attractive target whose pharmacological modulation in NSCLC may assist in enhancing the efficacy of or overcoming resistance to immune checkpoint inhibitors. In this review, we discuss the biological mechanisms through which STAT3 inhibition synergizes with or overcomes resistance to immune checkpoint inhibitors and highlight the therapeutic strategy of using drugs that target STAT3 as potential combination partners for immune checkpoint inhibitors in the management of NSCLC patients.
Collapse
Affiliation(s)
- Kostas A. Papavassiliou
- First University Department of Respiratory Medicine, Medical School, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Georgios Marinos
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
8
|
NT157 exerts antineoplastic activity by targeting JNK and AXL signaling in lung cancer cells. Sci Rep 2022; 12:17092. [PMID: 36224313 PMCID: PMC9556623 DOI: 10.1038/s41598-022-21419-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 09/27/2022] [Indexed: 01/04/2023] Open
Abstract
Combination therapies or multi-targeted drugs have been pointed out as an option to prevent the emergence of resistant clones, which could make long-term treatment more effective and translate into better clinical outcomes for cancer patients. The NT157 compound is a synthetic tyrphostin that leads to long-term inhibition of IGF1R/IRS1-2-, STAT3- and AXL-mediated signaling pathways. Given the importance of these signaling pathways for the development and progression of lung cancer, this disease becomes an interesting model for generating preclinical evidence on the cellular and molecular mechanisms underlying the antineoplastic activity of NT157. In lung cancer cells, exposure to NT157 decreased, in a dose-dependent manner, cell viability, clonogenicity, cell cycle progression and migration, and induced apoptosis (p < 0.05). In the molecular scenario, NT157 reduced expression of IRS1 and AXL and phosphorylation of p38 MAPK, AKT, and 4EBP1. Besides, NT157 decreased expression of oncogenes BCL2, CCND1, MYB, and MYC and increased genes related to cellular stress and apoptosis, JUN, BBC3, CDKN1A, CDKN1B, FOS, and EGR1 (p < 0.05), favoring a tumor-suppressive cell signaling network in the context of lung cancer. Of note, JNK was identified as a key kinase for NT157-induced IRS1 and IRS2 phosphorylation, revealing a novel axis involved in the mechanism of action of the drug. NT157 also presented potentiating effects on EGFR inhibitors in lung cancer cells. In conclusion, our preclinical findings highlight NT157 as a putative prototype of a multitarget drug that may contribute to the antineoplastic arsenal against lung cancer.
Collapse
|
9
|
Hashimoto S, Hashimoto A, Muromoto R, Kitai Y, Oritani K, Matsuda T. Central Roles of STAT3-Mediated Signals in Onset and Development of Cancers: Tumorigenesis and Immunosurveillance. Cells 2022; 11:cells11162618. [PMID: 36010693 PMCID: PMC9406645 DOI: 10.3390/cells11162618] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/12/2022] [Accepted: 08/20/2022] [Indexed: 02/07/2023] Open
Abstract
Since the time of Rudolf Virchow in the 19th century, it has been well-known that cancer-associated inflammation contributes to tumor initiation and progression. However, it remains unclear whether a collapse of the balance between the antitumor immune response via the immunological surveillance system and protumor immunity due to cancer-related inflammation is responsible for cancer malignancy. The majority of inflammatory signals affect tumorigenesis by activating signal transducer and activation of transcription 3 (STAT3) and nuclear factor-κB. Persistent STAT3 activation in malignant cancer cells mediates extremely widespread functions, including cell growth, survival, angiogenesis, and invasion and contributes to an increase in inflammation-associated tumorigenesis. In addition, intracellular STAT3 activation in immune cells causes suppressive effects on antitumor immunity and leads to the differentiation and mobilization of immature myeloid-derived cells and tumor-associated macrophages. In many cancer types, STAT3 does not directly rely on its activation by oncogenic mutations but has important oncogenic and malignant transformation-associated functions in both cancer and stromal cells in the tumor microenvironment (TME). We have reported a series of studies aiming towards understanding the molecular mechanisms underlying the proliferation of various types of tumors involving signal-transducing adaptor protein-2 as an adaptor molecule that modulates STAT3 activity, and we recently found that AT-rich interactive domain-containing protein 5a functions as an mRNA stabilizer that orchestrates an immunosuppressive TME in malignant mesenchymal tumors. In this review, we summarize recent advances in our understanding of the functional role of STAT3 in tumor progression and introduce novel molecular mechanisms of cancer development and malignant transformation involving STAT3 activation that we have identified to date. Finally, we discuss potential therapeutic strategies for cancer that target the signaling pathway to augment STAT3 activity.
Collapse
Affiliation(s)
- Shigeru Hashimoto
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
- Correspondence: (S.H.); (T.M.)
| | - Ari Hashimoto
- Department of Molecular Biology, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Ryuta Muromoto
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Yuichi Kitai
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Kenji Oritani
- Department of Hematology, International University of Health and Welfare, Narita 286-8686, Japan
| | - Tadashi Matsuda
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
- Correspondence: (S.H.); (T.M.)
| |
Collapse
|
10
|
Sun C, Liu X, Sun N, Zhang X, Shah M, Zhang G, Che Q, Zhu T, Li J, Li D. Cytotoxic Nitrobenzoyl Sesquiterpenoids from an Antarctica Sponge-Derived Aspergillus insulicola. JOURNAL OF NATURAL PRODUCTS 2022; 85:987-996. [PMID: 35380848 DOI: 10.1021/acs.jnatprod.1c01118] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive neoplastic diseases of the pancreas with fatal proliferation and metastasis and no medicine available for treatment. From an Antarctica sponge-derived fungus, Aspergillus insulicola HDN151418, four new nitrobenzoyl sesquiterpenoids, namely, insulicolides D-G (1-4), were isolated. Compounds 3 and 4 exhibited selective inhibition against human PDAC cell lines. Further studies indicated that compound 4 could significantly suppress cell proliferation to induce apoptosis and blocked migration and invasion of PDAC cells. Compound 4 could also avoid resistance and improved the therapeutic effect of the chemotherapy drug gemcitabine. A preliminary mechanism study showed that compound 4 can significantly inhibit the expression of EGFR and XIAP in PDAC cells. Altogether, 4 is a potential lead compound for anti-PDAC drug research.
Collapse
Affiliation(s)
- Chunxiao Sun
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Xiaoyu Liu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Ning Sun
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Xiaomin Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Mudassir Shah
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Guojian Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, People's Republic of China
| | - Qian Che
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Tianjiao Zhu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Jing Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Dehai Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, People's Republic of China
| |
Collapse
|
11
|
Yang R, Song Y, Shakoor K, Yi W, Peng C, Liu S. Insights into the role of STAT3 in intrahepatic cholangiocarcinoma (Review). Mol Med Rep 2022; 25:171. [PMID: 35302174 PMCID: PMC8971913 DOI: 10.3892/mmr.2022.12687] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/22/2022] [Indexed: 01/27/2023] Open
Abstract
Intrahepatic cholangiocarcinoma (ICC) is a primary malignant liver tumour whose incidence is second only to that of hepatocellular carcinoma. ICC is a highly heterogeneous disease arising from neoplastic transformation of intrahepatic biliary epithelial cells (cholangiocytes), and it is characterized by a very poor prognosis. Signal transducer and activator of transcription 3 (STAT3) is an important oncogene that is widely expressed in numerous cancers. STAT3 is a candidate target for the treatment of ICC. However, studies on STAT3 and the occurrence and development of ICC require improvements. Therefore, the present review summarized the mechanism of STAT3 in ICC and provided a theoretical basis for STAT3 to become an effective target for determining the prognosis and treatment of ICC.
Collapse
Affiliation(s)
- Ranzhiqiang Yang
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, P.R. China
| | - Yinghui Song
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, P.R. China
| | - Kashif Shakoor
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, P.R. China
| | - Weimin Yi
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, P.R. China
| | - Chuang Peng
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, P.R. China
| | - Sulai Liu
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, P.R. China
| |
Collapse
|
12
|
Parakh S, Ernst M, Poh AR. Multicellular Effects of STAT3 in Non-small Cell Lung Cancer: Mechanistic Insights and Therapeutic Opportunities. Cancers (Basel) 2021; 13:6228. [PMID: 34944848 PMCID: PMC8699548 DOI: 10.3390/cancers13246228] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common type of lung cancer and accounts for 85% of lung cancer cases. Aberrant activation of the Signal Transducer and Activator of Transcription 3 (STAT3) is frequently observed in NSCLC and is associated with a poor prognosis. Pre-clinical studies have revealed an unequivocal role for tumor cell-intrinsic and extrinsic STAT3 signaling in NSCLC by promoting angiogenesis, cell survival, cancer cell stemness, drug resistance, and evasion of anti-tumor immunity. Several STAT3-targeting strategies have also been investigated in pre-clinical models, and include preventing upstream receptor/ligand interactions, promoting the degradation of STAT3 mRNA, and interfering with STAT3 DNA binding. In this review, we discuss the molecular and immunological mechanisms by which persistent STAT3 activation promotes NSCLC development, and the utility of STAT3 as a prognostic and predictive biomarker in NSCLC. We also provide a comprehensive update of STAT3-targeting therapies that are currently undergoing clinical evaluation, and discuss the challenges associated with these treatment modalities in human patients.
Collapse
Affiliation(s)
- Sagun Parakh
- Department of Medical Oncology, The Olivia Newton-John Cancer and Wellness Centre, Austin Health, Heidelberg, VIC 3084, Australia;
- Tumor Targeting Laboratory, The Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC 3086, Australia;
| | - Matthias Ernst
- School of Cancer Medicine, La Trobe University, Melbourne, VIC 3086, Australia;
- Cancer and Inflammation Laboratory, The Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
| | - Ashleigh R. Poh
- School of Cancer Medicine, La Trobe University, Melbourne, VIC 3086, Australia;
- Cancer and Inflammation Laboratory, The Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
| |
Collapse
|
13
|
Xiong JW, Song SB, Xiong LM, Duan CH, Song Q, Yu DL, Zhang XQ. CircRPPH1 promotes cell proliferation, migration and invasion of non-small cell lung cancer (NSCLC) via the PI3K/AKT and JAK2/STAT3 signaling axes. J Biochem 2021; 171:245-252. [PMID: 34850004 DOI: 10.1093/jb/mvab129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 11/19/2021] [Indexed: 12/25/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) has markedly increased morbidity and mortality rate worldwide. Circular RNAs (circRNAs) were shown to regulate NSCLC progression. But, the underlying pathways of the circRPPH1-mediated regulation of NSCLC still need further exploration. We evaluated circRPPH1 levels in NSCLC tissues and cell lines via qRT-RCR. Moreover, using ectopic plasmid incorporation and siRNA assays, we analyzed the circRPPH1-mediated regulation of cell proliferation (CP), migration (CM), and invasion (CI) in NSCLC cell lines (H1975 and A549 cells), using CCK-8, colony forming, scratch wound, and transwell assays, respectively. CircRPPH1 levels were remarkably high in the NSCLC tissues and cell lines. The transfection experiments showed that circRPPH1 overexpression was able to promote CP, CM and CI of NSCLC cells, while CP, CM and CI were significantly restrained by the knockdown of circRPPH1. We also displayed that circRPPH1 knockdown suppressed the cell progression via inactivating the PI3K/AKT and JAK2/STAT3 signaling axes. Subsequently, in vivo experiment in nude mice was demonstrated that the inhibition of circRPPH1 could reduce the tumor growth of NSCLC. circRPPH1 may accelerate the growth and metastasis of NSCLC, in culture conditions and in animal models, by stimulating the PI3K/AKT and JAK2/STAT3 signaling axes, thus promoting the development of NSCLC.
Collapse
Affiliation(s)
- Jian-Wen Xiong
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Si-Bei Song
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Lin-Min Xiong
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Chuan-Hui Duan
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qian Song
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Dong-Liang Yu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xiao-Qiang Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| |
Collapse
|
14
|
Padda SK, Reckamp KL, Koczywas M, Neal JW, Kawashima J, Kong S, Huang DB, Kowalski M, Wakelee HA. A phase 1b study of erlotinib and momelotinib for the treatment of EGFR-mutated, tyrosine kinase inhibitor-naive metastatic non-small cell lung cancer. Cancer Chemother Pharmacol 2021; 89:105-115. [PMID: 34773474 PMCID: PMC8739290 DOI: 10.1007/s00280-021-04369-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/16/2021] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Preclinical evidence suggests the feedforward cytokine loop of interleukin-6/Janus kinases (JAK)/STAT3 plays a role in epidermal growth factor receptor tyrosine kinase inhibitor (EGFR TKI) resistance in EGFR-mutated non-small cell lung cancer (NSCLC). METHODS In this phase 1b study, the JAK1/2 and TANK-binding kinase 1 (TBK1) inhibitor momelotinib was evaluated in combination with erlotinib in patients with EGFR TKI-naive, EGFR-mutated NSCLC. After erlotinib lead-in (50, 75, 100, or 150 mg oral daily [QD]), momelotinib was combined and dose escalated in a 3 + 3 study design. The primary endpoint of maximum tolerated dose (MTD) of momelotinib was determined based on the incidence of dose-limiting toxicities (DLTs) during the first 28-day cycle. Secondary endpoints included efficacy and pharmacokinetics (PK). RESULTS Eleven patients were enrolled across 3 dose levels of momelotinib (100 mg QD, 200 mg QD, and 100 mg twice daily [BID]). The MTD was momelotinib 200 mg QD in combination with erlotinib. Two DLTs of grade 4 neutropenia without fever and grade 3 diarrhea occurred at momelotinib 100 mg BID. Most common treatment-emergent adverse events included diarrhea, dry skin, fatigue, and decreased appetite; the vast majority being grades 1-2. The overall response rate was 54.5% (90% CI 27.1-80.0; all partial) and median progression-free survival was 9.2 months (90% CI 6.2-12.4). Momelotinib did not affect the PK of erlotinib. CONCLUSIONS The JAK1/2 and TBK1 inhibitor momelotinib in combination with erlotinib did not appear to enhance benefit over the historical data of erlotinib monotherapy in patients with EGFR-mutated NSCLC. CLINICALTRIALS. GOV IDENTIFIER NCT02206763.
Collapse
Affiliation(s)
- Sukhmani K Padda
- Stanford University School of Medicine/Stanford Cancer Institute, Stanford, CA, USA. .,Cedars-Sinai Medical Center, 8700 Beverly Blvd, SCCT 1S31, Los Angeles, CA, 90048, USA.
| | - Karen L Reckamp
- Cedars-Sinai Medical Center, 8700 Beverly Blvd, SCCT 1S31, Los Angeles, CA, 90048, USA.,City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | - Joel W Neal
- Stanford University School of Medicine/Stanford Cancer Institute, Stanford, CA, USA
| | - Jun Kawashima
- Gilead Sciences, Inc., Foster City, CA, USA.,Sierra Oncology, Inc., Vancouver, BC, Canada
| | - Shengchun Kong
- Gilead Sciences, Inc., Foster City, CA, USA.,Genentech, Inc., South San Francisco, CA, USA
| | - Daniel B Huang
- The Oncology Institute of Hope and Innovation, Santa Ana, CA, USA
| | - Mark Kowalski
- Gilead Sciences, Inc., Foster City, CA, USA.,Sierra Oncology, Inc., Vancouver, BC, Canada
| | - Heather A Wakelee
- Stanford University School of Medicine/Stanford Cancer Institute, Stanford, CA, USA
| |
Collapse
|
15
|
Wang LL, Luo J, He ZH, Liu YQ, Li HG, Xie D, Cai MY. STEAP3 promotes cancer cell proliferation by facilitating nuclear trafficking of EGFR to enhance RAC1-ERK-STAT3 signaling in hepatocellular carcinoma. Cell Death Dis 2021; 12:1052. [PMID: 34741044 PMCID: PMC8571373 DOI: 10.1038/s41419-021-04329-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/30/2021] [Accepted: 10/13/2021] [Indexed: 12/24/2022]
Abstract
STEAP3 (Six-transmembrane epithelial antigen of the prostate 3, TSAP6, dudulin-2) has been reported to be involved in tumor progression in human malignancies. Nevertheless, how it participates in the progression of human cancers, especially HCC, is still unknown. In the present study, we found that STEAP3 was aberrantly overexpressed in the nuclei of HCC cells. In a large cohort of clinical HCC tissues, high expression level of nuclear STEAP3 was positively associated with tumor differentiation and poor prognosis (p < 0.001), and it was an independent prognostic factor for HCC patients. In HCC cell lines, nuclear expression of STEAP3 significantly promoted HCC cells proliferation by promoting stemness phenotype and cell cycle progression via RAC1-ERK-STAT3 and RAC1-JNK-STAT6 signaling axes. Through upregulating the expression and nuclear trafficking of EGFR, STEAP3 participated in regulating EGFR-mediated STAT3 transactivity in a manner of positive feedback. In summary, our findings support that nuclear expression of STEAP3 plays a critical oncogenic role in the progression of HCC via modulation on EGFR and intracellular signaling, and it could be a candidate for prognostic marker and therapeutic target in HCC.
Collapse
MESH Headings
- Animals
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Cycle/genetics
- Cell Cycle Proteins/metabolism
- Cell Line, Tumor
- Cell Nucleus/metabolism
- Cell Proliferation
- Disease Progression
- ErbB Receptors/metabolism
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- MAP Kinase Signaling System
- Male
- Mice, Inbred BALB C
- Mice, Nude
- Middle Aged
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Oxidoreductases/metabolism
- Phosphorylation
- Prognosis
- Protein Transport
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- STAT3 Transcription Factor/metabolism
- Signal Transduction
- Spheroids, Cellular/metabolism
- Spheroids, Cellular/pathology
- Treatment Outcome
- rac1 GTP-Binding Protein/metabolism
- Mice
Collapse
Affiliation(s)
- Li-Li Wang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jie Luo
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Zhang-Hai He
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Ye-Qing Liu
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hai-Gang Li
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Dan Xie
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China.
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Mu-Yan Cai
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China.
| |
Collapse
|
16
|
Circulating Tumour Cells (CTCs) in NSCLC: From Prognosis to Therapy Design. Pharmaceutics 2021; 13:pharmaceutics13111879. [PMID: 34834295 PMCID: PMC8619417 DOI: 10.3390/pharmaceutics13111879] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/27/2021] [Accepted: 10/30/2021] [Indexed: 02/08/2023] Open
Abstract
Designing optimal (neo)adjuvant therapy is a crucial aspect of the treatment of non-small-cell lung carcinoma (NSCLC). Standard methods of chemotherapy, radiotherapy, and immunotherapy represent effective strategies for treatment. However, in some cases with high metastatic activity and high levels of circulating tumour cells (CTCs), the efficacy of standard treatment methods is insufficient and results in treatment failure and reduced patient survival. CTCs are seen not only as an isolated phenomenon but also a key inherent part of the formation of metastasis and a key factor in cancer death. This review discusses the impact of NSCLC therapy strategies based on a meta-analysis of clinical studies. In addition, possible therapeutic strategies for repression when standard methods fail, such as the administration of low-toxicity natural anticancer agents targeting these phenomena (curcumin and flavonoids), are also discussed. These strategies are presented in the context of key mechanisms of tumour biology with a strong influence on CTC spread and metastasis (mechanisms related to tumour-associated and -infiltrating cells, epithelial–mesenchymal transition, and migration of cancer cells).
Collapse
|
17
|
Cong L, Yi J, Qiu S, Wang R, Jin S, Jiang R, Cong X. Effect of EG00229 on Radiation Resistance of Lung Adenocarcinoma Cells. J Cancer 2021; 12:6105-6117. [PMID: 34539883 PMCID: PMC8425201 DOI: 10.7150/jca.56123] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 08/08/2021] [Indexed: 12/25/2022] Open
Abstract
Background: Neuropilin 1 (NRP1) is a pleiotropic receptor that interacts with multiple ligands and their receptors and plays a critical role in the process of tumor metastasis and radiation resistance in endothelial cells and tumor cells. In this study, we sought to investigate the mechanistic role of NRP1 in the radiation resistance of non-small cell lung cancer (NSCLC) cells and the role of EG00229 (an inhibitor of NRP1) on reversing radiation resistance. Materials and Methods: A549 and H1299 NSCLC cells were used to construct radiation resistance models. Western blot, ELISA, and qRT-PCR were used to detect protein and mRNA levels of NRP1, epithelial-mesenchymal transition (EMT) markers, and molecules in signaling pathways. Immunofluorescence was used to measure changes in co-expression of NRP1 and VEGF-165 in radiation-resistant model cells. An immunoprecipitation assay was used to detect the binding capacity of NRP1 and VEGF-165. Results: We successfully created two radiation resistant models (A549RR and H1299-RR). The expression levels of NRP1, EMT-related proteins, and proteins in metastasis-related pathways were increased in NSCLC cells with radiation resistance. After adding EG00229, the expression levels and binding capacity of NRP1 and VEGF-165 proteins were significantly reduced. The expression of EMT-related proteins and proteins in metastasis-related pathways were reduced in NSCLC cells with radiation resistance. Conclusion: Our data provide an insight into the molecular mechanisms of radiation resistance and suggest that EG00229 may contribute to reversing the radiation resistance of NSCLC cells by inhibiting the binding of NRP1 and VEGF-165. Our findings could provide a novel theoretical and experimental foundation for improving the efficacy of lung cancer radiotherapy.
Collapse
Affiliation(s)
- Lele Cong
- Department of Dermatology, China-Japan Union Hospital, Jilin University, Changchun 130033, Jilin, China
| | - Junxuan Yi
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Shuang Qiu
- Tissue Bank, China-Japan Union Hospital, Jilin University, Changchun 130033, Jilin, China
| | - Rui Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Shunzi Jin
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Rihua Jiang
- Department of Dermatology, China-Japan Union Hospital, Jilin University, Changchun 130033, Jilin, China
| | - Xianling Cong
- Tissue Bank, China-Japan Union Hospital, Jilin University, Changchun 130033, Jilin, China
| |
Collapse
|
18
|
Wang D, Li Y, Chen X, Li P. Prognostic significance of volume-based 18F-FDG PET/CT parameters and correlation with PD-L1 expression in patients with surgically resected lung adenocarcinoma. Medicine (Baltimore) 2021; 100:e27100. [PMID: 34477147 PMCID: PMC8415941 DOI: 10.1097/md.0000000000027100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 08/11/2021] [Indexed: 12/09/2022] Open
Abstract
The aim of this study was to retrospectively analyze 18F-FDG positron emission tomography/computed tomography (18F-FDG PET/CT) metabolic variables, programmed death-ligand 1 (PD-L1) and phosphorylated signal transducer and activator of transcription 3 (p-STAT3) tumor expression, and other factors as predictors of disease-free survival (DFS) in patients with lung adenocarcinoma (LUAD) (stage IA-IIIA) who underwent surgical resection. We still lack predictor of immune checkpoint (programmed cell death-1 [PD-1]/PD-L1) inhibitors. Herein, we investigated the correlation between metabolic parameters from 18F-FDG PET/CT and PD-L1 expression in patients with surgically resected LUAD.Seventy-four patients who underwent 18F-FDG PET/CT prior to treatment were consecutively enrolled. The main 18F-FDG PET/CT-derived variables were primary tumor maximum standardized uptake value (SUVmax), metabolic tumor volume (MTV), and total lesion glycolysis (TLG). Surgical tumor specimens were analyzed for PD-L1 and p-STAT3 expression using immunohistochemistry. Correlations between immunohistochemistry results and 18F-FDG PET/CT-derived variables were compared. Associations of PD-L1 and p-STAT3 tumor expression, 18F-FDG PET/CT-derived variables, and other factors with DFS in resected LUAD were evaluated.All tumors were FDG-avid. The cutoff values of low and high SUVmax, MTV, and TLG were 12.60, 14.87, and 90.85, respectively. The results indicated that TNM stage, PD-L1 positivity, and high 18F-FDG PET/CT metabolic volume parameters (TLG ≥90.85 or MTV ≥14.87) were independent predictors of worse DFS in resected LUAD. No 18F-FDG metabolic parameters associated with PD-L1 expression were observed (chi-square test), but we found that patients with positive PD-L1 expression have significantly higher SUVmax (P = .01), MTV (P = .00), and TLG (P = .00) than patients with negative PD-L1 expression.18F-FDG PET/CT metabolic volume parameters (TLG ≥90.85 or MTV ≥14.87) were more helpful in prognostication than the conventional parameter (SUVmax), PD-L1 expression was an independent predictor of DFS in patients with resected LUAD. Metabolic parameters on 18F-FDG PET/CT have a potential role for 18F-FDG PET/CT in selecting candidate LUAD for treatment with checkpoint inhibitors.
Collapse
Affiliation(s)
- Dalong Wang
- Department of PET/CT, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yingci Li
- Department of PET/CT, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Xiaolin Chen
- Department of PET/CT, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ping Li
- Department of PET/CT, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
19
|
Lu Q, Ni Y, Wang W, Wang L, Jiang T, Shang L. Dynamin 3 Inhibits the Proliferation of Non-small-Cell Lung Cancer Cells by Suppressing c-MET-GBR2-STAT3 Complex Formation. Front Cell Dev Biol 2021; 9:641403. [PMID: 34490234 PMCID: PMC8416685 DOI: 10.3389/fcell.2021.641403] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 07/20/2021] [Indexed: 01/05/2023] Open
Abstract
Dynamin 3 (DNM3) has gained increased attention ever since its potential as a tumor suppressor was reported. However, its action in lung cancer (LC) is undefined. In this study, the role of DNM3 in LC development was investigated. DNM3 expression was found to be downregulated in tumors of patients with LC, especially those with metastasis. The DNM3 downregulation enhanced the proliferative and metastatic ability of LC cells, whereas its upregulation had the opposite effects. In vivo xenograft experiments confirmed that lung tumors with lower DNM3 expression had higher growth and metastatic abilities. Mechanistic studies revealed that DNM3 interacts with growth factor receptor-bound protein 2 (GBR2), thereby interrupting tyrosine-protein kinase Met (c-MET)-GBR2-signal transducer and activator of transcription 3 (STAT3) complex formation, which suppressed STAT3 activation. Therefore, the absence of DNM3 frees GBR2 to activate STAT3, which regulates the expression of genes related to LC proliferation and metastasis (e.g., cyclin D1 and Snail family transcriptional repressor 1). Additionally, the c-MET inhibitor crizotinib effectively suppressed LC cell proliferation and migration in vitro and in vivo, even with DNM3 depleted. Therefore, our study has demonstrated the antitumor effect of DNM3 in LC and suggests that the inhibition of c-MET might be a promising strategy for treating those LC patients with low DNM3 expression.
Collapse
Affiliation(s)
- Qiang Lu
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
| | - Yunfeng Ni
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
| | - Wuping Wang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
| | - Lei Wang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
| | - Tao Jiang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
| | - Lei Shang
- The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Department of Health Statistics, School of Public Health, The Air Force Military Medical University, Xi’an, China
| |
Collapse
|
20
|
Mohankumar K, Francis AP, Pajaniradje S, Rajagopalan R. Synthetic curcumin analog: inhibiting the invasion, angiogenesis, and metastasis in human laryngeal carcinoma cells via NF-kB pathway. Mol Biol Rep 2021; 48:6065-6074. [PMID: 34355287 DOI: 10.1007/s11033-021-06610-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/29/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND Laryngeal carcinoma, the most common among head and neck squamous cell carcinoma (HNSCC), induces 1% of all cancer deaths. Curcumin the active constituent of turmeric, is shown to be effective in the treatment of various cancers. In the present study, we explored the mechanistic role of bis-demethoxy curcumin analog (BDMC-A) as a chemotherapeutic agent. We investigated its inhibitory effect on invasion, angiogenesis, and metastasis in human laryngeal carcinoma (Hep-2) cells in comparison with curcumin. METHODS The effect of curcumin and BDMC-A on transcription factors (NF-κB, p65, c-Jun, c-Fos, STAT3, 5, PPAR-γ, β-catenin, COX-2, MMP-9, VEGF, TIMP-2) involved in signal transduction cascade, invasion, and angiogenesis in Hep-2 cells were quantified using Western blotting and RT-PCR technique. ELISA was used to measure the pro-inflammatory markers in Hep-2 cells treated with curcumin and BDMC-A. RESULTS The results showed that BDMC-A inhibits the transcription factors NF-κB, p65, c-Jun, c-Fos, STAT3, STAT5, PPAR-γ and β-catenin, which are responsible for tumor progression and malignancy. Moreover, BDMC-A treatment downregulated MMP-9, VEGF, TGF- β, IL-6 and IL-8 and upregulated TIMP-2 levels. The effects were more significant compared to curcumin. CONCLUSION Our overall results revealed that BDMC-A more effectively inhibited the markers of invasion, angiogenesis and metastasis in comparison with curcumin.
Collapse
Affiliation(s)
- Kumaravel Mohankumar
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, 605 014, India
| | - Arul Prakash Francis
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, 605 014, India
| | - Sankar Pajaniradje
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, 605 014, India
| | - Rukkumani Rajagopalan
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, 605 014, India.
| |
Collapse
|
21
|
Feng X, Ding W, Ma J, Liu B, Yuan H. Targeted Therapies in Lung Cancers: Current Landscape and Future Prospects. Recent Pat Anticancer Drug Discov 2021; 16:540-551. [PMID: 34132185 DOI: 10.2174/1574892816666210615161501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/09/2021] [Accepted: 03/31/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Lung cancer is the most common and malignant cancer worldwide. Targeted therapies have emerged as a promising treatment strategy for lung cancers. OBJECTIVE The objective of this study is to evaluate the current landscape of targets and finding promising targets for future new drug discovery for lung cancers by identifying the science-technology-clinical development pattern and mapping the interaction network of targets. METHODS Targets for cancers were classified into 3 groups based on a paper published in Nature. We search for scientific literature, patent documents and clinical trials of targets in Group 1 and Group 2 for lung cancers. Then, a target-target interaction network of Group 1 was constructed, and the science-technology-clinical(S-T-C) development patterns of targets in Group 1 were identified. Finally, based on the cluster distribution and the development pattern of targets in Group 1, interactions between the targets were employed to predict potential targets in Group 2 on drug development. RESULTS The target-target interaction(TTI)network of group 1 resulted in 3 clusters with different developmental stages. The potential targets in Group 2 are divided into 3 ranks. Level-1 is the first priority and level-3 is the last. Level-1 includes 16 targets, such as STAT3, CRKL, and PTPN11, that are mostly involved in signaling transduction pathways. Level-2 and level-3 contain 8 and 6 targets related to various biological functions. CONCLUSION This study will provide references for drug development in lung cancers, emphasizing that priorities should be given to targets in Level-1, whose mechanisms are worth further exploration.
Collapse
Affiliation(s)
- Xin Feng
- School of Business Administration, Shenyang Pharmaceutical University, Shenyang, China
| | - Wenqing Ding
- School of Business Administration, Shenyang Pharmaceutical University, Shenyang, China
| | - Junhong Ma
- School of Business Administration, Shenyang Pharmaceutical University, Shenyang, China
| | - Baijun Liu
- School of Business Administration, Shenyang Pharmaceutical University, Shenyang, China
| | - Hongmei Yuan
- School of Business Administration, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
22
|
Kortekaas RK, Burgess JK, van Orsoy R, Lamb D, Webster M, Gosens R. Therapeutic Targeting of IL-11 for Chronic Lung Disease. Trends Pharmacol Sci 2021; 42:354-366. [PMID: 33612289 DOI: 10.1016/j.tips.2021.01.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/11/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023]
Abstract
Interleukin (IL)-11 was originally recognized as an immunomodulatory and hematopoiesis-inducing cytokine. However, although IL-11 is typically not found in healthy individuals, it is now becoming evident that IL-11 may play a role in diverse pulmonary conditions, including IPF, asthma, and lung cancer. Additionally, experimental strategies targeting IL-11, such as humanized antibodies, have recently been developed, revealing the therapeutic potential of IL-11. Thus, further insight into the underlying mechanisms of IL-11 in lung disease may lead to the ability to interfere with pathological conditions that have a clear need for disease-modifying treatments, such as IPF. In this review, we outline the effects, expression, signaling, and crosstalk of IL-11 and focus on its role in lung disease and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Rosa K Kortekaas
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Janette K Burgess
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Medical Biology and Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Roël van Orsoy
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - David Lamb
- Department of Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Megan Webster
- Department of Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
23
|
Tolomeo M, Cascio A. The Multifaced Role of STAT3 in Cancer and Its Implication for Anticancer Therapy. Int J Mol Sci 2021; 22:ijms22020603. [PMID: 33435349 PMCID: PMC7826746 DOI: 10.3390/ijms22020603] [Citation(s) in RCA: 191] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/24/2020] [Accepted: 01/05/2021] [Indexed: 12/12/2022] Open
Abstract
Signal transducer and activator of transcription (STAT) 3 is one of the most complex regulators of transcription. Constitutive activation of STAT3 has been reported in many types of tumors and depends on mechanisms such as hyperactivation of receptors for pro-oncogenic cytokines and growth factors, loss of negative regulation, and excessive cytokine stimulation. In contrast, somatic STAT3 mutations are less frequent in cancer. Several oncogenic targets of STAT3 have been recently identified such as c-myc, c-Jun, PLK-1, Pim1/2, Bcl-2, VEGF, bFGF, and Cten, and inhibitors of STAT3 have been developed for cancer prevention and treatment. However, despite the oncogenic role of STAT3 having been widely demonstrated, an increasing amount of data indicate that STAT3 functions are multifaced and not easy to classify. In fact, the specific cellular role of STAT3 seems to be determined by the integration of multiple signals, by the oncogenic environment, and by the alternative splicing into two distinct isoforms, STAT3α and STAT3β. On the basis of these different conditions, STAT3 can act both as a potent tumor promoter or tumor suppressor factor. This implies that the therapies based on STAT3 modulators should be performed considering the pleiotropic functions of this transcription factor and tailored to the specific tumor type.
Collapse
|
24
|
Yang PL, Liu LX, Li EM, Xu LY. STAT3, the Challenge for Chemotherapeutic and Radiotherapeutic Efficacy. Cancers (Basel) 2020; 12:cancers12092459. [PMID: 32872659 PMCID: PMC7564975 DOI: 10.3390/cancers12092459] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/25/2020] [Accepted: 08/27/2020] [Indexed: 02/05/2023] Open
Abstract
Chemoradiotherapy is one of the most effective and extensively used strategies for cancer treatment. Signal transducer and activator of transcription 3 (STAT3) regulates vital biological processes, such as cell proliferation and cell growth. It is constitutively activated in various cancers and limits the application of chemoradiotherapy. Accumulating evidence suggests that STAT3 regulates resistance to chemotherapy and radiotherapy and thereby impairs therapeutic efficacy by mediating its feedback loop and several target genes. The alternative splicing product STAT3β is often identified as a dominant-negative regulator, but it enhances sensitivity to chemotherapy and offers a new and challenging approach to reverse therapeutic resistance. We focus here on exploring the role of STAT3 in resistance to receptor tyrosine kinase (RTK) inhibitors and radiotherapy, outlining the potential of targeting STAT3 to overcome chemo(radio)resistance for improving clinical outcomes, and evaluating the importance of STAT3β as a potential therapeutic approach to overcomes chemo(radio)resistance. In this review, we discuss some new insights into the effect of STAT3 and its subtype STAT3β on chemoradiotherapy sensitivity, and we explore how these insights influence clinical treatment and drug development for cancer.
Collapse
Affiliation(s)
- Ping-Lian Yang
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, China; (P.-L.Y.); (L.-X.L.)
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Lu-Xin Liu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, China; (P.-L.Y.); (L.-X.L.)
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, China; (P.-L.Y.); (L.-X.L.)
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
- Correspondence: (E.-M.L.); (L.-Y.X.); Tel.: +86-754-88900460 (L.-Y.X.); Fax: +86-754-88900847 (L.-Y.X.)
| | - Li-Yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, China; (P.-L.Y.); (L.-X.L.)
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, China
- Correspondence: (E.-M.L.); (L.-Y.X.); Tel.: +86-754-88900460 (L.-Y.X.); Fax: +86-754-88900847 (L.-Y.X.)
| |
Collapse
|
25
|
Yuan Y, Liu L, Wang Y, Liu S. Reduced expression of CYLD promotes cell survival and inflammation in gefitinib-treated NSCLC PC-9 cells: Targeting CYLD may be beneficial for acquired resistance to gefitinib therapy. Cell Biol Int 2020; 44:1911-1918. [PMID: 32441799 DOI: 10.1002/cbin.11397] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 05/08/2020] [Accepted: 05/18/2020] [Indexed: 12/15/2022]
Abstract
The application of tyrosine kinase inhibitors (TKIs) to the epidermal growth factor receptor (EGFR) has been proven to be highly effective for non-small-cell lung cancer (NSCLC). However, patients often evolve into acquired resistance. The secondary mutations in EGFR account for nearly half of the acquired resistance. While the remaining 50% of patients exhibit tolerance to EGFR-TKIs with unclear mechanism(s). Cylindromatosis (CYLD), a deubiquitinase, functions as a tumor suppressor to regulate cell apoptosis, proliferation, and immune response, and so on. The role of CYLD in NSCLC EGFR-TKI resistance remains elusive. Here, we found CYLD was upregulated in PC-9 cells, whereas downregulated in PC-9 acquired gefitinib-resistant (PC-9/GR) cells in response to the treatment of gefitinib, which is consistent with the results in the Gene Expression Omnibus database. Overexpression of CYLD promoted a more apoptotic death ratio in PC-9/GR cells than that in PC-9 cells. In addition, silencing the expression of CYLD resulted in an increase of the expression level of interleukin-6, transforming growth factor-β and tumor necrosis factor-α, which may contribute to acquired resistance of PC-9 cells to gefitinib. Taken together, our data in vitro demonstrate that PC-9/GR cells downregulated CYLD expression, enhanced subsequent CYLD-dependent antiapoptotic capacity and inflammatory response, which may provide a possible target for acquired gefitinib-resistant treatment in NSCLC.
Collapse
Affiliation(s)
- Yuan Yuan
- Central Laboratory of Binhu Hospital, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Liu Liu
- Department of Oncology, Binhu Hospital, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yi Wang
- Department of Oncology, Binhu Hospital, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Shangquan Liu
- Central Laboratory of Binhu Hospital, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
26
|
STAT3: Versatile Functions in Non-Small Cell Lung Cancer. Cancers (Basel) 2020; 12:cancers12051107. [PMID: 32365499 PMCID: PMC7281271 DOI: 10.3390/cancers12051107] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 12/22/2022] Open
Abstract
Signal Transducer and Activator of Transcription 3 (STAT3) activation is frequently found in non-small cell lung cancer (NSCLC) patient samples/cell lines and STAT3 inhibition in NSCLC cell lines markedly impairs their survival. STAT3 also plays a pivotal role in driving tumor-promoting inflammation and evasion of anti-tumor immunity. Consequently, targeting STAT3 either directly or by inhibition of upstream regulators such as Interleukin-6 (IL-6) or Janus kinase 1/2 (JAK1/2) is considered as a promising treatment strategy for the management of NSCLC. In contrast, some studies also report STAT3 being a tumor suppressor in a variety of solid malignancies, including lung cancer. Here, we provide a concise overview of STAT3‘s versatile roles in NSCLC and discuss the yins and yangs of STAT3 targeting therapies.
Collapse
|
27
|
Mohan CD, Rangappa S, Preetham HD, Chandra Nayaka S, Gupta VK, Basappa S, Sethi G, Rangappa KS. Targeting STAT3 signaling pathway in cancer by agents derived from Mother Nature. Semin Cancer Biol 2020; 80:157-182. [DOI: 10.1016/j.semcancer.2020.03.016] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/23/2020] [Accepted: 03/28/2020] [Indexed: 02/07/2023]
|
28
|
Ke W, Zhang L, Dai Y. The role of IL-6 in immunotherapy of non-small cell lung cancer (NSCLC) with immune-related adverse events (irAEs). Thorac Cancer 2020; 11:835-839. [PMID: 32043828 PMCID: PMC7113041 DOI: 10.1111/1759-7714.13341] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/10/2020] [Accepted: 01/12/2020] [Indexed: 12/14/2022] Open
Abstract
IL-6 is a cytokine that plays an important role in response to injury or infection and is a promising biomarker for predicting poor prognosis and therapeutic targets in non-small cell lung cancer (NSCLC). This article reviews the biochemical mechanism, function and genotype of IL-6, and summarizes the diagnostic and prognostic value of IL-6 level. Anti-IL-6 therapy does not affect the effect of immunocheckpoint inhibitors (ICIs), but enhances its anticancer function, which may be the treatment option for immune-related adverse events (irAEs) in the future. Therefore, IL-6 may be a therapeutic target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Wanhai Ke
- Arrhythmia Center, Fuwai Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Zhang
- Respiratory Medicine Department, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yan Dai
- Arrhythmia Center, Fuwai Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
29
|
Prince GMSH, Yang TY, Lin H, Chen MC. Mechanistic insight of cyclin-dependent kinase 5 in modulating lung cancer growth. CHINESE J PHYSIOL 2019; 62:231-240. [PMID: 31793458 DOI: 10.4103/cjp.cjp_67_19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Lung harbors the growth of primary and secondary tumors. Even though numerous factors regulate the complex signal transduction and cytoskeletal remodeling toward the progression of lung cancer, cyclin-dependent kinase 5 (Cdk5), a previously known kinase in the central nervous system, has raised much attention in the recent years. Patients with aberrant Cdk5 expression also lead to poor survival. Cdk5 has already been employed in various cellular processes which shape the fate of cancer. In lung cancer, Cdk5 mainly regulates tumor suppressor genes, carcinogenesis, cytoskeletal remodeling, and immune checkpoints. Inhibiting Cdk5 by using drugs, siRNA or CRISP-Cas9 system has rendered crucial therapeutic advantage in the combat against lung cancer. Thus, the relation of Cdk5 to lung cancer needs to be addressed in detail. In this review, we will discuss various cellular events modulated by Cdk5 and we will go further into their underlying mechanism in lung cancer.
Collapse
Affiliation(s)
| | - Tsung-Ying Yang
- Department of Internal Medicine, Division of Chest Medicine, Taichung Veterans General Hospital, Taichung; Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ho Lin
- Department of Life Sciences; Program in Translational Medicine and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Mei-Chih Chen
- Department of Nursing, Asia University; Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
30
|
Han L, Yao S, Cao S, Mo G, Li J, Cao Y, Huang F. Triterpenoid Saponins from Anemone flaccida Suppress Tumor Cell Proliferation by Regulating MAPK, PD1/PDL1, and STAT3 Signaling Pathways and Altering Cancer Metabolism. Onco Targets Ther 2019; 12:10917-10930. [PMID: 31849495 PMCID: PMC6913295 DOI: 10.2147/ott.s212666] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 11/26/2019] [Indexed: 12/18/2022] Open
Abstract
Purpose Natural triterpenoid saponins isolated from Anemone flaccida Fr. Schmidt have exhibited anti-cancer properties and exerted remarkable inhibitory effects on tumor growth. Herein, we investigated the potential mechanism involved in the suppression of hepatocellular carcinoma (HCC) development by triterpenoid saponins in a mouse model. Methods An HCC model was established in H22 tumor-bearing mice and triterpenoid saponins were administered at various doses. Immunofluorescence, flow cytometry, and western blot were performed to analyze the effect of triterpenoid saponins on immune response in tumor tissues. Metabolomic analysis was carried out to assess the metabolites involved in mediating the effect of triterpenoid saponins on tumor tissues. Results Triterpenoid saponins induced anti-tumor immune response by decreasing the number of Treg cells, increasing that of B cells, natural killer cells, and CD3+/CD28+ T cells, and reducing the secretion of inflammatory factors including nuclear factor-κB, cyclooxygenase-2, and microsomal prostaglandin E synthase-1. In addition, triterpenoid saponins inhibited tumor growth and induced the apoptosis of HCC cells by blocking the activation of PD1/PD-L1, ERK1/2, p38 MAPK, JNK, and STAT3 signaling pathways. Furthermore, triterpenoid saponins regulated tumor immune response by upregulating a number of metabolites (including 1,3-diaminopropane, lauric acid, 2,4-diaminobutyric acid 2, and ribitol) and modulating the metabolism of histidine, arginine, proline, beta-alanine, glycine, serine, and threonine. Conclusion The findings suggested that triterpenoid saponins interfered with multiple signaling cascades involved in tumorigenesis and tumor metabolism and have potential applications in HCC therapy.
Collapse
Affiliation(s)
- Lintao Han
- Key Laboratory of Traditional Chinese Medicine Resource and Prescription, Ministry of Education, Wuhan, Hubei 430061, People's Republic of China
| | - Shiqi Yao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, People's Republic of China
| | - Sa Cao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, People's Republic of China
| | - Guoyan Mo
- Key Laboratory of Traditional Chinese Medicine Resource and Prescription, Ministry of Education, Wuhan, Hubei 430061, People's Republic of China
| | - Jingjing Li
- Key Laboratory of Traditional Chinese Medicine Resource and Prescription, Ministry of Education, Wuhan, Hubei 430061, People's Republic of China
| | - Yan Cao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, People's Republic of China
| | - Fang Huang
- Key Laboratory of Traditional Chinese Medicine Resource and Prescription, Ministry of Education, Wuhan, Hubei 430061, People's Republic of China
| |
Collapse
|
31
|
Wan Mohd Tajuddin WNB, Lajis NH, Abas F, Othman I, Naidu R. Mechanistic Understanding of Curcumin's Therapeutic Effects in Lung Cancer. Nutrients 2019; 11:E2989. [PMID: 31817718 PMCID: PMC6950067 DOI: 10.3390/nu11122989] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/22/2019] [Accepted: 11/30/2019] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is among the most common cancers with a high mortality rate worldwide. Despite the significant advances in diagnostic and therapeutic approaches, lung cancer prognoses and survival rates remain poor due to late diagnosis, drug resistance, and adverse effects. Therefore, new intervention therapies, such as the use of natural compounds with decreased toxicities, have been considered in lung cancer therapy. Curcumin, a natural occurring polyphenol derived from turmeric (Curcuma longa) has been studied extensively in recent years for its therapeutic effects. It has been shown that curcumin demonstrates anti-cancer effects in lung cancer through various mechanisms, including inhibition of cell proliferation, invasion, and metastasis, induction of apoptosis, epigenetic alterations, and regulation of microRNA expression. Several in vitro and in vivo studies have shown that these mechanisms are modulated by multiple molecular targets such as STAT3, EGFR, FOXO3a, TGF-β, eIF2α, COX-2, Bcl-2, PI3KAkt/mTOR, ROS, Fas/FasL, Cdc42, E-cadherin, MMPs, and adiponectin. In addition, limitations, strategies to overcome curcumin bioavailability, and potential side effects as well as clinical trials were also reviewed.
Collapse
Affiliation(s)
- Wan Nur Baitty Wan Mohd Tajuddin
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; (W.N.B.W.M.T.); (I.O.)
| | - Nordin H. Lajis
- Laboratory of Natural Products, Faculty of Science, Universiti Putra Malaysia, UPM, Serdang 43400, Malaysia; (N.H.L.); (F.A.)
| | - Faridah Abas
- Laboratory of Natural Products, Faculty of Science, Universiti Putra Malaysia, UPM, Serdang 43400, Malaysia; (N.H.L.); (F.A.)
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, UPM, Serdang 43400, Malaysia
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; (W.N.B.W.M.T.); (I.O.)
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; (W.N.B.W.M.T.); (I.O.)
| |
Collapse
|
32
|
He H, Zhuo R, Dai J, Wang X, Huang X, Wang H, Xu D. Chelerythrine induces apoptosis via ROS-mediated endoplasmic reticulum stress and STAT3 pathways in human renal cell carcinoma. J Cell Mol Med 2019; 24:50-60. [PMID: 31568643 PMCID: PMC6933352 DOI: 10.1111/jcmm.14295] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/21/2019] [Accepted: 03/07/2019] [Indexed: 12/15/2022] Open
Abstract
Renal cell carcinoma (RCC) is a heterogeneous histological disease and it is one of the most common kidney cancer. The treatment of RCC has been improved for the past few years, but its mortality still remains high. Chelerythrine (CHE) is a natural benzo[c]phenanthridine alkaloid and a widely used broad‐range protein kinase C inhibitor which has anti‐cancer effect on various types of human cancer cells. However, its effect on RCC has not been fully elucidated. In this study, we evaluated the effect and mechanism of CHE on RCC cells. Our study showed that CHE induced colony formation inhibition and G2/M cell cycle arrest in a dose‐dependent manner in RCC cells. In addition, CHE increased cellular ROS level, leading to endoplasmic reticulum (ER) stress, inactivating STAT3 activities and inducing apoptosis in RCC cells which were suppressed by NAC, a special ROS inhibitor. We further found that both knockdown of ATF4 protein and overexpression of STAT3 protein could reduce CHE‐induced apoptosis in Caki cells. These results demonstrated that the apoptosis induced by CHE was mediated by ROS‐caused ER stress and STAT3 inactivation. Collectively, our studies provided support for CHE as a potential new therapeutic agent for the management of RCC.
Collapse
Affiliation(s)
- Hongchao He
- Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ran Zhuo
- Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jun Dai
- Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiaojing Wang
- Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xin Huang
- Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Haofei Wang
- Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Danfeng Xu
- Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
33
|
Amin Marashi F, Torabi A, Beaudry F. Granulosa cells exposed to fibroblast growth factor 8 and 18 reveal early onset of cell growth and survival. Int J Reprod Biomed 2019; 17:435-442. [PMID: 31508568 PMCID: PMC6719512 DOI: 10.18502/ijrm.v17i6.4815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 08/29/2018] [Accepted: 10/17/2018] [Indexed: 11/24/2022] Open
Abstract
Background Fibroblast growth factors (FGFs) are growth factors that have diverse biological activities including broad mitogenic and cell survival activities. They function through the activation of a specific tyrosine kinase receptor that transduces the signal by activating several intracellular signaling pathways. Objective To identify the different signaling pathways involved in the mechanism of action of FGF8 and FGF18 on ovine granulosa cells using mass spectrometry. Materials and Methods Ovine ovarian granulosa cells were harvested from adult sheep independently at the stage of the estrous cycle and were cultured at a density of 500,000 viable cells in 1 ml DMEM/F12 medium for five days. The cells were then treated on day 5 of culture with 10 ng/mL FGF8 and FGF18 for 30 minutes, and total cell protein was collected for mass spectrometry. Results Mass spectrometry showed that both FGF8 and FGF18 significantly induce simultaneous upregulation of several proteins, including ATF1, STAT3, MAPK1, MAPK3, MAPK14, PLCG1, PLCG2, PKCA, PIK3CA, RAF1, GAB1, and BAG2 (> 1.5-fold; p < 0.01).
Conclusion ATF1 and STAT3 are important transcription factors involved in cell growth, proliferation and survival, and consequently can hamper or rescue the normal ovine reproductive system function.
Collapse
Affiliation(s)
- Fatemeh Amin Marashi
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, Québec, Canada J2S 7C6
| | - Ali Torabi
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, Québec, Canada J2S 7C6.,Groupe de Recherche en Pharmacologie Animale du Québec (GREPAQ), Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, Québec, Canada
| | - Francis Beaudry
- Groupe de Recherche en Pharmacologie Animale du Québec (GREPAQ), Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, Québec, Canada
| |
Collapse
|
34
|
Zhu D, Yu Y, Wang W, Wu K, Liu D, Yang Y, Zhang C, Qi Y, Zhao S. Long noncoding RNA PART1 promotes progression of non-small cell lung cancer cells via JAK-STAT signaling pathway. Cancer Med 2019; 8:6064-6081. [PMID: 31436388 PMCID: PMC6792487 DOI: 10.1002/cam4.2494] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/09/2019] [Accepted: 07/31/2019] [Indexed: 12/22/2022] Open
Abstract
Non‐small cell lung cancer (NSCLC), the major type of lung cancer, becomes the greatest threat to the life of people. Growing evidence shows prostate androgen‐regulated transcript 1 (PART1) is considered as effective markers for prostate cancer, and has been shown to be associated with poor prognosis of NSCLC. However, the tumorigenic mechanism of PART1 in NSCLC remains to be investigated. In this study, we found that the expression of PART1 was robustly induced in NSCLC tissues and cell lines. Functional studies established that overexpression of PART1 could promote NSCLC cell proliferation, migration, and invasion, while interference of PART1 inhibited NSCLC progression. Our results also identified miR‐635 as a novel target of PART1, whose expression was inhibited by PART1 in NSCLC cell lines. Moreover, gain‐ and loss‐of‐function studies revealed that PART1 could sponge miR‐635 and increase the expression of Janus kinase (JAK) and signal transducer and activator of transcription proteins (STATs). Finally, we deciphered the molecular mechanism by which PART1 contributed to promotion of NSCLC cell progression via phosphorylation and activation of JAK‐STAT signaling pathway. The animal experiment further confirmed that interference of NSCLC could suppress in vivo tumorigenic ability of NSCLC with favorable pharmacological activity via inactivation of JAK‐STAT signaling pathway. In conclusion, our findings clarified the biologic significance of PART1/miR‐635/JAK‐STAT axis in NSCLC progression and provided novel evidence that PART1 may be a new potential therapeutic target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Dengyan Zhu
- Department of Thoracic Surgery, The First Affiliate Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Yu
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Wang
- Department of Thoracic Surgery, Henan Medical Association, Zhengzhou, China
| | - Kai Wu
- Department of Thoracic Surgery, The First Affiliate Hospital of Zhengzhou University, Zhengzhou, China
| | - Donglei Liu
- Department of Thoracic Surgery, The First Affiliate Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Yang
- Department of Thoracic Surgery, The First Affiliate Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunyang Zhang
- Department of Thoracic Surgery, The First Affiliate Hospital of Zhengzhou University, Zhengzhou, China
| | - Yu Qi
- Department of Thoracic Surgery, The First Affiliate Hospital of Zhengzhou University, Zhengzhou, China
| | - Song Zhao
- Department of Thoracic Surgery, The First Affiliate Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
35
|
Xiao X, Wang S, Long G. C-reactive protein is a significant predictor of improved survival in patients with advanced non-small cell lung cancer. Medicine (Baltimore) 2019; 98:e16238. [PMID: 31261584 PMCID: PMC6616372 DOI: 10.1097/md.0000000000016238] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
This study tries to evaluate the associations between circulating C-reactive protein (CRP) and the overall survival of patients with non-small cell lung cancer (NSCLC).One hundred ninety-two patients with advanced NSCLC who treated with chemotherapy were enrolled in this study. The cut-off value of CRP concentration was 5.0 mg/L. The patients were divided into low, intermediate and high 3 groups respectively according to the baseline level of CRP before the treatment. Kaplan-Meier analysis and Cox proportional-hazard models were used to evaluate the relationship between the CRP and overall survival time of patients.After adjusting for age, gender, smoking history, pathologic type, CRP was a significant independent impact which predicts the survival prognosis of patients with NSCLC. For all patients, the hazard ratio with high CRP levels for NSCLC-specific survival was 1.83 [95%confidenceinterval (CI) = 0.96, 3.48] compared with low CRP levels. The level of CRP was significantly correlated with survival time (hazard ratio = 1.77; 95% CI = 0.73, 4.26) for the patient with first-line chemotherapy. Patients with high level of circulating CRP also responded poorly to chemotherapy.A high level of circulating CRP was associated with a poor response and worse survival in patients with NSCLC.
Collapse
|
36
|
Huynh J, Chand A, Gough D, Ernst M. Therapeutically exploiting STAT3 activity in cancer - using tissue repair as a road map. Nat Rev Cancer 2019; 19:82-96. [PMID: 30578415 DOI: 10.1038/s41568-018-0090-8] [Citation(s) in RCA: 365] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The tightly orchestrated temporal and spatial control of signal transducer and activator of transcription 3 (STAT3) activity in epithelial, immune and stromal cells is critical for wound healing and tissue repair. Excessive STAT3 activation within cancer cells and cells of the tumour microenvironment can be viewed as a neoplastic mimic of an inflammation-driven repair response that collectively promotes tumour progression. In addition to the canonical transcriptional pathways by which STAT3 promotes stem cell-like characteristics, survival, proliferation, metastatic potential and immune evasion, cytoplasmic STAT3 activity fuels tumour growth by metabolic and other non-transcriptional mechanisms. Here, we review the tumour-modulating activities of STAT3 in light of its role as a signalling node integrating inflammatory responses during wound healing. Accordingly, many of the cytokines that contribute to the para-inflammatory state of most solid malignancies converge on and underpin dysregulated STAT3 activity. Targeting of these cytokines, their cognate receptors and associated signalling cascades in clinical trials is beginning to demonstrate therapeutic efficacy, given that interference with STAT3 activity is likely to simultaneously curb the growth of cancer cells and augment antitumour immunity.
Collapse
Affiliation(s)
- Jennifer Huynh
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
| | - Ashwini Chand
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
| | - Daniel Gough
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia.
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia.
| |
Collapse
|
37
|
Yang L, Li J, Xu L, Lin S, Xiang Y, Dai X, Liang G, Huang X, Zhu J, Zhao C. Rhein shows potent efficacy against non-small-cell lung cancer through inhibiting the STAT3 pathway. Cancer Manag Res 2019; 11:1167-1176. [PMID: 30774444 PMCID: PMC6362962 DOI: 10.2147/cmar.s171517] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Non-small-cell lung cancer (NSCLC) comprises about 85% of all lung cancers and is usually diagnosed at an advanced stage with poor prognosis. The IL-6/STAT3 signaling pathway plays a pivotal role in NSCLC biology. Rhein is a lipophilic anthraquinone extensively found in medicinal herbs. Emerging evidence suggests that Rhein has significant antitumor effects, supporting the potential uses of Rhein as an antitumor agent. METHODS Cell viability and colony formation were performed to examine Rhein's potent anti-proliferative effect in human NSCLC cell lines PC-9, H460 and A549. Flow cytometry-based assay was employed to study whether Rhein could affect cell apoptosis and cycle. The expression level of P-STAT3, apoptosis and cycle-related proteins Bcl-2, Bax, MDM2, CDC2, P53 and CyclinB1 were detected by Western blotting. The xenograft models were used to evaluate the in vivo effect of Rhein. RESULTS We found that Rhein could significantly reduce the viability and stimulate apoptosis in human NSCLC cells in a dose-dependent manner. Western blot analysis results suggested that the antitumor effect of Rhein might be mediated via STAT3 inhibition. Rhein upregulated the expression of the proapoptotic protein Bax and downregulated the expression of the antiapoptotic protein Bcl-2. In addition, Rhein induced the arrest of NSCLC cells in the G2/M phase of the cell cycle and dose dependently inhibited the expression of cycle-related proteins. The Rhein also inhibited tumor growth in H460 xenograft models. CONCLUSION Rhein shows potent efficacy against NSCLC through inhibiting the STAT3 pathway. Our results also suggest that Rhein has a promising potential to be used as a novel antitumor agent for the treatment of NSCLC.
Collapse
Affiliation(s)
- Lehe Yang
- Department of Respiratory Medicine, Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, People's Republic of China, ,
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China,
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, People's Republic of China
| | - Jifa Li
- Department of Respiratory Medicine, Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, People's Republic of China, ,
| | - Lingyuan Xu
- Department of Respiratory Medicine, Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, People's Republic of China, ,
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China,
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, People's Republic of China
| | - Shichong Lin
- Department of Respiratory Medicine, Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, People's Republic of China, ,
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China,
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, People's Republic of China
| | - Youqun Xiang
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, People's Republic of China
| | - Xuanxuan Dai
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, People's Republic of China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China,
| | - Xiaoying Huang
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, People's Republic of China
| | - Jiandong Zhu
- Department of Respiratory Medicine, Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, People's Republic of China, ,
| | - Chengguang Zhao
- Department of Respiratory Medicine, Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, People's Republic of China, ,
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China,
| |
Collapse
|
38
|
Chong SJF, Lai JXH, Eu JQ, Bellot GL, Pervaiz S. Reactive Oxygen Species and Oncoprotein Signaling-A Dangerous Liaison. Antioxid Redox Signal 2018; 29:1553-1588. [PMID: 29186971 DOI: 10.1089/ars.2017.7441] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
SIGNIFICANCE There is evidence to implicate reactive oxygen species (ROS) in tumorigenesis and its progression. This has been associated with the interplay between ROS and oncoproteins, resulting in enhanced cellular proliferation and survival. Recent Advances: To date, studies have investigated specific contributions of the crosstalk between ROS and signaling networks in cancer initiation and progression. These investigations have challenged the established dogma of ROS as agents of cell death by demonstrating a secondary function that fuels cell proliferation and survival. Studies have thus identified (onco)proteins (Bcl-2, STAT3/5, RAS, Rac1, and Myc) in manipulating ROS level as well as exploiting an altered redox environment to create a milieu conducive for cancer formation and progression. CRITICAL ISSUES Despite these advances, drug resistance and its association with an altered redox metabolism continue to pose a challenge at the mechanistic and clinical levels. Therefore, identifying specific signatures, altered protein expressions, and modifications as well as protein-protein interplay/function could not only enhance our understanding of the redox networks during cancer initiation and progression but will also provide novel targets for designing specific therapeutic strategies. FUTURE DIRECTIONS Not only a heightened realization is required to unravel various gene/protein networks associated with cancer formation and progression, particularly from the redox standpoint, but there is also a need for developing more sensitive tools for assessing cancer redox metabolism in clinical settings. This review attempts to summarize our current knowledge of the crosstalk between oncoproteins and ROS in promoting cancer cell survival and proliferation and treatment strategies employed against these oncoproteins. Antioxid. Redox Signal.
Collapse
Affiliation(s)
- Stephen Jun Fei Chong
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Jolin Xiao Hui Lai
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Jie Qing Eu
- 2 Cancer Science Institute , Singapore, Singapore
| | - Gregory Lucien Bellot
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,3 Department of Hand and Reconstructive Microsurgery, National University Health System , Singapore, Singapore
| | - Shazib Pervaiz
- 1 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,4 NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , Singapore, Singapore .,5 National University Cancer Institute, National University Health System , Singapore, Singapore .,6 School of Biomedical Sciences, Curtin University , Perth, Australia
| |
Collapse
|
39
|
Genome-wide identification of transcription factors that are critical to non-small cell lung cancer. Cancer Lett 2018; 434:132-143. [DOI: 10.1016/j.canlet.2018.07.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/04/2018] [Accepted: 07/13/2018] [Indexed: 12/25/2022]
|
40
|
Kaida H, Azuma K, Kawahara A, Sadashima E, Hattori S, Takamori S, Akiba J, Fujimoto K, Rominger A, Murakami T, Ishii K, Ishibashi M. The assessment of correlation and prognosis among 18F-FDG uptake parameters, Glut1, pStat1 and pStat3 in surgically resected non-small cell lung cancer patients. Oncotarget 2018; 9:31971-31984. [PMID: 30174790 PMCID: PMC6112832 DOI: 10.18632/oncotarget.25865] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 07/13/2018] [Indexed: 12/12/2022] Open
Abstract
Introduction To assess the correlation among 18F-FDG uptake, Glut1, pStat1 and pStat3, and to investigate the relationship between the prognosis and 18F-FDG uptake and these molecular markers in surgically resected non-small cell lung cancer (NSCLC) patients. Results Knockdown of Glut1 led to a significant increase in pStat1 expression. Glut1 expression positively correlated with the SUVmax, SUVmean, and TLG significantly (P<0.001). pStat3 expression negatively correlated with all PET parameters significantly (P<0.001). pStat1 had positive weak correlations with the SUVmax and SUVmean. All PET parameters and Glut1 were significantly associated with DFS (P<0.05). TLG, MTV, Glut1 and pStat1 were significantly associated with OS (P<0.05). Conclusion pStat3 and Glut1 may be associated with 18F-FDG uptake mechanism. TLG, MTV, and Glut1 may be independent prognostic factors. Methods The SUVmax, SUVmean, MTV and TLG of primary lesions were calculated in 140 patients. The expressions of Glut1 and Stat pathway proteins in NSCLC cell lines were examined by immune blots. Excised tumor tissue was analyzed by immunohistochemistry. OS and DFS were evaluated by the Kaplan-Meier method. The difference in survival between subgroups was analyzed by log-rank test. The prognostic significance of clinicopathological, molecular and PET parameters was assessed by Cox proportional hazard regression analysis.
Collapse
Affiliation(s)
- Hayato Kaida
- Department of Radiology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Koichi Azuma
- Division of Respirology, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Akihiko Kawahara
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Fukuoka, Japan
| | - Eiji Sadashima
- Life Science, Saga-Ken Medical Centre Koseikan, Saga, Saga, Japan
| | - Satoshi Hattori
- Department of Biomedical Statistics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shinzo Takamori
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Jun Akiba
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Fukuoka, Japan
| | - Kiminori Fujimoto
- Department of Radiology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Takamichi Murakami
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Kazunari Ishii
- Department of Radiology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Masatoshi Ishibashi
- Department of Radiology, Fukuoka Tokushukai Medical Center, Kasuga, Fukuoka, Japan
| |
Collapse
|
41
|
Hu J, Zhang H, Cao M, Wang L, Wu S, Fang B. Auranofin Enhances Ibrutinib's Anticancer Activity in EGFR-Mutant Lung Adenocarcinoma. Mol Cancer Ther 2018; 17:2156-2163. [PMID: 30065099 DOI: 10.1158/1535-7163.mct-17-1173] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 03/27/2018] [Accepted: 07/25/2018] [Indexed: 11/16/2022]
Abstract
We previously found that ibrutinib has anticancer activity in EGFR-mutant non-small cell lung cancer (NSCLC). One of our recent studies showed that auranofin, a gold complex that has been used to treat rheumatoid arthritis, inhibited the PI3K/AKT/mTOR pathway and promoted apoptosis in some NSCLC cells. Because the PI3K/AKT/mTOR pathway is one of the major downstream pathways of EGFR, we hypothesized that ibrutinib's activity might be enhanced by combination therapy with auranofin in NSCLC cells. To this end, we examined ibrutinib's dose responses in EGFR-mutant H1975, PC9, and H1650 cells and in EGFR wild-type Calu3 and H460 cells in the presence or absence of auranofin. Although low concentrations of auranofin alone demonstrated mild anticancer activities, its presence dramatically enhanced ibrutinib's activity in H1975, PC9, and H1650 cells (IC50 value reduced 10- to 100-fold), but had only mild effect on Calu3 and H460 cells, demonstrating that ibrutinib's anti-EGFR activity is enhanced when it is combined with auranofin. A mechanistic analysis revealed that ibrutinib alone induced dramatic inhibition of the MEK/ERK pathway in both H1975 and H1650 cells, whereas auranofin alone inhibited the AKT/mTOR pathway. The combination of ibrutinib and auranofin led to a dramatically enhanced inhibition of the expression or phosphorylation of multiple key nodes in the AKT/mTOR and MEK/ERK pathways in both cell lines. In mice, the combination of ibrutinib and auranofin significantly suppressed the growth of H1975 xenografted tumors without inducing obvious toxic effects. Our results demonstrate the feasibility of improving ibrutinib's anti-EGFR activity for NSCLC using combination therapy with auranofin. Mol Cancer Ther; 17(10); 2156-63. ©2018 AACR.
Collapse
Affiliation(s)
- Jing Hu
- The 4th Department of Internal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China. .,Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Huijuan Zhang
- The 4th Department of Internal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Mengru Cao
- The 4th Department of Internal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.,Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Li Wang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shuhong Wu
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bingliang Fang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
42
|
Zhang X, Zhang S, Sun Q, Jiao W, Yan Y, Zhang X. Compound K Induces Endoplasmic Reticulum Stress and Apoptosis in Human Liver Cancer Cells by Regulating STAT3. Molecules 2018; 23:E1482. [PMID: 29921768 PMCID: PMC6099685 DOI: 10.3390/molecules23061482] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 06/11/2018] [Accepted: 06/14/2018] [Indexed: 02/06/2023] Open
Abstract
The ginsenoside compound K (20-O-β-d-glucopyranosyl-20(S)-protopanaxadiol; CK) is an intestinal bacterial metabolite of ginseng protopanaxadiol saponin that has been reported to induce apoptosis in many cancer cells; however, the precise mechanisms of its activity in human hepatocellular carcinoma (HCC) cells remain unclear. Herein, we demonstrated that CK inhibited the growth and colony formation of HepG2 and SMMC-7721 cells, phenotypes that were mediated by inducing apoptosis. Meanwhile, CK showed lower toxicity in normal hepatoma cells. After treating HepG2 and SMMC-7721 cells with CK, p-STAT3 levels decreased, the three branches of the unfolded protein response were activated, and levels of endoplasmic reticulum stress (ERS)-related proteins were increased. We also revealed that CK decreased the DNA-binding capacity of STAT3. Moreover, silencing STAT3 with CRISPR/Cas9 technology enhanced CK-induced ERS and apoptosis. Finally, we showed that CK inhibited the growth of liver cancer xenografts with little toxicity. Mice bearing human HCC xenografts that were treated with CK showed increased GRP78 expression and decreased p-STAT3 levels. Taken together, these data showed that CK induced ERS and apoptosis by inhibiting p-STAT3 in human liver cancer cells; thus, CK might be a potential therapeutic candidate for human HCC.
Collapse
Affiliation(s)
- Xuan Zhang
- College of Medicine, Yanbian University, Yanji 133000, China.
| | - Silin Zhang
- College of Medicine, Yanbian University, Yanji 133000, China.
| | - Qitong Sun
- College of Medicine, Yanbian University, Yanji 133000, China.
| | - Wenjun Jiao
- College of Medicine, Yanbian University, Yanji 133000, China.
| | - Yan Yan
- College of Medicine, Yanbian University, Yanji 133000, China.
| | - Xuewu Zhang
- College of Medicine, Yanbian University, Yanji 133000, China.
| |
Collapse
|
43
|
Bandola-Simon J, Roche PA. Dysfunction of antigen processing and presentation by dendritic cells in cancer. Mol Immunol 2018; 113:31-37. [PMID: 29628265 DOI: 10.1016/j.molimm.2018.03.025] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 01/31/2018] [Accepted: 03/29/2018] [Indexed: 12/23/2022]
Abstract
The ability to mount an effective anti-tumor immune response requires coordinate control of CD4 T cell and CD8 T cell function by antigen presenting cells (APCs). Unfortunately, tumors create an immunosuppressive microenvironment that helps protect tumor cells from immune recognition. In many cases this defect can be traced back to a failure of APCs (most importantly dendritic cells (DCs)) to recognize, process, and present tumor antigens to T cells. In this review, we will summarize work addressing the role of different DC subsets in anti-tumor immunity and the various mechanisms used by tumor cells to suppress the ability of APCs to stimulate potent anti-tumor T cell responses.
Collapse
Affiliation(s)
- Joanna Bandola-Simon
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Paul A Roche
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States.
| |
Collapse
|
44
|
Proteomic identification of the oncoprotein STAT3 as a target of a novel Skp1 inhibitor. Oncotarget 2018; 8:2681-2693. [PMID: 27835873 PMCID: PMC5356833 DOI: 10.18632/oncotarget.13153] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 10/14/2016] [Indexed: 12/17/2022] Open
Abstract
The S phase kinase-associated protein 1 (Skp1), an adaptor protein of the Skp1-Cul1-F-box protein complex, binds the ubiquitin E3 ligase Skp2 and is critical to its biological functions. Targeting of Skp1 by a small compound 6-O-angeloylplenolin (6-OAP) results in dissociation and degradation of Skp2 and mitotic arrest of lung cancer cells. Here, by using a proteome microarray containing 16,368 proteins and a biotinylated 6-OAP, we identified 99 proteins that could bind 6-OAP, with Skp1 and STAT3 sitting at the central position of the 6-OAP interactome. 6-OAP formed hydrogen bonds with Ser611/Ser613/Arg609 at the SH2 domain of STAT3 and inhibited the constitutive and interleukin-6-induced phosphorylated STAT3 (pSTAT3), leading to inhibitory effects on lung cancer cells and suppression of Skp2 transcription. STAT3 was overexpressed in tumor samples compared to counterpart normal lung tissues and was inversely associated with prognosis of the patients. 6-OAP inhibited tumor growth in SCID mice intravenously injected with lung cancer cells, and downregulated both STAT3 and Skp2 in tumor samples. Given that 6-OAP is a Skp1 inhibitor, our data suggest that this compound may target Skp1 and STAT3 to suppress Skp2, augmenting its anti-lung cancer activity.
Collapse
|
45
|
Yang XW, Li L, Hou GJ, Yan XZ, Xu QG, Chen L, Zhang BH, Shen F. STAT3 overexpression promotes metastasis in intrahepatic cholangiocarcinoma and correlates negatively with surgical outcome. Oncotarget 2018; 8:7710-7721. [PMID: 28032598 PMCID: PMC5352354 DOI: 10.18632/oncotarget.13846] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 11/30/2016] [Indexed: 12/21/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) promotes tumor progression in many types of cancer. In this study, we analyzed the prognostic value of this marker in human intrahepatic cholangiocarcinoma (ICC). Using real-time PCR, western blot and immunohistochemistry assays, we found that STAT3 is overexpressed in ICC patients. STAT3 expression correlated with several clinicopathological features, including tumor size, pathological satellite, vascular invasion, undifferentiated-type histology, lymph node metastasis and TNM stage in two independent cohorts of ICC patients. Patients with high STAT3 levels had a poor prognosis in terms of overall survival (OS) and disease-free survival (DFS). Multivariate survival analysis indicated that STAT3 is an independent prognostic factor for OS and DFS. Furthermore, we observed that STAT3 overexpression promotes the invasion, metastasis and proliferation of ICC cells in vitro and in vivo, and also promotes STAT3 phosphorylation. These findings suggest that STAT3 expression correlated negatively with surgical outcome and inhibition of STAT3 expression may constitute a novel target for the treatment of ICC patients.
Collapse
Affiliation(s)
- Xin-Wei Yang
- Department of Laparoscopy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Liang Li
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai 200438, China
| | - Guo-Jun Hou
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Xin-Zhou Yan
- Department of Laparoscopy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Qin-Guo Xu
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Lei Chen
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai 200438, China
| | - Bao-Hua Zhang
- Department of Laparoscopy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Feng Shen
- Department of Comprehensive Treatment, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| |
Collapse
|
46
|
Oh S, Kim H, Nam K, Shin I. Egr-1 is required for neu/HER2-induced mammary tumors. Cell Signal 2018; 45:102-109. [PMID: 29408223 DOI: 10.1016/j.cellsig.2018.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/11/2018] [Accepted: 02/01/2018] [Indexed: 01/23/2023]
Abstract
Egr-1 is known to function mainly as a tumor suppressor through direct regulation of multiple tumor suppressor genes. To determine the role of Egr-1 in breast tumors in vivo, we used mouse models of breast cancer induced by HER2/neu. We compared neu-overexpressing Egr-1 knockout mice (neu/Egr-1 KO) to neu-overexpressing Egr-1 wild type or heterozygote mice (neu/Egr-1 WT or neu/Egr-1 het) with regard to onset of tumor appearance and number of tumors per mouse. In addition, to examine the role of Egr-1 in vitro, we established neu/Egr-1 WT and KO tumor cell lines derived from breast tumors developed in each mouse. Egr-1 deletion delayed tumor development in vivo and decreased the rate of cell growth in vitro. These results suggest that Egr-1 plays an oncogenic role in HER2/neu-driven mammary tumorigenesis.
Collapse
Affiliation(s)
- Sunhwa Oh
- Department of Life Science, Hanyang University, Seoul 04763, Republic of Korea
| | - Hyungjoo Kim
- Department of Life Science, Hanyang University, Seoul 04763, Republic of Korea
| | - KeeSoo Nam
- Department of Life Science, Hanyang University, Seoul 04763, Republic of Korea
| | - Incheol Shin
- Department of Life Science, Hanyang University, Seoul 04763, Republic of Korea; Natural Science Institute, Hanyang University, Seoul 04763, Republic of Korea.
| |
Collapse
|
47
|
Chhabra Y, Wong HY, Nikolajsen LF, Steinocher H, Papadopulos A, Tunny KA, Meunier FA, Smith AG, Kragelund BB, Brooks AJ, Waters MJ. A growth hormone receptor SNP promotes lung cancer by impairment of SOCS2-mediated degradation. Oncogene 2018; 37:489-501. [PMID: 28967904 PMCID: PMC5799715 DOI: 10.1038/onc.2017.352] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 08/13/2017] [Accepted: 08/16/2017] [Indexed: 02/07/2023]
Abstract
Both humans and mice lacking functional growth hormone (GH) receptors are known to be resistant to cancer. Further, autocrine GH has been reported to act as a cancer promoter. Here we present the first example of a variant of the GH receptor (GHR) associated with cancer promotion, in this case lung cancer. We show that the GHRP495T variant located in the receptor intracellular domain is able to prolong the GH signal in vitro using stably expressing mouse pro-B-cell and human lung cell lines. This is relevant because GH secretion is pulsatile, and extending the signal duration makes it resemble autocrine GH action. Signal duration for the activated GHR is primarily controlled by suppressor of cytokine signalling 2 (SOCS2), the substrate recognition component of the E3 protein ligase responsible for ubiquitinylation and degradation of the GHR. SOCS2 is induced by a GH pulse and we show that SOCS2 binding to the GHR is impaired by a threonine substitution at Pro 495. This results in decreased internalisation and degradation of the receptor evident in TIRF microscopy and by measurement of mature (surface) receptor expression. Mutational analysis showed that the residue at position 495 impairs SOCS2 binding only when a threonine is present, consistent with interference with the adjacent Thr494. The latter is key for SOCS2 binding, together with nearby Tyr487, which must be phosphorylated for SOCS2 binding. We also undertook nuclear magnetic resonance spectroscopy approach for structural comparison of the SOCS2 binding scaffold Ile455-Ser588, and concluded that this single substitution has altered the structure of the SOCS2 binding site. Importantly, we find that lung BEAS-2B cells expressing GHRP495T display increased expression of transcripts associated with tumour proliferation, epithelial-mesenchymal transition and metastases (TWIST1, SNAI2, EGFR, MYC and CCND1) at 2 h after a GH pulse. This is consistent with prolonged GH signalling acting to promote cancer progression in lung cancer.
Collapse
Affiliation(s)
- Y Chhabra
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - H Y Wong
- University of Queensland Centre for Clinical Research, The University of Queensland, Herston, Queensland, Australia
| | - L F Nikolajsen
- Structural Biology and NMR Laboratory (SBiNLab), Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - H Steinocher
- Structural Biology and NMR Laboratory (SBiNLab), Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - A Papadopulos
- The Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - K A Tunny
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - F A Meunier
- The Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - A G Smith
- School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute, Queensland University of Technology, Woolloongabba, Queensland, Australia
| | - B B Kragelund
- Structural Biology and NMR Laboratory (SBiNLab), Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - A J Brooks
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - M J Waters
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
48
|
Wu P, Wu D, Zhao L, Huang L, Shen G, Huang J, Chai Y. Prognostic role of STAT3 in solid tumors: a systematic review and meta-analysis. Oncotarget 2017; 7:19863-83. [PMID: 26959884 PMCID: PMC4991424 DOI: 10.18632/oncotarget.7887] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 02/13/2016] [Indexed: 12/22/2022] Open
Abstract
Accumulated studies have provided controversial evidences of the association between signal transducer and activator of transcription proteins 3 (STAT3) expression and survival of human solid tumors. To address this inconsistency, we performed a meta-analysis with 63 studies identified from PubMed, Medline and EBSCO. We found STAT3 overexpression was significantly associated with worse 3-year overall survival (OS) (OR = 2.06, 95% CI = 1.57 to 2.71, P < 0.00001) and 5-year OS (OR = 2.00, 95% CI = 1.53 to 2.63, P < 0.00001) of human solid tumors. Similar results were observed when disease free survival (DFS) were analyzed. Subgroup analysis showed that elevated STAT3 expression was associated with poor prognosis of gastric cancer, lung cancer, gliomas, hepatic cancer, osteosarcoma, prostate cancer, pancreatic cancer but better prognosis of breast cancer. The correlation between STAT3 and survival of solid tumors was related to its phosphorylated state. High expression level of STAT3 was also associated with advanced tumor stage. In conclusion, elevated STAT3 expression is associated with poor survival in most solid tumors. STAT3 is a valuable biomarker for prognosis prediction and a promising therapeutic target in human solid tumors.
Collapse
Affiliation(s)
- Pin Wu
- Department of Thoracic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China.,Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Dang Wu
- Department of Radiation Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China.,Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Lufeng Zhao
- Department of Thoracic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Lijian Huang
- Department of Thoracic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Gang Shen
- Department of Thoracic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jian Huang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China.,Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Ying Chai
- Department of Thoracic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| |
Collapse
|
49
|
Hu C, Wang A, Wu H, Qi Z, Li X, Yan XE, Chen C, Yu K, Zou F, Wang W, Wang W, Wu J, Liu J, Wang B, Wang L, Ren T, Zhang S, Yun CH, Liu J, Liu Q. Discovery and characterization of a novel irreversible EGFR mutants selective and potent kinase inhibitor CHMFL-EGFR-26 with a distinct binding mode. Oncotarget 2017; 8:18359-18372. [PMID: 28407693 PMCID: PMC5392334 DOI: 10.18632/oncotarget.15443] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 01/23/2017] [Indexed: 12/18/2022] Open
Abstract
EGFR T790M mutation accounts for about 40-55% drug resistance for the first generation EGFR kinase inhibitors in the NSCLC. Starting from ibrutinib, a highly potent irreversible BTK kinase inhibitor, which was also found to be moderately active to EGFR T790M mutant, we discovered a highly potent irreversible EGFR inhibitor CHMFL-EGFR-26, which is selectively potent against EGFR mutants including L858R, del19, and L858R/T790M. It displayed proper selectivity window between the EGFR mutants and the wide-type. CHMFL-EGFR-26 exhibited good selectivity profile among 468 kinases/mutants tested (S score (1)=0.02). In addition, X-ray crystallography revealed a distinct “DFG-in” and “cHelix-out” inactive binding mode between CHMFL-EGFR-26 and EGFR T790M protein. The compound showed highly potent anti-proliferative efficacy against EGFR mutant but not wide-type NSCLC cell lines through effective inhibition of the EGFR mediated signaling pathway, induction of apoptosis and arresting of cell cycle progression. CHMFL-EGFR-26 bore acceptable pharmacokinetic properties and demonstrated dose-dependent tumor growth suppression in the H1975 (EGFR L858R/T790M) and PC-9 (EGFR del19) inoculated xenograft mouse models. Currently CHMFL-EGFR-26 is undergoing extensive pre-clinical evaluation for the clinical trial purpose.
Collapse
Affiliation(s)
- Chen Hu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,University of Science and Technology of China, Hefei, Anhui 230036, P. R. China
| | - Aoli Wang
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,University of Science and Technology of China, Hefei, Anhui 230036, P. R. China
| | - Hong Wu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory, Hefei, Anhui 230031, P. R. China
| | - Ziping Qi
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory, Hefei, Anhui 230031, P. R. China
| | - Xixiang Li
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory, Hefei, Anhui 230031, P. R. China
| | - Xiao-E Yan
- Institute of Systems Biomedicine, Department of Biophysics, Beijing Key Laboratory of Tumor Systems Biology and Center for Molecular and Translational Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Cheng Chen
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,University of Science and Technology of China, Hefei, Anhui 230036, P. R. China
| | - Kailin Yu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,University of Science and Technology of China, Hefei, Anhui 230036, P. R. China
| | - Fengming Zou
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory, Hefei, Anhui 230031, P. R. China
| | - Wenchao Wang
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory, Hefei, Anhui 230031, P. R. China
| | - Wei Wang
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory, Hefei, Anhui 230031, P. R. China
| | - Jiaxin Wu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,University of Science and Technology of China, Hefei, Anhui 230036, P. R. China
| | - Juan Liu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,University of Science and Technology of China, Hefei, Anhui 230036, P. R. China
| | - Beilei Wang
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,University of Science and Technology of China, Hefei, Anhui 230036, P. R. China
| | - Li Wang
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,University of Science and Technology of China, Hefei, Anhui 230036, P. R. China
| | - Tao Ren
- Precision Targeted Therapy Discovery Center, Institute of Technology Innovation, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230088, P. R. China
| | - Shanchun Zhang
- CHMFL-HCMTC Target Therapy Joint Laboratory, Hefei, Anhui 230031, P. R. China.,Hefei Cosource Medicine Technology Co. LTD., Hefei, Anhui 230031, P. R. China
| | - Cai-Hong Yun
- Institute of Systems Biomedicine, Department of Biophysics, Beijing Key Laboratory of Tumor Systems Biology and Center for Molecular and Translational Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Jing Liu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory, Hefei, Anhui 230031, P. R. China
| | - Qingsong Liu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,University of Science and Technology of China, Hefei, Anhui 230036, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory, Hefei, Anhui 230031, P. R. China.,Precision Targeted Therapy Discovery Center, Institute of Technology Innovation, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230088, P. R. China
| |
Collapse
|
50
|
Clay TD, Russell PA, Do H, Sundararajan V, Conron M, Wright GM, Solomon B, Dobrovic A, McLachlan SA, Moore MM. EGFR and KRAS mutations do not enrich for the activation of IL-6, JAK1 or phosphorylated STAT3 in resected lung adenocarcinoma. Med Oncol 2017; 34:175. [PMID: 28879441 DOI: 10.1007/s12032-017-1031-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 08/29/2017] [Indexed: 01/11/2023]
Abstract
Resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) against EGFR mutant lung adenocarcinoma develops after a median of nine to thirteen months. Upregulation of the interleukin-6 (IL-6)/Janus kinase (JAK)/signal transducers and activators of transcription (STAT) pathway may be a potential source of resistance to EGFR TKIs. We undertook a detailed assessment of the IL-6/JAK1/phosphorylated STAT3 (pSTAT3) pathway in resected lung adenocarcinoma specimens, with special interest in whether the presence of an EGFR mutation enriched for pSTAT3 positivity. Tumours from 143 patients with resected lung adenocarcinoma were assessed. EGFR and KRAS mutation status were scanned for with high-resolution melting and confirmed by polymerase chain reaction. Immunohistochemisty (IHC) was performed for IL-6, gp130, JAK1 and pSTAT3. Two methods for assigning IHC positivity were assessed (the presence of any positivity, and the presence of positivity at an H score >40). We found statistically significant associations between IL-6, JAK1 and pSTAT3 measured by IHC, consistent with the activation of the pathway in clinical specimens. No relationship was demonstrated between members of this pathway and oncogenic mutations in EGFR or KRAS. However, a proportion of tumours with EGFR mutations showed staining for IL-6, JAK1 and pSTAT3. No correlations with clinicopathologic features or survival outcomes were found for IL-6, JAK1 or pSTAT3 staining. The presence of EGFR or KRAS mutations did not enrich for the activation of IL-6, JAK1 or pSTAT3. pSTAT3 may still play a role in resistance to EGFR TKIs in clinical practice.
Collapse
Affiliation(s)
- Timothy D Clay
- St John of God Hospital, Suite C202, 12 Salvado Road, Subiaco, 6008, Australia. .,Department of Medicine, St Vincent's Hospital, Melbourne Medical School, University of Melbourne, Melbourne, Australia.
| | - Prudence A Russell
- Department of Pathology, St Vincent's Hospital, Melbourne, Melbourne, Australia.,Department of Pathology, University of Melbourne, Melbourne, Australia
| | - Hongdo Do
- Department of Pathology, University of Melbourne, Melbourne, Australia.,Translational Genomics and Epigenomics Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Vijaya Sundararajan
- Department of Medicine, St Vincent's Hospital, Melbourne Medical School, University of Melbourne, Melbourne, Australia
| | - Matthew Conron
- Department of Medicine, St Vincent's Hospital, Melbourne Medical School, University of Melbourne, Melbourne, Australia.,Department of Respiratory Medicine, St Vincent's Hospital, Melbourne, Melbourne, Australia
| | - Gavin M Wright
- Department of Thoracic Surgery, St Vincent's Hospital, Melbourne, Melbourne, Australia.,Department of Thoracic Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia.,Department of Surgery, University of Melbourne, Melbourne, Australia
| | - Benjamin Solomon
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Alexander Dobrovic
- Department of Pathology, University of Melbourne, Melbourne, Australia.,Translational Genomics and Epigenomics Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Sue-Anne McLachlan
- Department of Medicine, St Vincent's Hospital, Melbourne Medical School, University of Melbourne, Melbourne, Australia.,Department of Medical Oncology, St Vincent's Hospital, Melbourne, Melbourne, Australia
| | - Melissa M Moore
- Department of Medicine, St Vincent's Hospital, Melbourne Medical School, University of Melbourne, Melbourne, Australia.,Department of Medical Oncology, St Vincent's Hospital, Melbourne, Melbourne, Australia
| |
Collapse
|