1
|
Wei R, Liao X, Li J, Mu X, Ming Y, Peng Y. Novel humanized monoclonal antibodies against ROR1 for cancer therapy. Mol Cancer 2024; 23:165. [PMID: 39138527 PMCID: PMC11321157 DOI: 10.1186/s12943-024-02075-y] [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: 06/12/2024] [Accepted: 07/29/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Overexpression of receptor tyrosine kinase-like orphan receptor 1 (ROR1) contributes to cancer cell proliferation, survival and migration, playing crucial roles in tumor development. ROR1 has been proposed as a potential therapeutic target for cancer treatment. This study aimed to develop novel humanized ROR1 monoclonal antibodies and investigate their anti-tumor effects. METHODS ROR1 expression in tumor tissues and cell lines was analyzed by immunohistochemistry and flow cytometry. Antibodies from mouse hybridomas were humanized by the complementarity-determining region (CDR) grafting technique. Surface plasmon resonance spectroscopy, ELISA assay and flow cytometry were employed to characterize humanized antibodies. In vitro cellular assay and in vivo mouse experiment were conducted to comprehensively evaluate anti-tumor activity of these antibodies. RESULTS ROR1 exhibited dramatically higher expression in lung adenocarcinoma, liver cancer and breast cancer, and targeting ROR1 by short-hairpin RNAs significantly inhibited proliferation and migration of cancer cells. Two humanized ROR1 monoclonal antibodies were successfully developed, named h1B8 and h6D4, with high specificity and affinity to ROR1 protein. Moreover, these two antibodies effectively suppressed tumor growth in the lung cancer xenograft mouse model, c-Myc/Alb-cre liver cancer transgenic mouse model and MMTV-PyMT breast cancer mouse model. CONCLUSIONS Two humanized monoclonal antibodies targeting ROR1, h1B8 and h6D4, were successfully developed and exhibited remarkable anti-tumor activity in vivo.
Collapse
Affiliation(s)
- Rong Wei
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xun Liao
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiao Li
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaoyu Mu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yue Ming
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yong Peng
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China.
| |
Collapse
|
2
|
Li L, Huang W, Ren X, Wang Z, Ding K, Zhao L, Zhang J. Unlocking the potential: advancements and future horizons in ROR1-targeted cancer therapies. SCIENCE CHINA. LIFE SCIENCES 2024:10.1007/s11427-024-2685-9. [PMID: 39145866 DOI: 10.1007/s11427-024-2685-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 07/11/2024] [Indexed: 08/16/2024]
Abstract
While receptor tyrosine kinase-like orphan receptor 1 (ROR1) is typically expressed at low levels or absent in normal tissues, its expression is notably elevated in various malignant tumors and conditions, including chronic lymphocytic leukemia (CLL), breast cancer, ovarian cancer, melanoma, and lung adenocarcinoma. This distinctive feature positions ROR1 as an attractive target for tumor-specific treatments. Currently, several targeted drugs directed at ROR1 are undergoing clinical development, including monoclonal antibodies, antibody-drug conjugates (ADCs), and chimeric antigen receptor T-cell therapy (CAR-T). Additionally, there are four small molecule inhibitors designed to bind to ROR1, presenting promising avenues for the development of PROTAC degraders targeting ROR1. This review offers updated insights into ROR1's structural and functional characteristics, embryonic development implications, cell survival signaling pathways, and evolutionary targeting strategies, all of which have the potential to advance the treatment of malignant tumors.
Collapse
Affiliation(s)
- Lin Li
- State Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Weixue Huang
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Xiaomei Ren
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Zhen Wang
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Ke Ding
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China.
| | - Linxiang Zhao
- State Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Jinwei Zhang
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China.
| |
Collapse
|
3
|
Xian J, Sinha N, Girgis C, Oh CS, Cring MR, Widhopf GF, Kipps TJ. Variant Transcript of ROR1 ENST00000545203 Does Not Encode ROR1 Protein. Biomedicines 2024; 12:1573. [PMID: 39062146 PMCID: PMC11274362 DOI: 10.3390/biomedicines12071573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Drs. John and Ford reported in biomedicines that a variant transcript encoding receptor tyrosine kinase-like orphan receptor 1 (ROR1), namely ENST00000545203 or variant 3 (ROR1V3), was a predominant ROR1 transcript of neoplastic or normal cells in the Bioinformatic database, including GTEx and the 33 datasets from TCGA. Unlike the full-length ROR1 transcript, Drs. John and Ford deduced that ROR1V3 encoded a cytoplasmic ROR1 protein lacking an apparent signal peptide necessary for transport to the cell surface, which they presumed made it unlikely to function as a surface receptor for Wingless/Integrated (Wnt) factors. Moreover, they speculated that studies evaluating ROR1 via immunohistochemistry using any one of several anti-ROR1 mAbs actually may have detected cytoplasmic protein encoded by ROR1V3 and that anti-cancer therapies targeting surface ROR1 thus would be ineffective against "cytoplasmic ROR1-positive" cancers that express predominately ROR1V3. We generated lentivirus vectors driving the expression of full-length ROR1 or the ROR1v3 upstream of an internal ribosome entry site (IRES) of the gene encoding a red fluorescent reporter protein. Although we find that cells that express ROR1 have surface and cytoplasmic ROR1 protein, cells that express ROR1v3 neither have surface nor cytoplasmic ROR1, which is consistent with our finding that ROR1v3 lacks an in-frame initiation codon for ribosomal translation into protein. We conclude that the detection of ROR1 protein in various cancers cannot be ascribed to the expression of ROR1v3.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Thomas J. Kipps
- Center for Novel Therapeutics, Moores Cancer Center, Department of Medicine, University of California, San Diego, CA 92037, USA; (J.X.); (N.S.); (C.G.); (C.S.O.); (M.R.C.); (G.F.W.II)
| |
Collapse
|
4
|
Wu ZL, Wang Y, Jia XY, Wang YG, Wang H. Receptor tyrosine kinase-like orphan receptor 1: A novel antitumor target in gastrointestinal cancers. World J Clin Oncol 2024; 15:603-613. [PMID: 38835843 PMCID: PMC11145958 DOI: 10.5306/wjco.v15.i5.603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/20/2024] [Accepted: 04/17/2024] [Indexed: 05/21/2024] Open
Abstract
Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is a member of the type I receptor tyrosine kinase family. ROR1 is pivotal in embryonic development and cancer, and serves as a biomarker and therapeutic target. It has soluble and membrane-bound subtypes, with the latter highly expressed in tumors. ROR1 is conserved throughout evolution and may play a role in the development of gastrointestinal cancer through multiple signaling pathways and molecular mechanisms. Studies suggest that overexpression of ROR1 may increase tumor invasiveness and metastasis. Additionally, ROR1 may regulate the cell cycle, stem cell characteristics, and interact with other signaling pathways to affect cancer progression. This review explores the structure, expression and role of ROR1 in the development of gastrointestinal cancers. It discusses current antitumor strategies, outlining challenges and prospects for treatment.
Collapse
Affiliation(s)
- Zheng-Long Wu
- Xinyuan Institute of Medicine and Biotechnology, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
- Department of Oncology, Zhejiang Xiaoshan Hospital, Hangzhou 311201, Zhejiang Province, China
| | - Ying Wang
- Xinyuan Institute of Medicine and Biotechnology, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Xiao-Yuan Jia
- Xinyuan Institute of Medicine and Biotechnology, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Yi-Gang Wang
- Xinyuan Institute of Medicine and Biotechnology, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Hui Wang
- Department of Oncology, Zhejiang Xiaoshan Hospital, Hangzhou 311201, Zhejiang Province, China
| |
Collapse
|
5
|
Shatsky RA, Batra-Sharma H, Helsten T, Schwab RB, Pittman EI, Pu M, Weihe E, Ghia EM, Rassenti LZ, Molinolo A, Cabrera B, Breitmeyer JB, Widhopf GF, Messer K, Jamieson C, Kipps TJ, Parker BA. A phase 1b study of zilovertamab in combination with paclitaxel for locally advanced/unresectable or metastatic Her2-negative breast cancer. Breast Cancer Res 2024; 26:32. [PMID: 38408999 PMCID: PMC10895766 DOI: 10.1186/s13058-024-01782-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/09/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Zilovertamab is a humanized monoclonal antibody targeting ROR1, an onco-embryonic antigen expressed by malignant cells of a variety of solid tumors, including breast cancer. A prior phase 1 study showed that zilovertamab was well tolerated and effective in inhibiting ROR1-signaling, which leads to activation of ERK1/2, NF-κB, and NRF2 target genes. This phase 1b study evaluated the safety and tolerability of zilovertamab with paclitaxel in patients with advanced breast cancer. PATIENTS AND METHODS Eligible patients had locally advanced, unresectable, or metastatic HER2- breast cancer with Eastern Cooperative Group performance status of 0-2 and without prior taxane therapy in the advanced setting. Study treatment included 600 mg of zilovertamab administered intravenously (IV) on Days 1 and 15 of Cycle 1 and then Day 1 of each 28-day cycle along with paclitaxel weekly at 80 mg/m2 IV. RESULTS Study patients had received a median of 4 prior therapies (endocrine therapy + chemotherapy) for locally advanced, unresectable, or metastatic disease. No patient discontinued therapy due to toxicity ascribed to zilovertamab. Adverse events were consistent with the known safety profile of paclitaxel. Of 16 patients, 6 (38%) had a partial response, and 6/16 (38%) patients had stable disease as best tumor response. CONCLUSION The combination of zilovertamab and paclitaxel was safe and well tolerated in heavily pre-treated advanced breast cancer patients. Further evaluation of ROR1 targeting in breast cancer patients with zilovertamab is warranted. TRIAL REGISTRATION NCT02776917. Registered on ClinicalTrials.gov on 05/17/2016.
Collapse
Affiliation(s)
- Rebecca A Shatsky
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive Mail Code 0987, La Jolla, San Diego, CA, 92093, USA
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Hemali Batra-Sharma
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive Mail Code 0987, La Jolla, San Diego, CA, 92093, USA
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Teresa Helsten
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive Mail Code 0987, La Jolla, San Diego, CA, 92093, USA
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Richard B Schwab
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive Mail Code 0987, La Jolla, San Diego, CA, 92093, USA
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Emily I Pittman
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive Mail Code 0987, La Jolla, San Diego, CA, 92093, USA
| | - Minya Pu
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive Mail Code 0987, La Jolla, San Diego, CA, 92093, USA
| | - Elizabeth Weihe
- Department of Radiology, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Emanuela M Ghia
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive Mail Code 0987, La Jolla, San Diego, CA, 92093, USA
- Center for Novel Therapeutics, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Laura Z Rassenti
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive Mail Code 0987, La Jolla, San Diego, CA, 92093, USA
- Center for Novel Therapeutics, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Alfredo Molinolo
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive Mail Code 0987, La Jolla, San Diego, CA, 92093, USA
| | - Betty Cabrera
- University of California San Diego California Institute for Regenerative Medicine Alpha Clinic, La Jolla, San Diego, CA, USA
| | | | - George F Widhopf
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
- Center for Novel Therapeutics, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Karen Messer
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive Mail Code 0987, La Jolla, San Diego, CA, 92093, USA
- Herbert Wertheim School of Public Health, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Catriona Jamieson
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive Mail Code 0987, La Jolla, San Diego, CA, 92093, USA
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
- University of California San Diego California Institute for Regenerative Medicine Alpha Clinic, La Jolla, San Diego, CA, USA
- Sanford Stem Cell Institute, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Thomas J Kipps
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive Mail Code 0987, La Jolla, San Diego, CA, 92093, USA
- Center for Novel Therapeutics, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Barbara A Parker
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive Mail Code 0987, La Jolla, San Diego, CA, 92093, USA.
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA.
| |
Collapse
|
6
|
Zabeti Touchaei A, Vahidi S, Samadani AA. Decoding the regulatory landscape of lncRNAs as potential diagnostic and prognostic biomarkers for gastric and colorectal cancers. Clin Exp Med 2024; 24:29. [PMID: 38294554 PMCID: PMC10830721 DOI: 10.1007/s10238-023-01260-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/12/2023] [Indexed: 02/01/2024]
Abstract
Colorectal cancer (CRC) and gastric cancer (GC) are major contributors to cancer-related mortality worldwide. Despite advancements in understanding molecular mechanisms and improved drug treatments, the overall survival rate for patients remains unsatisfactory. Metastasis and drug resistance are major challenges contributing to the high mortality rate in both CRC and GC. Recent research has shed light on the role of long noncoding RNAs (lncRNAs) in the development and progression of these cancers. LncRNAs regulate gene expression through various mechanisms, including epigenetic modifications and interactions with microRNAs (miRNAs) and proteins. They can serve as miRNA precursors or pseudogenes, modulating gene expression at transcriptional and post-transcriptional levels. Additionally, circulating lncRNAs have emerged as non-invasive biomarkers for the diagnosis, prognosis, and prediction of drug therapy response in CRC and GC. This review explores the intricate relationship between lncRNAs and CRC/GC, encompassing their roles in cancer development, progression, and chemoresistance. Furthermore, it discusses the potential of lncRNAs as therapeutic targets in these malignancies. The interplay between lncRNAs, miRNAs, and tumor microenvironment is also highlighted, emphasizing their impact on the complexity of cancer biology. Understanding the regulatory landscape and molecular mechanisms governed by lncRNAs in CRC and GC is crucial for the development of effective diagnostic and prognostic biomarkers, as well as novel therapeutic strategies. This review provides a comprehensive overview of the current knowledge and paves the way for further exploration of lncRNAs as key players in the management of CRC and GC.
Collapse
Affiliation(s)
| | - Sogand Vahidi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Ali Akbar Samadani
- Guilan Road Trauma Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
7
|
Qiao JX, Witmer MR, Lee V, Wang TC, Reid PC, Arioka Y, Farr G, Hill-Drzewi M, Schweizer L, Yamniuk A, Cheng L, Abramczyk B, Corbett M, Calambur D, Szapiel N, Ryseck R, Ponath P, Poss MA, Carter P. Exploration of macrocyclic peptide binders to the extracellular CRD domain of human receptor tyrosine kinase-like orphan receptor 1 (ROR1). Bioorg Med Chem Lett 2024; 98:129589. [PMID: 38097140 DOI: 10.1016/j.bmcl.2023.129589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 12/03/2023] [Accepted: 12/07/2023] [Indexed: 12/20/2023]
Abstract
Elevated levels of receptor tyrosine kinase-like orphan receptor 1 (RORl) expression are observed in multiple hematological and solid tumors, but not in most of the healthy adult tissues, identifying ROR1 as an attractive target for tumor-specific therapy. Herein we will describe the discovery of macrocyclic peptides as binders of the extracellular Cysteine-Rich Domain (CRD) of human ROR1 via mRNA in vitro selection technology using the PDPS platform, followed by exploration of sidechain SAR of parent macrocycle peptides, fluorescently labeled analogs, and a Peptide Drug Conjugate (PDC). The parent macrocyclic peptides represented by Compound 1 and Compound 14 displayed nanomolar cell-based binding to ROR1 and relatively good internalization in 786-O and MDA-MB-231 tumor cell lines. However, these peptides were not observed to induce apoptosis in Mia PaCa-2 cells, a model pancreatic tumor cell line with a relatively low level of cell surface expression of ROR1.
Collapse
Affiliation(s)
| | - Mark R Witmer
- Molecular Discovery Technology, Princeton, NJ 08543, United States
| | - Ving Lee
- Discovery Chemistry, Princeton, NJ 08543, United States
| | - Tammy C Wang
- Discovery Chemistry, Princeton, NJ 08543, United States
| | - Patrick C Reid
- PeptiDream 3-25-23 Tonomachi, Kawasaki-Ku, Kawasaki-Shi, Kanagawa 210-0821, Japan
| | - Yuki Arioka
- PeptiDream 3-25-23 Tonomachi, Kawasaki-Ku, Kawasaki-Shi, Kanagawa 210-0821, Japan
| | - Glen Farr
- Leads Discovery & Optimization, Princeton, NJ 08543, United States
| | | | - Liang Schweizer
- Leads Discovery & Optimization, Princeton, NJ 08543, United States
| | - Aaron Yamniuk
- Molecular Discovery Technology, Princeton, NJ 08543, United States
| | - Lin Cheng
- Molecular Discovery Technology, Princeton, NJ 08543, United States
| | - Bozena Abramczyk
- Molecular Discovery Technology, Princeton, NJ 08543, United States
| | - Martin Corbett
- Molecular Discovery Technology, Princeton, NJ 08543, United States
| | - Deepa Calambur
- Molecular Discovery Technology, Princeton, NJ 08543, United States
| | - Nicolas Szapiel
- Molecular Discovery Technology, Princeton, NJ 08543, United States
| | - Rolf Ryseck
- Molecular Discovery Technology, Princeton, NJ 08543, United States
| | - Paul Ponath
- Discovery Biology, Bristol-Myers Squibb, Princeton, NJ 08543, United States
| | | | - Percy Carter
- Discovery Chemistry, Princeton, NJ 08543, United States
| |
Collapse
|
8
|
Zhang N, Li Y. Receptor tyrosine kinases: biological functions and anticancer targeted therapy. MedComm (Beijing) 2023; 4:e446. [PMID: 38077251 PMCID: PMC10701465 DOI: 10.1002/mco2.446] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 10/16/2024] Open
Abstract
Receptor tyrosine kinases (RTKs) are a class of protein kinases that play crucial roles in various cellular processes, including cell migration, morphological differentiation, cell growth, and angiogenesis. In humans, 58 RTKs have been identified and categorized into 20 distinct families based on the composition of their extracellular regions. RTKs are primarily activated by specific ligands that bind to their extracellular region. They not only regulate tumor transformation, proliferation, metastasis, drug resistance, and angiogenesis, but also initiate and maintain the self-renewal and cloning ability of cancer stem cells. Accurate diagnosis and grading of tumors with dysregulated RTKs are essential in clinical practice. There is a growing body of evidence supporting the benefits of RTKs-targeted therapies for cancer patients, and researchers are actively exploring new targets and developing targeted agents. However, further optimization of RTK inhibitors is necessary to effectively target the diverse RTK alterations observed in human cancers. This review provides insights into the classification, structure, activation mechanisms, and expression of RTKs in tumors. It also highlights the research advances in RTKs targeted anticancer therapy and emphasizes their significance in optimizing cancer diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Nan Zhang
- Chongqing University Cancer Hospital, School of MedicineChongqing UniversityChongqingChina
| | - Yongsheng Li
- Chongqing University Cancer Hospital, School of MedicineChongqing UniversityChongqingChina
- Department of Medical OncologyChongqing University Cancer HospitalChongqingChina
| |
Collapse
|
9
|
Heabah NAEG, Darwish SA, Eid AM. Evaluation of the prognostic significance of receptor tyrosine kinase-like orphan receptor 1 (ROR1) in lung carcinoma and its relation to lymphangiogenesis and epithelial mesenchymal transition. Pathol Res Pract 2023; 248:154703. [PMID: 37481855 DOI: 10.1016/j.prp.2023.154703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
Exploring the carcinogenic mechanisms of lung carcinoma helps to discover novel prognostic biomarkers and develop new therapeutic options to improve patient's survival. Receptor Tyrosine Kinase-Like Orphan Receptor 1 (ROR1), a transmembrane protein, contributes to cancer progression and metastasis; via stimulation of epithelial mesenchymal transition (EMT) and promotion of angiogenesis. This makes ROR1 an important target for tumor therapy. This study aimed to evaluate expression of ROR1, E-cadherin (a marker of EMT), and D2-40 (a marker of lymphangiogenesis) in lung carcinoma and associate their expressions with the available clinicopathological parameters and patients' survival. Immunohistochemical staining using ROR1, E-cadherin, and D2-40 was performed for 78 cases of lung carcinoma. Kaplan-Meier survival curves and Cox-regression analysis were done. High ROR1 expression was detected in 46.2% of cases. Significant relations were found between high ROR1 expression and larger tumor size (P < 0.001), poorly differentiated tumors (P = 0.001), advanced tumor stages (P < 0.001), positive lymph nodal status (P < 0.001), decreased E-cadherin expression (P < 0.001), and high lymphovascular density (LVD) (P < 0.001). Patients' progression free survival (PFS) and overall survival (OS) were shorter with high ROR1 expression. High ROR1 expression, high LVD, large tumor size, and adenocarcinoma histopathological type were independent risk factors for OS in lung carcinoma patients. High ROR1 expression is associated with poor prognostic parameters in lung carcinoma patients including higher grade, advanced stages, high LVD, epithelial mesenchymal transition, as well as decreased PFS and OS.
Collapse
Affiliation(s)
| | - Sara A Darwish
- Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Tanta University, Egypt
| | | |
Collapse
|
10
|
Meng S, Li M, Qin L, Lv J, Wu D, Zheng D, Jia H, Chen D, Wu Q, Long Y, Tang Z, Tang Y, Yang L, Yao Y, Luo X, Li P. The onco-embryonic antigen ROR1 is a target of chimeric antigen T cells for colorectal cancer. Int Immunopharmacol 2023; 121:110402. [PMID: 37301125 DOI: 10.1016/j.intimp.2023.110402] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/18/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023]
Abstract
Colorectal cancer is globally ranked second in both incidence and mortality rate. It usually develops during the middle or late stages of diagnosis, and is characterized by easy metastasis, poor prognosis, and a significant decline in postoperative quality of life. ROR1 is an excellent oncoembryonic antigen in numerous immunotherapy treatments for tumors. Additionally, it is overexpressed in colorectal cancer. To fill the void in CRC treatment with ROR1 as a target of CAR-T immunotherapy, we designed and prepared antiROR1-CART. This third-generation CAR-T cell can effectively inhibit the growth of colorectal cancer in vitro and in vivo.
Collapse
Affiliation(s)
- Shangsen Meng
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ming Li
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China; Department of Health Sciences, Institute of Physical Science and Information Technology, Anhui University, Hefei, China
| | - Le Qin
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jiang Lv
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, China
| | - Di Wu
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Diwei Zheng
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Heng Jia
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China; Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Dongmei Chen
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Qiting Wu
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Youguo Long
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhaoyang Tang
- Guangdong Zhaotai In Vivo Biomedicine Co. Ltd., Guangzhou 510700, China
| | - Yanlai Tang
- Department of Paediatrics, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Lihua Yang
- Department of Paediatrics, Zhujiang Hospital, Southern China Medical University, Guangzhou, Guangdong 510280, China
| | - Yao Yao
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xuequn Luo
- Department of Paediatrics, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China.
| | - Peng Li
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, China; Department of Surgery of the Faculty of Medicine, The Chinese University of Hong Kong (CUHK), China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China.
| |
Collapse
|
11
|
Lee KJ, Kim NH, Kim HS, Kim Y, Lee JJ, Kim JH, Cho HY, Jeong SY, Park ST. The Role of ROR1 in Chemoresistance and EMT in Endometrial Cancer Cells. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59050994. [PMID: 37241228 DOI: 10.3390/medicina59050994] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023]
Abstract
Background and Objectives: Receptor tyrosine kinase-like orphan receptor type 1 (ROR1) plays a critical role in embryogenesis and is overexpressed in many malignant cells. These characteristics allow ROR1 to be a potential new target for cancer treatment. The aim of this study was to investigate the role of ROR1 through in vitro experiments in endometrial cancer cell lines. Materials and Methods: ROR1 expression was identified in endometrial cancer cell lines using Western blot and RT-qPCR. The effects of ROR1 on cell proliferation, invasion, migration, and epithelial-mesenchymal transition (EMT) markers were analyzed in two endometrial cancer cell lines (HEC-1 and SNU-539) using either ROR1 silencing or overexpression. Additionally, chemoresistance was examined by identifying MDR1 expression and IC50 level of paclitaxel. Results: The ROR1 protein and mRNA were highly expressed in SNU-539 and HEC-1 cells. High ROR1 expression resulted in a significant increase in cell proliferation, migration, and invasion. It also resulted in a change of EMT markers expression, a decrease in E-cadherin expression, and an increase in Snail expression. Moreover, cells with ROR1 overexpression had a higher IC50 of paclitaxel and significantly increased MDR1 expression. Conclusions: These in vitro experiments showed that ROR1 is responsible for EMT and chemoresistance in endometrial cancer cell lines. Targeting ROR1 can inhibit cancer metastasis and may be a potential treatment method for patients with endometrial cancer who exhibit chemoresistance.
Collapse
Affiliation(s)
- Kyung-Jun Lee
- Institute of New Frontier Research Team, Hallym University, Chuncheon 24252, Republic of Korea
| | - Nam-Hyeok Kim
- Institute of New Frontier Research Team, Hallym University, Chuncheon 24252, Republic of Korea
| | - Hyeong Su Kim
- Institute of New Frontier Research Team, Hallym University, Chuncheon 24252, Republic of Korea
- Division of Hemato-Oncology, Department of Internal Medicine, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| | - Youngmi Kim
- Institute of New Frontier Research Team, Hallym University, Chuncheon 24252, Republic of Korea
| | - Jae-Jun Lee
- Institute of New Frontier Research Team, Hallym University, Chuncheon 24252, Republic of Korea
- Department of Anesthesiology and Pain Medicine, Chuncheon Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Chuncheon 24253, Republic of Korea
| | - Jung Han Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| | - Hye-Yon Cho
- Institute of New Frontier Research Team, Hallym University, Chuncheon 24252, Republic of Korea
- Department of Obstetrics and Gynecology, Dongtan Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Hwaseong 18450, Republic of Korea
| | - Soo Young Jeong
- Institute of New Frontier Research Team, Hallym University, Chuncheon 24252, Republic of Korea
- Department of Obstetrics and Gynecology, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| | - Sung Taek Park
- Institute of New Frontier Research Team, Hallym University, Chuncheon 24252, Republic of Korea
- Department of Obstetrics and Gynecology, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| |
Collapse
|
12
|
Ghaderi A, Okhovat MA, Lehto J, De Petris L, Manouchehri Doulabi E, Kokhaei P, Zhong W, Rassidakis GZ, Drakos E, Moshfegh A, Schultz J, Olin T, Österborg A, Mellstedt H, Hojjat-Farsangi M. A Small Molecule Targeting the Intracellular Tyrosine Kinase Domain of ROR1 (KAN0441571C) Induced Significant Apoptosis of Non-Small Cell Lung Cancer (NSCLC) Cells. Pharmaceutics 2023; 15:pharmaceutics15041148. [PMID: 37111634 PMCID: PMC10145660 DOI: 10.3390/pharmaceutics15041148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 04/09/2023] Open
Abstract
The ROR1 receptor tyrosine kinase is expressed in embryonic tissues but is absent in normal adult tissues. ROR1 is of importance in oncogenesis and is overexpressed in several cancers, such as NSCLC. In this study, we evaluated ROR1 expression in NSCLC patients (N = 287) and the cytotoxic effects of a small molecule ROR1 inhibitor (KAN0441571C) in NSCLC cell lines. ROR1 expression in tumor cells was more frequent in non-squamous (87%) than in squamous (57%) carcinomas patients, while 21% of neuroendocrine tumors expressed ROR1 (p = 0.0001). A significantly higher proportion of p53 negative patients in the ROR1+ group than in the p53 positive non-squamous NSCLC patients (p = 0.03) was noted. KAN0441571C dephosphorylated ROR1 and induced apoptosis (Annexin V/PI) in a time- and dose-dependent manner in five ROR1+ NSCLC cell lines and was superior compared to erlotinib (EGFR inhibitor). Apoptosis was confirmed by the downregulation of MCL-1 and BCL-2, as well as PARP and caspase 3 cleavage. The non-canonical Wnt pathway was involved. The combination of KAN0441571C and erlotinib showed a synergistic apoptotic effect. KAN0441571C also inhibited proliferative (cell cycle analyses, colony formation assay) and migratory (scratch wound healing assay) functions. Targeting NSCLC cells by a combination of ROR1 and EGFR inhibitors may represent a novel promising approach for the treatment of NSCLC patients.
Collapse
Affiliation(s)
- Amineh Ghaderi
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Mohammad-Ali Okhovat
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Jemina Lehto
- Kancera AB, Nanna Svartz Väg 4, 171 65 Solna, Sweden
| | - Luigi De Petris
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna, Karolinska Institutet, 171 64 Stockholm, Sweden
- Thoracic Oncology Center, Karolinska Comprehensive Cancer Center, 171 76 Solna, Sweden
| | - Ehsan Manouchehri Doulabi
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Parviz Kokhaei
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna, Karolinska Institutet, 171 64 Stockholm, Sweden
- Department of Immunology, Arak University of Medical Sciences, Arak 3848170001, Iran
| | - Wen Zhong
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Georgios Z. Rassidakis
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Elias Drakos
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna, Karolinska Institutet, 171 64 Stockholm, Sweden
- Department of Pathology, Medical School, University of Crete, 700 13 Heraklion, Greece
| | - Ali Moshfegh
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna, Karolinska Institutet, 171 64 Stockholm, Sweden
- Kancera AB, Nanna Svartz Väg 4, 171 65 Solna, Sweden
| | - Johan Schultz
- Kancera AB, Nanna Svartz Väg 4, 171 65 Solna, Sweden
| | - Thomas Olin
- Kancera AB, Nanna Svartz Väg 4, 171 65 Solna, Sweden
| | - Anders Österborg
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna, Karolinska Institutet, 171 64 Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital Solna, 171 64 Solna, Sweden
| | - Håkan Mellstedt
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Mohammad Hojjat-Farsangi
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna, Karolinska Institutet, 171 64 Stockholm, Sweden
| |
Collapse
|
13
|
Jahangirian E, Zargan J, Rabbani H, Zamani J. Investigating the inhibitory and penetrating properties of three novel anticancer and antimicrobial scorpion peptides via molecular docking and molecular dynamic simulation. J Biomol Struct Dyn 2023; 41:15354-15385. [PMID: 36927377 DOI: 10.1080/07391102.2023.2188956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/28/2023] [Indexed: 03/18/2023]
Abstract
The two types of bladder cancer, muscle invasive and non-muscle invasive (NMIBC), are among the most prevalent cancers worldwide. Despite this, even though muscle-invasive bladder cancer is more deadly, NMIBC requires more therapy due to a greater recurrence rate and more extended and expensive care. Immunotherapy, intravesical chemotherapy, cystoscopy, and transurethral resection (TUR) are among the treatments available. Crude scorpion venomand purified proteins and peptides, can suppress cancer metastasis in an in vitro or in vivo context, suppress cancer growth, halt the cell cycle, and cause cell apoptosis, according to an increasing number of experimental and preclinical studies. In this research, three novels discovered peptides (P2, P3 and P4. ProteomeXchange: PXD036231) from Buthotus saulcyi and, Odontobuthus doriae scorpions were used along with a peptide called pantinin (as a control). The phylogenetic tree showed that the peptides belong to Chaperonin HSP60, Chrysophsin2 and Pheromone-binding protein2, respectively. These peptides were docked with four known antigens, BAGE, BLCAP, PRAME and ROR1 related to bladder cancer and three bacterial antigens FliC, FliD and FimH to investigate their antimicrobial and anticancer properties. The results showed that peptides 2 and 3 have the best binding rate. The MD simulation results also confirmed the binding of peptides 2 and 3 to antigens. The penetration power of peptides 2 and 3 in the membrane of cancer cells and bacterial cells was also simulated, and the results of RMSD and PD confirmed it. QSAR suggests that peptides 2 and 3 can act as anti-cancer and anti-microbial peptides.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ehsan Jahangirian
- Department of Biology, Faculty of Basic Science, Imam Hossein University, Tehran, Iran
| | - Jamil Zargan
- Department of Biology, Faculty of Basic Science, Imam Hossein University, Tehran, Iran
| | - Hodjattallah Rabbani
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Javad Zamani
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
14
|
Jeong SY, Lee KJ, Cha J, Park SY, Kim HS, Kim JH, Lee JJ, Kim N, Park ST. Meta-Analysis of Survival Effects of Receptor Tyrosine Kinase-like Orphan Receptor 1 (ROR1). MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58121867. [PMID: 36557069 PMCID: PMC9784027 DOI: 10.3390/medicina58121867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Background and Objectives: Identification and targeting of membrane proteins in tumor cells is one of the key steps in the development of cancer drugs. The receptor tyrosine kinase-like orphan receptor (ROR) type 1 is a type-I transmembrane protein expressed in various cancer tissues, which is in contrast to its limited expression in normal tissues. These characteristics make ROR1 a candidate target for cancer treatment. This study aimed to identify the prognostic value of ROR1 expression in cancers. Materials and Methods: We conducted a comprehensive systematic search of electronic databases (PubMed) from their inception to September 2021. The included studies assessed the effect of ROR1 on overall survival (OS) and progression-free survival (PFS). Hazard ratios (HR) from collected data were pooled in a meta-analysis using Revman version 5.4 with generic inverse-variance and random effects modeling. Results: A total of fourteen studies were included in the final analysis. ROR1 was associated with worse OS (HR 1.95, 95% confidence interval (CI) 1.50−2.54; p < 0.001) with heterogeneity. The association between poor OS and ROR1 expression was high in endometrial cancer, followed by ovarian cancer, and diffuse large B cell lymphoma. In addition, ROR1 was associated with poor PFS (HR 1.84, 95% CI 1.60−2.10; p < 0.001), but heterogeneity was not statistically significant. In subgroup analysis, high ROR1 expression showed a significantly higher rate of advanced stage or lymph node metastasis. Conclusions: This meta-analysis provides evidence that ROR1 expression is associated with adverse outcome in cancer survival. This result highlights ROR1 as a target for developmental therapeutics in cancers.
Collapse
Affiliation(s)
- Soo Young Jeong
- Department of Obstetrics and Gynecology, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea
- Institute of New Frontier Research Team, Hallym University, Chuncheon 24252, Republic of Korea
| | - Kyung-jun Lee
- Institute of New Frontier Research Team, Hallym University, Chuncheon 24252, Republic of Korea
| | - Jieum Cha
- Department of Obstetrics and Gynecology, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| | - So Yoon Park
- Department of Obstetrics and Gynecology, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| | - Hyeong Su Kim
- Institute of New Frontier Research Team, Hallym University, Chuncheon 24252, Republic of Korea
- Division of Hemato-Oncology, Department of Internal Medicine, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| | - Jung Han Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| | - Jae-Jun Lee
- Institute of New Frontier Research Team, Hallym University, Chuncheon 24252, Republic of Korea
- Departments of Anesthesiology and Pain Medicine, Chuncheon Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Chuncheon 24253, Republic of Korea
| | - Namhyeok Kim
- Institute of New Frontier Research Team, Hallym University, Chuncheon 24252, Republic of Korea
| | - Sung Taek Park
- Department of Obstetrics and Gynecology, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea
- Institute of New Frontier Research Team, Hallym University, Chuncheon 24252, Republic of Korea
- Correspondence: ; Tel.: +82-2-829-5151; Fax: +82-2-833-5323
| |
Collapse
|
15
|
Flanagan DJ, Woodcock SA, Phillips C, Eagle C, Sansom OJ. Targeting ligand-dependent wnt pathway dysregulation in gastrointestinal cancers through porcupine inhibition. Pharmacol Ther 2022; 238:108179. [PMID: 35358569 PMCID: PMC9531712 DOI: 10.1016/j.pharmthera.2022.108179] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 12/20/2022]
Abstract
Gastrointestinal cancers are responsible for more cancer deaths than any other system of the body. This review summarises how Wnt pathway dysregulation contributes to the development of the most common gastrointestinal cancers, with a particular focus on the nature and frequency of upstream pathway aberrations. Tumors with upstream aberrations maintain a dependency on the presence of functional Wnt ligand, and are predicted to be tractable to inhibitors of Porcupine, an enzyme that plays a key role in Wnt secretion. We summarise available pre-clinical efficacy data from Porcupine inhibitors in vitro and in vivo, as well as potential toxicities and the data from early phase clinical trials. We appraise the rationale for biomarker-defined targeted approaches, as well as outlining future opportunities for combination with other therapeutics.
Collapse
Affiliation(s)
- Dustin J Flanagan
- Cancer Research UK Beatson Institute, Glasgow, UK; Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | | | | | | | - Owen J Sansom
- Cancer Research UK Beatson Institute, Glasgow, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
16
|
The Anti-ROR1 Monoclonal Antibody Zilovertamab Inhibits the Proliferation of Ovarian and Endometrial Cancer Cells. Pharmaceutics 2022; 14:pharmaceutics14040837. [PMID: 35456672 PMCID: PMC9033071 DOI: 10.3390/pharmaceutics14040837] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/29/2022] [Accepted: 04/04/2022] [Indexed: 02/04/2023] Open
Abstract
The non-canonical Wnt signalling receptor ROR1 is aberrantly expressed in numerous cancers, including ovarian and endometrial cancer. We previously reported that silencing ROR1 could inhibit the proliferation and metastatic potential of ovarian and endometrial cancer cells in vitro. Zilovertamab is an ROR1-targeting humanised monoclonal antibody, with demonstrated safety and efficacy in clinical trials of several ROR1-related malignancies. The aim of this study was to investigate the potential of zilovertamab alone, or in combination with commonly utilised gynaecological cancer therapies (cisplatin, paclitaxel and the PARP inhibitor—Olaparib) on high-grade serous ovarian cancer (HGSOC), including models of platinum resistance and homologous recombination deficiency (CaOV3, CaOV3CisR, PEO1 and PEO4) and endometrial cancer (EC) cell lines (Ishikawa and KLE). The effect of zilovertamab (at 25 µg/mL or 50 µg/mL) +/− agents was investigated using the IncuCyte S3 Live Cell imaging system. Zilovertamab alone inhibited the proliferation of HGSOC and EC cells in vitro, including in models of platinum resistance and homologous recombination deficiency. In general, the addition of commonly used chemotherapies to a fixed dose of zilovertamab did not enhance the observed anti-proliferative activity. This study supports the potential of zilovertamab, or other ROR1-targeting therapies, for treating women with HGSOC and EC.
Collapse
|
17
|
Huang G, Mao J. Identification of a 12-Gene Signature and Hub Genes Involved in Kidney Wilms Tumor via Integrated Bioinformatics Analysis. Front Oncol 2022; 12:877796. [PMID: 35480093 PMCID: PMC9038080 DOI: 10.3389/fonc.2022.877796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/07/2022] [Indexed: 01/23/2023] Open
Abstract
Wilms tumor (WT), also known as nephroblastoma, is a rare primary malignancy in all kinds of tumor. With the development of second-generation sequencing, the discovery of new tumor markers and potential therapeutic targets has become easier. This study aimed to explore new WT prognostic biomarkers. In this study, WT-miRNA datasets GSE57370 and GSE73209 were selected for expression profiling to identify differentially expressed genes. The key gene miRNA, namely hsa-miR-30c-5p, was identified by overlapping, and the target gene of candidate hsa-miR-30c-5p was predicted using an online database. Furthermore, 384 genes were obtained by intersecting them with differentially expressed genes in the TARGET-WT database, and the genes were analyzed for pathway and functional enrichment. Kaplan–Meier survival analysis of the 384 genes yielded a total of 25 key genes associated with WT prognosis. Subsequently, a prediction model with 12 gene signatures (BCL6, CCNA1, CTHRC1, DGKD, EPB41L4B, ERRFI1, LRRC40, NCEH1, NEBL, PDSS1, ROR1, and RTKN2) was developed. The model had good predictive power for the WT prognosis at 1, 3, and 5 years (AUC: 0.684, 0.762, and 0.774). Finally, ERRFI1 (hazard ratios [HR] = 1.858, 95% confidence intervals [CI]: 1.298–2.660) and ROR1 (HR = 0.780, 95% CI: 0.609–0.998) were obtained as independent predictors of prognosis in WT patients by single, multifactorial Cox analysis.
Collapse
|
18
|
Carlsen L, Schorl C, Huntington K, Hernandez-Borrero L, Jhaveri A, Zhang S, Zhou L, El-Deiry WS. Pan-drug and drug-specific mechanisms of 5-FU, irinotecan (CPT-11), oxaliplatin, and cisplatin identified by comparison of transcriptomic and cytokine responses of colorectal cancer cells. Oncotarget 2021; 12:2006-2021. [PMID: 34611476 PMCID: PMC8487728 DOI: 10.18632/oncotarget.28075] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 08/28/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) caused over 900,000 deaths worldwide in 2020. A majority of late-stage CRC patients are treated with 5-fluorouracil (5-FU) combined with either irinotecan (CPT-11), oxaliplatin, or both. Despite their widespread use, the mechanisms of efficacy and toxicity of these drugs remain incompletely understood. While previous work has investigated cellular responses to these agents individually, we directly compare the transcriptomic and cytokine profiles of HCT116 wild-type and p53-/- colorectal cancer cells treated with these drugs and report pan-drug, drug-specific, drug class-specific, p53-independent, and p53-dependent signatures. We observed downregulation of histone genes by 5-FU (that significantly correlates with improved survival in CRC patients) and upregulation of FOS and ATF3 by oxaliplatin (which may contribute to peripheral neuropathy). BTG2 was identified as a top gene upregulated by all four drugs, suggesting its critical role in the cellular response to chemotherapy in CRC. Soluble TRAILR2 (death receptor 5; DR5) is a decoy receptor for TRAIL, an apoptosis-inducing cytokine. TRAILR2 was down-regulated by oxaliplatin and 5-FU, was not affected by CPT-11, and was increased by cisplatin. There was an increase in IL-8 by oxaliplatin and increase in ferritin by cisplatin which may contribute to cancer cell survival. Novel drug-specific mechanisms of efficacy or toxicity identified in these signatures may be targeted with combination therapies or development of new targeted therapies. Together, the findings here contribute to our understanding of the molecular bases of efficacy and toxicity of chemotherapeutic agents often used for treatment of GI cancer such as CRC.
Collapse
Affiliation(s)
- Lindsey Carlsen
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, USA.,Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,Pathobiology Graduate Program, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Christoph Schorl
- The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, USA.,Department of Molecular Biology, Cell Biology and Biochemistry, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,Genomics Core Facility, Brown University, Providence, RI 02903, USA.,Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Kelsey Huntington
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, USA.,Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,Pathobiology Graduate Program, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Liz Hernandez-Borrero
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, USA.,Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,Pathobiology Graduate Program, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Aakash Jhaveri
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Shengliang Zhang
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, USA.,Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Lanlan Zhou
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, USA.,Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,The Joint Program in Cancer Biology, Brown University and the Lifespan Health System, Providence, RI 02903, USA.,Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,Pathobiology Graduate Program, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA.,Hematology-Oncology Division, Department of Medicine, Rhode Island Hospital and Brown University, Providence, RI 02903, USA.,Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| |
Collapse
|
19
|
Abolhasan R, Khalilzadeh B, Yousefi H, Samemaleki S, Chakari-Khiavi F, Ghorbani F, Pourakbari R, Kamrani A, Khataee A, Rad TS, Rashidi MR, Yousefi M, AghebatiMaleki L. Ultrasensitive and label free electrochemical immunosensor for detection of ROR1 as an oncofetal biomarker using gold nanoparticles assisted LDH/rGO nanocomposite. Sci Rep 2021; 11:14921. [PMID: 34290319 PMCID: PMC8295321 DOI: 10.1038/s41598-021-94380-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 06/30/2021] [Indexed: 11/09/2022] Open
Abstract
In the present article, we developed a highly sensitive label-free electrochemical immunosensor based on NiFe-layered double hydroxides (LDH)/reduced graphene oxide (rGO)/gold nanoparticles modified glassy carbon electrode for the determination of receptor tyrosine kinase-like orphan receptor (ROR)-1. In this electrochemical immunoassay platform, NiFe-LDH/rGO was used due to great electron mobility, high specific surface area and flexible structures, while Au nanoparticles were prepared and coated on the modified electrodes to improve the detection sensitivity and ROR1 antibody immobilizing (ROR1Ab). The modification procedure was approved by using cyclic voltammetry and differential pulse voltammetry based on the response of peak current to the step by step modifications. Under optimum conditions, the experimental results showed that the immunosensor revealed a sensitive response to ROR1 in the range of 0.01-1 pg mL-1, and with a lower limit of quantification of 10 attogram/mL (10 ag mL-1). Furthermore, the designed immunosensor was applied for the analysis of ROR1 in several serum samples of chronic lymphocytic leukemia suffering patients with acceptable results, and it also exhibited good selectivity, reproducibility and stability.
Collapse
Affiliation(s)
- Rozita Abolhasan
- Stem Cell Research Center (SCRC), Tabriz University of Medical Sciences, 51664-14766, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Balal Khalilzadeh
- Stem Cell Research Center (SCRC), Tabriz University of Medical Sciences, 51664-14766, Tabriz, Iran. .,Biosensor Sciences and Technologies Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Hadi Yousefi
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| | - Sahar Samemaleki
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Forough Chakari-Khiavi
- Pharmaceutical Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farzaneh Ghorbani
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Pourakbari
- Stem Cell Research Center (SCRC), Tabriz University of Medical Sciences, 51664-14766, Tabriz, Iran
| | - Amin Kamrani
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Khataee
- Research Laboratory of Advanced Water and Wastewater Treatment Processes, Department of Applied Chemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran.,Department of Environmental Engineering, Gebze Technical University, 41400, Gebze, Turkey.,Department of Materrial Science and Physical Chemistry of Materials, South Ural State University, 454080, Chelyabinsk, Russian Federation
| | - Tannaz Sadeghi Rad
- Research Laboratory of Advanced Water and Wastewater Treatment Processes, Department of Applied Chemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Mohammad Reza Rashidi
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leili AghebatiMaleki
- Stem Cell Research Center (SCRC), Tabriz University of Medical Sciences, 51664-14766, Tabriz, Iran. .,Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran. .,Immunology Research Center, Tabriz University of Medical Sciences, PO Box 6446-14155, Tabriz, Iran.
| |
Collapse
|
20
|
Zhao Y, Zhang D, Guo Y, Lu B, Zhao ZJ, Xu X, Chen Y. Tyrosine Kinase ROR1 as a Target for Anti-Cancer Therapies. Front Oncol 2021; 11:680834. [PMID: 34123850 PMCID: PMC8193947 DOI: 10.3389/fonc.2021.680834] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022] Open
Abstract
Receptor tyrosine kinase ROR1 plays an essential role in embryogenesis and is overexpressed in many types of malignant tumors. Studies have demonstrated that it plays an important role in oncogenesis by activating cell survival signaling events, particularly the non-canonical WNT signaling pathway. Antibody-based immunotherapies targeting ROR1 have been developed and evaluated in preclinical and clinical studies with promising outcomes. However, small molecule inhibitors targeting ROR1 are underappreciated because of the initial characterization of ROR1 as a peusdokinase. The function of ROR1 as a tyrosine kinase remains poorly understood, although accumulating evidence have demonstrated its intrinsic tyrosine kinase activity. In this review, we analyzed the structural and functional features of ROR1 and discussed therapeutic strategies targeting this kinase.
Collapse
Affiliation(s)
- Yuming Zhao
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Dengyang Zhang
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yao Guo
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Bo Lu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Zhizhuang Joe Zhao
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Xiaojun Xu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yun Chen
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| |
Collapse
|
21
|
Nema S, Kallianpur S, Kumar A, Nema R, Vishwakarma S, Nema SK. Do patients with oral squamous cell carcinoma express receptor tyrosine kinase-like orphan receptor 1? Results of an observational study. J Oral Maxillofac Pathol 2021; 25:105-109. [PMID: 34349419 PMCID: PMC8272474 DOI: 10.4103/jomfp.jomfp_293_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 09/25/2020] [Accepted: 10/16/2020] [Indexed: 12/14/2022] Open
Abstract
Context The receptor tyrosine kinase-like orphan receptor 1 (ROR1) is a transmembrane protein of the receptor tyrosine kinase family. The expression of ROR1 has been linked to cancers. Aims This study aimed to investigate the expression of ROR1 in oral squamous cell carcinoma (OSCC). Settings and Design This prospective observational study was conducted at a tertiary referral center for treatment of oral carcinoma from November 2013 to December 2016. Subjects and Methods One-step quantitative reverse transcription-polymerase chain reaction (30 oral cancer tissues and ten normal oral tissue samples) was performed to characterize the expression of the ROR1 gene in oral cancer. Statistical Analysis Used Analyses of all tumor samples were carried out at least twice, and the mean value was calculated. The differences in ROR1 messenger RNA (mRNA) expression between OSCC tissue and nontumorous gingival tissue was statistically analyzed using Mann-Whitney U-test. The correlations between the clinicopathological parameters and ROR1 mRNA expression were analyzed using Kruskal-Wallis test χ2 value. Results There were 17, 5, 3 and 1 cases of OSCC of buccal mucosa, tongue and lower alveolus lip, respectively. Nearly 88.5% of cases had a history of tobacco consumption. The most common OSCC type was T2N1M0. There was no difference in ROR1 fold change between controls and cases (P = 0.06), but there was a trend for downregulation of ROR1 expression from controls to cases. Subgroup analysis revealed the downregulation of ROR1 expression in controls versus Grade II that was significant (P = 0.04). Conclusions There was no change in the expression of ROR1 between cases and controls. A study involving a larger sample size needs to be formulated and conducted for investigating the relation between expression and regulation of ROR1 in OSCC.
Collapse
Affiliation(s)
- Swati Nema
- Department of Oral Pathology and Microbiology, Peoples College of Dental Sciences Peoples University, Bhopal, Madhya Pradesh, India
| | - Shreenivas Kallianpur
- Department of Oral Pathology and Microbiology, Peoples College of Dental Sciences Peoples University, Bhopal, Madhya Pradesh, India
| | - Ashok Kumar
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Rajeev Nema
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Supriya Vishwakarma
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Sandeep Kumar Nema
- Associate Professor of Orthopedics Jawaharlal Institute of Postgraduate Medical Education and Research Pondicherry, Pondicherry, India
| |
Collapse
|
22
|
Lopez-Bergami P, Barbero G. The emerging role of Wnt5a in the promotion of a pro-inflammatory and immunosuppressive tumor microenvironment. Cancer Metastasis Rev 2021; 39:933-952. [PMID: 32435939 DOI: 10.1007/s10555-020-09878-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Wnt5a is the prototypical activator of the non-canonical Wnt pathways, and its overexpression has been implicated in the progression of several tumor types by promoting cell motility, invasion, EMT, and metastasis. Recent evidences have revealed a novel role of Wnt5a in the phosphorylation of the NF-κB subunit p65 and the activation of the NF-κB pathway in cancer cells. In this article, we review the molecular mechanisms and mediators defining a Wnt5a/NF-κB signaling pathway and propose that the aberrant expression of Wnt5a in some tumors drives a Wnt5a/NF-κB/IL-6/STAT3 positive feedback loop that amplifies the effects of Wnt5a. The evidences discussed here suggest that Wnt5a has a double effect on the tumor microenvironment. First, it activates an autocrine ROR1/Akt/p65 pathway that promotes inflammation and chemotaxis of immune cells. Then, Wnt5a activates a TLR/MyD88/p50 pathway exclusively in myelomonocytic cells promoting the synthesis of the anti-inflammatory cytokine IL-10 and a tolerogenic phenotype. As a result of these mechanisms, Wnt5a plays a negative role on immune cell function that contributes to an immunosuppressive tumor microenvironment and would contribute to resistance to immunotherapy. Finally, we summarized the development of different strategies targeting either Wnt5a or the Wnt5a receptor ROR1 that can be helpful for cancer therapy by contributing to generate a more immunostimulatory tumor microenvironment.
Collapse
Affiliation(s)
- Pablo Lopez-Bergami
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimonides, Hidalgo 775, Buenos Aires, Argentina. .,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| | - Gastón Barbero
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimonides, Hidalgo 775, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
23
|
Menck K, Heinrichs S, Baden C, Bleckmann A. The WNT/ROR Pathway in Cancer: From Signaling to Therapeutic Intervention. Cells 2021; 10:cells10010142. [PMID: 33445713 PMCID: PMC7828172 DOI: 10.3390/cells10010142] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
The WNT pathway is one of the major signaling cascades frequently deregulated in human cancer. While research had initially focused on signal transduction centered on β-catenin as a key effector activating a pro-tumorigenic transcriptional response, nowadays it is known that WNT ligands can also induce a multitude of β-catenin-independent cellular pathways. Traditionally, these comprise WNT/planar cell polarity (PCP) and WNT/Ca2+ signaling. In addition, signaling via the receptor tyrosine kinase-like orphan receptors (RORs) has gained increasing attention in cancer research due to their overexpression in a multitude of tumor entities. Active WNT/ROR signaling has been linked to processes driving tumor development and progression, such as cell proliferation, survival, invasion, or therapy resistance. In adult tissue, the RORs are largely absent, which has spiked the interest in them for targeted cancer therapy. Promising results in preclinical and initial clinical studies are beginning to unravel the great potential of such treatment approaches. In this review, we summarize seminal findings on the structure and expression of the RORs in cancer, their downstream signaling, and its output in regard to tumor cell function. Furthermore, we present the current clinical anti-ROR treatment strategies and discuss the state-of-the-art, as well as the challenges of the different approaches.
Collapse
Affiliation(s)
- Kerstin Menck
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, 48149 Münster, Germany; (K.M.); (S.H.); (C.B.)
- West German Cancer Center, University Hospital Münster, 48149 Münster, Germany
| | - Saskia Heinrichs
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, 48149 Münster, Germany; (K.M.); (S.H.); (C.B.)
- West German Cancer Center, University Hospital Münster, 48149 Münster, Germany
| | - Cornelia Baden
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, 48149 Münster, Germany; (K.M.); (S.H.); (C.B.)
- West German Cancer Center, University Hospital Münster, 48149 Münster, Germany
| | - Annalen Bleckmann
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, 48149 Münster, Germany; (K.M.); (S.H.); (C.B.)
- West German Cancer Center, University Hospital Münster, 48149 Münster, Germany
- Department of Hematology/Medical Oncology, University Medical Center Göttingen, 37099 Göttingen, Germany
- Correspondence: ; Tel.: +49-0251-8352712
| |
Collapse
|
24
|
Bayat AA, Sadeghi N, Fatemi R, Nowroozi MR, Ohadian Moghadam S, Borzuee M, Radmanesh A, Khodadoost M, Sarrafzadeh AR, Zarei O, Rabbani H. Monoclonal Antibody Against ROR1 Induces Apoptosis in Human Bladder Carcinoma Cells. Avicenna J Med Biotechnol 2020; 12:165-171. [PMID: 32695279 PMCID: PMC7368111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 03/16/2020] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Receptor tyrosine kinase-like Orphan Receptor 1 (ROR1) is one of the promising cell surface antigens for targeting cancer cells. The aim of this study was to evaluate ROR1 cell surface expression in bladder cancer cells using a murine anti-ROR1 monoclonal antibody (mAb) called 5F1-B10 as well as investigate its potential in apoptosis induction. METHODS Expression of ROR1 in two human bladder cell lines, 5637 and EJ138, as well as a non-cancerous human cell line, Human Fetal Foreskin Fibroblast (HFFF), was examined by flow cytometry and immunocytochemistry. Immunohistochemical staining of cancer and normal bladder tissues was also performed. RESULTS The flow cytometry results showed that 5F1-B10 mAb could recognize ROR1 molecules in 86.1% and 45.6% of 5637 and EJ138 cells, respectively. The expression level of ROR1 was 5.49% in HFFF cells. The immunocytochemistry and immunohistochemistry staining results also confirmed the presence of ROR1 on the surface of both bladder cancer cells and tissues, respectively. The obtained data from apoptosis assay demonstrated that 5F1-B10 mAb could induce apoptosis in both 5637 and EJ138 cell lines. CONCLUSION Taken together, our finding indicates the role of ROR1 in bladder cancer cell survival and suggests this receptor might be a promising target for developing novel therapeutic agents against bladder carcinoma.
Collapse
Affiliation(s)
- Ali-Ahmad Bayat
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Niloufar Sadeghi
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Ramina Fatemi
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | | | | | - Mohadeseh Borzuee
- Uro-Oncology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amin Radmanesh
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Mahmood Khodadoost
- Faculty of Traditional Medicine, Traditional Medicine and Materia Medica Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Omid Zarei
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Hodjattallah Rabbani
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| |
Collapse
|
25
|
Zhou Q, Zhou S, Wang H, Li Y, Xiao X, Yang J. Stable silencing of ROR1 regulates cell cycle, apoptosis, and autophagy in a lung adenocarcinoma cell line. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:1108-1120. [PMID: 32509086 PMCID: PMC7270672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 04/14/2020] [Indexed: 06/11/2023]
Abstract
Lung cancer has the highest mortality and recurrence rate among cancers in the world. Receptor tyrosine kinase-like orphan receptor 1 (ROR1) has been widely recognized for its role in promoting the growth and metastasis of lung cancer, but its comprehensive role and molecular mechanisms in regulating cell cycle, apoptosis, and autophagy remain unclear. In this study, a series of ROR1-stably silenced monoclonal clones from lung adenocarcinoma cell lines PC9, PC9erlo, and NCI-H1975 were successfully selected and confirmed by qRT-PCR, western blot, and flow cytometry, and used as cell models in the following assays. Our study clearly shows that blocking ROR1 significantly downregulates cell cycle-inducing molecules such as CDK4 and Cyclin E1, and anti-apoptotic molecules such as Bcl-XL and Bcl-2, while it markedly upregulates pro-apoptotic molecules such as Bak, Caspase-3, and Caspase-7, which extends our previous observation on the molecular mechanism of ROR1-mediated tumor growth in lung adenocarcinoma. Our data also show that silencing ROR1 promotes autophagy since the key molecules involved in autophagy including ATG7, ATG12, BNIP3L, LC3A, LC3B, and NBS1 were up-regulated. We further screened key phosphokinase signaling pathways downstream of ROR1 in lung adenocarcinoma by a human phospho-kinase array. Our data indicate that blocking ROR1 could deactivate Akt, then activate GSK-3α/β by de-phosphorylation, and finally deactivate mTOR. In this way blocking ROR1 could effectively regulate the cell cycle, apoptosis, and autophagy in lung cancer.
Collapse
Affiliation(s)
- Qi Zhou
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine Chengdu 611137, Sichuan, P. R. China
| | - Shiyi Zhou
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine Chengdu 611137, Sichuan, P. R. China
| | - Huili Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine Chengdu 611137, Sichuan, P. R. China
| | - Yanping Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine Chengdu 611137, Sichuan, P. R. China
| | - Xiaoqian Xiao
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine Chengdu 611137, Sichuan, P. R. China
| | - Jiahui Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine Chengdu 611137, Sichuan, P. R. China
| |
Collapse
|
26
|
Tsubaki M, Takeda T, Noguchi M, Jinushi M, Seki S, Morii Y, Shimomura K, Imano M, Satou T, Nishida S. Overactivation of Akt Contributes to MEK Inhibitor Primary and Acquired Resistance in Colorectal Cancer Cells. Cancers (Basel) 2019; 11:cancers11121866. [PMID: 31769426 PMCID: PMC6966459 DOI: 10.3390/cancers11121866] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/21/2019] [Accepted: 11/21/2019] [Indexed: 12/21/2022] Open
Abstract
RAS and BRAF-mutated colorectal cancers are associated with resistance to chemotherapy and poor prognosis, highlighting the need for new therapeutic strategies. Although these cancers sometimes respond to mitogen activated protein kinase kinase (MEK) inhibitor treatment, they often acquire resistance via mechanisms, which are poorly understood. Here, we investigated the mechanism of MEK inhibitor resistance in primary- and acquired-resistant cells. Cell viability was examined using the trypan blue dye exclusion assay. Protein expression was analyzed by western blotting. Somatic mutations in colorectal cancer cells were investigated using the polymerase chain reaction array. PD0325901 and trametinib induced cell death in LoVo and Colo-205 cells but not in DLD-1 and HT-29 cells, which have a PIK3CA mutation constitutively activating Akt and NF-κB. Treatment with PD0325901 and trametinib suppressed ERK1/2 activation in all four cell lines but only induced Akt and NF-κB activation in DLD-1 and HT-29 cells. Inhibition of Akt but not NF-κB, overcame MEK inhibitor resistance in DLD-1 and HT-29 cells. Acquired-resistant LoVo/PR, Colo-205/PR and LoVo/TR cells have constitutively active Akt due to a M1043V mutation in the kinase activation loop of PIK3CA and Akt inhibitor resensitized these cells to MEK inhibitor. These results demonstrate that the overactivation of Akt plays a critical role in MEK inhibitor primary and acquired resistance and implicate combined Akt/MEK inhibition as a potentially useful treatment for RAS/BRAF-mutated colorectal cancer.
Collapse
Affiliation(s)
- Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
| | - Tomoya Takeda
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
| | - Masaki Noguchi
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
| | - Minami Jinushi
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
| | - Shiori Seki
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
| | - Yuusuke Morii
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
- Department of Phamacy, Municipal Ikeda Hospital, Ikeda, Osaka 563-8510, Japan;
| | - Kazunori Shimomura
- Department of Phamacy, Municipal Ikeda Hospital, Ikeda, Osaka 563-8510, Japan;
| | - Motohiro Imano
- Department of Surgery, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-0014, Japan;
| | - Takao Satou
- Department of Pathology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-0014, Japan.;
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
- Correspondence:
| |
Collapse
|
27
|
Wang L, Cho KB, Li Y, Tao G, Xie Z, Guo B. Long Noncoding RNA (lncRNA)-Mediated Competing Endogenous RNA Networks Provide Novel Potential Biomarkers and Therapeutic Targets for Colorectal Cancer. Int J Mol Sci 2019; 20:E5758. [PMID: 31744051 PMCID: PMC6888455 DOI: 10.3390/ijms20225758] [Citation(s) in RCA: 405] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer and has a high metastasis and reoccurrence rate. Long noncoding RNAs (lncRNAs) play an important role in CRC growth and metastasis. Recent studies revealed that lncRNAs participate in CRC progression by coordinating with microRNAs (miRNAs) and protein-coding mRNAs. LncRNAs function as competitive endogenous RNAs (ceRNAs) by competitively occupying the shared binding sequences of miRNAs, thus sequestering the miRNAs and changing the expression of their downstream target genes. Such ceRNA networks formed by lncRNA/miRNA/mRNA interactions have been found in a broad spectrum of biological processes in CRC, including liver metastasis, epithelial to mesenchymal transition (EMT), inflammation formation, and chemo-/radioresistance. In this review, we summarize typical paradigms of lncRNA-associated ceRNA networks, which are involved in the underlying molecular mechanisms of CRC initiation and progression. We comprehensively discuss the competitive crosstalk among RNA transcripts and the novel targets for CRC prognosis and therapy.
Collapse
Affiliation(s)
- Liye Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX,77204, USA; (K.B.C.); (Y.L.); (G.T.); (Z.X.)
| | | | | | | | | | - Bin Guo
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX,77204, USA; (K.B.C.); (Y.L.); (G.T.); (Z.X.)
| |
Collapse
|
28
|
Miyake N, Ochi N, Yamane H, Fukazawa T, Ikeda T, Yokota E, Takeyama M, Nakagawa N, Nakanishi H, Kohara H, Nagasaki Y, Kawahara T, Ichiyama N, Yamatsuji T, Naomoto Y, Takigawa N. Targeting ROR1 in combination with pemetrexed in malignant mesothelioma cells. Lung Cancer 2019; 139:170-178. [PMID: 31809978 DOI: 10.1016/j.lungcan.2019.10.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 09/24/2019] [Accepted: 10/24/2019] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is overexpressed in a subset of malignant cells. However, it remains unknown whether ROR1 is targetable in malignant mesothelioma (MM). Therefore, in this study, we investigated the effects of ROR1 inhibition in mesothelioma cells. MATERIALS AND METHODS Growth inhibition, colony formation, apoptosis, and mRNA/protein levels using siRNA-transfected MM cells were evaluated. Cluster analysis using Gene Expression Omnibus repository of transcriptomic information was also performed. RESULTS Our results indicated that in three (H2052, H2452, and MESO-1) among four MM cell lines, ROR1 inhibition had anti-proliferative and apoptotic effects and suppressed the activation of AKT and STAT3. Although growth inhibition by siROR1 was minimal in another mesothelioma cell line (H28), colony formation was significantly suppressed. Microarray, quantitative polymerase chain reaction, and Western blot analyses showed that there were differences in the suppression of mRNA and proteins between H2452 and H28 cells transfected with siROR1 compared with those transfected with control siRNA. Cluster analysis further showed that MM tumors had relatively high ROR1 expression, although the cluster in them was different from that in MM cell lines. Thymidylate synthase, a target of pemetrexed, was downregulated in H2452 cells transfected with siROR1. Accordingly, a combination of pemetrexed with siROR1 was found to be effective in the three MM cell lines we studied. CONCLUSION Our findings may provide novel therapeutic insight into the treatment of advanced MM.
Collapse
Affiliation(s)
- Noriko Miyake
- General Medical Center Research Unit, Kawasaki Medical School, 2-6-1 Nakasange, Kita-ku, Okayama 700-8505, Japan
| | - Nobuaki Ochi
- Department of General Internal Medicine 4, Kawasaki Medical School, 2-6-1 Nakasange, Kita-ku, Okayama 700-8505, Japan
| | - Hiromichi Yamane
- Department of General Internal Medicine 4, Kawasaki Medical School, 2-6-1 Nakasange, Kita-ku, Okayama 700-8505, Japan
| | - Takuya Fukazawa
- General Medical Center Research Unit, Kawasaki Medical School, 2-6-1 Nakasange, Kita-ku, Okayama 700-8505, Japan; Department of General Surgery, Kawasaki Medical School, 2-6-1 Nakasange, Kita-ku, Okayama 700-8505, Japan
| | - Tomoko Ikeda
- General Medical Center Research Unit, Kawasaki Medical School, 2-6-1 Nakasange, Kita-ku, Okayama 700-8505, Japan
| | - Etsuko Yokota
- General Medical Center Research Unit, Kawasaki Medical School, 2-6-1 Nakasange, Kita-ku, Okayama 700-8505, Japan
| | - Masami Takeyama
- Department of General Internal Medicine 4, Kawasaki Medical School, 2-6-1 Nakasange, Kita-ku, Okayama 700-8505, Japan
| | - Nozomu Nakagawa
- Department of General Internal Medicine 4, Kawasaki Medical School, 2-6-1 Nakasange, Kita-ku, Okayama 700-8505, Japan
| | - Hidekazu Nakanishi
- Department of General Internal Medicine 4, Kawasaki Medical School, 2-6-1 Nakasange, Kita-ku, Okayama 700-8505, Japan
| | - Hiroyuki Kohara
- Department of General Internal Medicine 4, Kawasaki Medical School, 2-6-1 Nakasange, Kita-ku, Okayama 700-8505, Japan
| | - Yasunari Nagasaki
- Department of General Internal Medicine 4, Kawasaki Medical School, 2-6-1 Nakasange, Kita-ku, Okayama 700-8505, Japan
| | - Tatsuyuki Kawahara
- Department of General Internal Medicine 4, Kawasaki Medical School, 2-6-1 Nakasange, Kita-ku, Okayama 700-8505, Japan
| | - Naruhiko Ichiyama
- Department of General Internal Medicine 4, Kawasaki Medical School, 2-6-1 Nakasange, Kita-ku, Okayama 700-8505, Japan
| | - Tomoki Yamatsuji
- Department of General Surgery, Kawasaki Medical School, 2-6-1 Nakasange, Kita-ku, Okayama 700-8505, Japan
| | - Yoshio Naomoto
- Department of General Surgery, Kawasaki Medical School, 2-6-1 Nakasange, Kita-ku, Okayama 700-8505, Japan
| | - Nagio Takigawa
- General Medical Center Research Unit, Kawasaki Medical School, 2-6-1 Nakasange, Kita-ku, Okayama 700-8505, Japan; Department of General Internal Medicine 4, Kawasaki Medical School, 2-6-1 Nakasange, Kita-ku, Okayama 700-8505, Japan.
| |
Collapse
|
29
|
Deng F, Miller J. A review on protein markers of exosome from different bio-resources and the antibodies used for characterization. J Histotechnol 2019; 42:226-239. [PMID: 31432761 DOI: 10.1080/01478885.2019.1646984] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Exosomes are small membrane vesicles (ranging from 30 nm to 150 nm), secreted by different cell types upon fusion of multivesicular bodies (MVB) to the cell plasma membrane under a variety of normal and pathological conditions. Through transferring their cargos such as proteins, lipids and nucleic acids from donor cells to recipient cells, exosomes play a crucial role in cell-to-cell communication. Due to their presence in most body fluids (such as blood, breast milk, saliva, urine, bile, pancreatic juice, cerebrospinal and peritoneal fluids), and their role in carrying bioactive molecules from the cells of origin, exosomes have attracted great interest in their diagnostic and prognostic value for various diseases and therapeutic approaches. Although a large body of literature has documented the importance of exosomes over the past decade, there is no article systematically summarizing protein markers of exosome from different resources and the antibodies that are suited to characterize exosomes. In this review, we briefly summarize the exosome marker proteins, exosomal biomarkers for different diseases, and the antibodies suitable for different bio-resources exosomes characterization.
Collapse
Affiliation(s)
- Fengyan Deng
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, MO, USA
| | - Josh Miller
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, MO, USA
| |
Collapse
|
30
|
Liu HC, Peng YS, Lee HC. miRDRN-miRNA disease regulatory network: a tool for exploring disease and tissue-specific microRNA regulatory networks. PeerJ 2019; 7:e7309. [PMID: 31404401 PMCID: PMC6688598 DOI: 10.7717/peerj.7309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/17/2019] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND MicroRNA (miRNA) regulates cellular processes by acting on specific target genes, and cellular processes proceed through multiple interactions often organized into pathways among genes and gene products. Hundreds of miRNAs and their target genes have been identified, as are many miRNA-disease associations. These, together with huge amounts of data on gene annotation, biological pathways, and protein-protein interactions are available in public databases. Here, using such data we built a database and web service platform, miRNA disease regulatory network (miRDRN), for users to construct disease and tissue-specific miRNA-protein regulatory networks, with which they may explore disease related molecular and pathway associations, or find new ones, and possibly discover new modes of drug action. METHODS Data on disease-miRNA association, miRNA-target association and validation, gene-tissue association, gene-tumor association, biological pathways, human protein interaction, gene ID, gene ontology, gene annotation, and product were collected from publicly available databases and integrated. A large set of miRNA target-specific regulatory sub-pathways (RSPs) having the form (T, G 1, G 2) was built from the integrated data and stored, where T is a miRNA-associated target gene, G 1 (G 2) is a gene/protein interacting with T (G 1). Each sequence (T, G 1, G 2) was assigned a p-value weighted by the participation of the three genes in molecular interactions and reaction pathways. RESULTS A web service platform, miRDRN (http://mirdrn.ncu.edu.tw/mirdrn/), was built. The database part of miRDRN currently stores 6,973,875 p-valued RSPs associated with 116 diseases in 78 tissue types built from 207 diseases-associated miRNA regulating 389 genes. miRDRN also provides facilities for the user to construct disease and tissue-specific miRNA regulatory networks from RSPs it stores, and to download and/or visualize parts or all of the product. User may use miRDRN to explore a single disease, or a disease-pair to gain insights on comorbidity. As demonstrations, miRDRN was applied: to explore the single disease colorectal cancer (CRC), in which 26 novel potential CRC target genes were identified; to study the comorbidity of the disease-pair Alzheimer's disease-Type 2 diabetes, in which 18 novel potential comorbid genes were identified; and, to explore possible causes that may shed light on recent failures of late-phase trials of anti-AD, BACE1 inhibitor drugs, in which genes downstream to BACE1 whose suppression may affect signal transduction were identified.
Collapse
Affiliation(s)
- Hsueh-Chuan Liu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan
| | - Yi-Shian Peng
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan
| | - Hoong-Chien Lee
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan
- Department of Physics, Chung Yuan Christian University, Zhongli District, Taoyuan City, Taiwan
| |
Collapse
|
31
|
Gorroño-Etxebarria I, Aguirre U, Sanchez S, González N, Escobar A, Zabalza I, Quintana JM, Vivanco MDM, Waxman J, Kypta RM. Wnt-11 as a Potential Prognostic Biomarker and Therapeutic Target in Colorectal Cancer. Cancers (Basel) 2019; 11:E908. [PMID: 31261741 PMCID: PMC6679153 DOI: 10.3390/cancers11070908] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/20/2019] [Accepted: 06/22/2019] [Indexed: 02/05/2023] Open
Abstract
The expression of the secreted factor Wnt-11 is elevated in several types of cancer, including colorectal cancer, where it promotes cancer cell migration and invasion. Analysis of colorectal cancer gene expression databases associated WNT11 mRNA expression with increased likelihood of metastasis in a subset of patients. WNT11 expression was correlated with the expression of the Wnt receptors FZD6, RYK, and PTK7, and the combined expression of WNT11, FZD6 and RYK or PTK7 was associated with an increased risk of 5-year mortality rates. Immunohistochemical analysis of Wnt-11 in a cohort of 357 colorectal cancer patients found significantly higher Wnt-11 levels in tumors, compared with benign tissue. Elevated Wnt-11 levels occurred more frequently in rectal tumors than in colonic tumors and in tumors from women than men. In univariate analysis, increased Wnt-11 expression was also associated with tumor invasion and increased 5-year mortality. High Wnt-11 levels were not associated with high levels of nuclear β-catenin, suggesting Wnt-11 is not simply an indicator for activation of β-catenin-dependent signaling. Expression of Wnt-11 in colorectal cancer cell lines expressing low endogenous Wnt-11 inhibited β-catenin/Tcf activity and increased ATF2-dependent transcriptional activity. WNT11 gene silencing and antibody-mediated inhibition of Wnt-11 in colorectal cancer cell lines expressing high Wnt-11 reduced their capacity for invasion. Together, these observations suggest that Wnt-11 could be a potential target for the treatment of patients with invasive colorectal cancer.
Collapse
Affiliation(s)
| | - Urko Aguirre
- Research Unit, Galdakao-Usansolo Hospital, 48960 Galdakao, Spain
- Kronikgune Institute, Health Services Research on Chronic Patients Network (REDISSEC), 48902 Bilbao, Spain
| | - Saray Sanchez
- Cancer Heterogeneity Lab, CIC bioGUNE, 48160 Derio, Spain
| | - Nerea González
- Research Unit, Galdakao-Usansolo Hospital, 48960 Galdakao, Spain
- Kronikgune Institute, Health Services Research on Chronic Patients Network (REDISSEC), 48902 Bilbao, Spain
| | - Antonio Escobar
- Kronikgune Institute, Health Services Research on Chronic Patients Network (REDISSEC), 48902 Bilbao, Spain
- Research Unit, Basurto University Hospital, Osakidetza, 48013 Bilbao, Spain
| | - Ignacio Zabalza
- Department of Pathology, Galdakao-Usansolo Hospital and Biocruces-Bizkaia Institute, 48960 Galdakao, Spain
| | - José Maria Quintana
- Research Unit, Galdakao-Usansolo Hospital, 48960 Galdakao, Spain
- Kronikgune Institute, Health Services Research on Chronic Patients Network (REDISSEC), 48902 Bilbao, Spain
| | | | - Jonathan Waxman
- Department of Surgery and Cancer, Imperial College London, W12 0NN London, UK
| | - Robert M Kypta
- Cancer Heterogeneity Lab, CIC bioGUNE, 48160 Derio, Spain.
- Department of Surgery and Cancer, Imperial College London, W12 0NN London, UK.
| |
Collapse
|
32
|
Saleh RR, Antrás JF, Peinado P, Pérez-Segura P, Pandiella A, Amir E, Ocaña A. Prognostic value of receptor tyrosine kinase-like orphan receptor (ROR) family in cancer: A meta-analysis. Cancer Treat Rev 2019; 77:11-19. [PMID: 31174180 DOI: 10.1016/j.ctrv.2019.05.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/25/2019] [Accepted: 05/26/2019] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Identification of membrane proteins expressed exclusively on tumor cells is a goal for cancer drug development. The receptor tyrosine kinase-like orphan receptor type 1 and 2 (ROR1/2), are type-I transmembrane proteins expressed in cancer but not in adult normal tissue. Here, we explore the prognostic role ROR1/2 expression on patient outcome. METHODS A systematic search of electronic databases identified publications exploring the effect of ROR1/2 on overall survival (OS). Hazard ratios (HR) from collected data were pooled in a meta-analysis using generic inverse-variance and random effects modeling. Subgroup analyses were conducted based on disease site or tumor type. RESULTS Twenty five studies met the inclusion criteria. ROR1 was associated with worse overall survival (HR 2.13, 95% confidence interval (CI) 1.62-2.80; P < 0.001) with subgroup analysis showing the strongest association between ROR1 and OS was in lung cancer. There was no significant difference between solid tumors and hematological malignancies (HR 2.15, 95% CI 1.52-3.06 vs. HR 2.02, 95% CI 1.46-2.84; subgroup difference P = 0.80). ROR2 was also associated with worse OS (HR 1.84, 95% CI 1.43-2.38; P < 0.001). There was no significant difference between disease sites although the highest association seen was in head and neck cancers (HR 3.19, 95% CI 1.13-8.97) and the lowest in gynecological cancers (HR 1.19, 95% CI 0.71-2.00; subgroup difference P = 0.10). CONCLUSIONS ROR1 and ROR2 expression is associated with adverse outcome in several tumors. ROR1/2 warrants study as a target for developmental therapeutics.
Collapse
Affiliation(s)
- Ramy R Saleh
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, Ontario, Canada
| | - Jesús Fuentes Antrás
- Experimental Therapeutics Unit, Medical Oncology Department. Hospital Clínico San Carlos, and IdISSC, Madrid, Spain
| | - Paloma Peinado
- Experimental Therapeutics Unit, Medical Oncology Department. Hospital Clínico San Carlos, and IdISSC, Madrid, Spain
| | - Pedro Pérez-Segura
- Experimental Therapeutics Unit, Medical Oncology Department. Hospital Clínico San Carlos, and IdISSC, Madrid, Spain
| | - Atanasio Pandiella
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain; CIC-Universidad de Salamanca, Salamanca, Spain
| | - Eitan Amir
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, Ontario, Canada
| | - Alberto Ocaña
- Experimental Therapeutics Unit, Medical Oncology Department. Hospital Clínico San Carlos, and IdISSC, Madrid, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain; Centro Regional de Investigaciones Biomédicas, Castilla-La Mancha University (UCLM), Albacete, Spain.
| |
Collapse
|
33
|
Liu X, Pu W, He H, Fan X, Zheng Y, Zhou JK, Ma R, He J, Zheng Y, Wu K, Zhao Y, Yang SY, Wang C, Wei YQ, Wei XW, Peng Y. Novel ROR1 inhibitor ARI-1 suppresses the development of non-small cell lung cancer. Cancer Lett 2019; 458:76-85. [PMID: 31125641 DOI: 10.1016/j.canlet.2019.05.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 02/05/2023]
Abstract
Limited drug response and severe drug resistance confer the high mortality of non-small-cell lung cancer (NSCLC), a leading cause of cancer death worldwide. There is an urgent need for novel treatment against NSCLC. Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is aberrantly overexpressed and participats in NSCLC development and EGFR-TKIs-induced drug resistance. Increasing evidences indicate that oncogenic ROR1 is a potential target for NSCLC therapy. However, nearly no ROR1 inhibitor was reported until now. Here, combining the computer-aided drug design and cell-based activity screening, we discover (R)-5,7-bis(methoxymethoxy)-2-(4-methoxyphenyl)chroman-4-one (ARI-1) as a novel ROR1 inhibitor. Biological evaluation demonstrates that ARI-1 specifically targets the extracellular frizzled domain of ROR1 and potently suppresses NSCLC cell proliferation and migration by regulating PI3K/AKT/mTOR signaling in a ROR1-dependent manner. Moreover, ARI-1 significantly inhibits tumor growth in vivo without obvious toxicity. Intriguingly, ARI-1 is effective to EGFR-TKIs-resistant NSCLC cells with high ROR1 expression. Therefore, our work suggests that the ROR1 inhibitor ARI-1 is a novel drug candidate for NSCLC treatment, especially for EGFR-TKIs-resisted NSCLC with high ROR1 expression.
Collapse
Affiliation(s)
- Xuesha Liu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenchen Pu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Huaiyu He
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xin Fan
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; College of Life Science, Sichuan University, Chengdu 610065, China
| | - Yuanyuan Zheng
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jian-Kang Zhou
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Rui Ma
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Juan He
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuzhu Zheng
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ke Wu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yun Zhao
- College of Life Science, Sichuan University, Chengdu 610065, China
| | - Sheng-Yong Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chun Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Yu-Quan Wei
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xia-Wei Wei
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yong Peng
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
34
|
Mao Y, Xu L, Wang J, Zhang L, Hou N, Xu J, Wang L, Yang S, Chen Y, Xiong L, Zhu J, Fan W, Xu J. ROR1 associates unfavorable prognosis and promotes lymphoma growth in DLBCL by affecting PI3K/Akt/mTOR signaling pathway. Biofactors 2019; 45:416-426. [PMID: 30801854 DOI: 10.1002/biof.1498] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 01/02/2019] [Accepted: 01/31/2019] [Indexed: 12/23/2022]
Abstract
The receptor-tyrosine-kinase (RTK)-like orphan receptor 1 (ROR1) is a transmembrane glycoprotein regarded as a tumor-associated antigen. ROR1 plays an important role in cancer development, but the detailed function of ROR1 in diffuse large B-cell lymphoma (DLBCL) remains unclear. In this study, we first detected ROR1 expression and evaluated the relationship between ROR1 expression and the clinicopathological characteristics of DLBCL patients. Next we employed shRNA-mediated knockdown of ROR1 in DLBCL cell line to explore the characteristics of ROR1 in DLBCL development both in vitro and in vivo. The results showed a significantly higher level of ROR1 in DLBCL tissues than in lymphatic hyperplasia tissues. High ROR1 expression was correlated with unfavorable prognosis in DLBCL patients. Furthermore, ROR1 knockdown inhibited the growth and induced the apoptosis in DLBCL cells and xenografts. In addition, shROR1 inhibited activation of the PI3K/Akt/mTOR signaling pathway, both in vitro and in vivo. Taken together, our results suggest that ROR1 is a novel prognostic marker for DLBCL survival and ROR1 significantly promotes DLBCL tumorigenesis by regulating the PI3K/Akt/mTOR signaling pathway. Targeting ROR1 may provide a promising strategy for DLBCL treatment. © 2019 BioFactors, 45(3):416-426, 2019.
Collapse
Affiliation(s)
- Yuan Mao
- Department of Hematology and Oncology, Department of Geriatric Lung Cancer Laboratory, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Hospital, Nanjing, China
| | - Li Xu
- Department of Pathology, Jiangsu Cancer Hospital, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jun Wang
- Department of Hematology and Oncology, Department of Geriatric Lung Cancer Laboratory, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Hospital, Nanjing, China
| | - Louqian Zhang
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, Jiangsu, China
| | - Nan Hou
- Department of Hematology and Oncology, Department of Geriatric Lung Cancer Laboratory, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Hospital, Nanjing, China
- Department of Hematology and Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Juqing Xu
- Department of Hematology and Oncology, Department of Geriatric Lung Cancer Laboratory, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Hospital, Nanjing, China
| | - Lin Wang
- Department of Hematology and Oncology, Department of Geriatric Lung Cancer Laboratory, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Hospital, Nanjing, China
| | - Shu Yang
- Department of Hematology and Oncology, Department of Geriatric Lung Cancer Laboratory, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Hospital, Nanjing, China
| | - Yan Chen
- Department of Pathology, Jiangsu Cancer Hospital, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lin Xiong
- Department of Pathology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jin Zhu
- Huadong Medical Institute of Biotechniques, Nanjing, China
| | - Weifei Fan
- Department of Hematology and Oncology, Department of Geriatric Lung Cancer Laboratory, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Hospital, Nanjing, China
| | - Jiaren Xu
- Department of Hematology and Oncology, Department of Geriatric Lung Cancer Laboratory, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Hospital, Nanjing, China
- Department of Hematology and Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
35
|
García-Aranda M, Redondo M. Targeting Receptor Kinases in Colorectal Cancer. Cancers (Basel) 2019; 11:cancers11040433. [PMID: 30934752 PMCID: PMC6521260 DOI: 10.3390/cancers11040433] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 03/19/2019] [Accepted: 03/25/2019] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer is the third most common malignancy in men and the second most common cancer in women. Despite the success of screening programs and the development of adjuvant therapies, the global burden of colorectal cancer is expected to increase by 60% to more than 2.2 million new cases and 1.1 million deaths by 2030. In recent years, a great effort has been made to demonstrate the utility of protein kinase inhibitors for cancer treatment. Considering this heterogeneous disease is defined by mutations that activate different Receptor Tyrosine Kinases (RTKs) and affect downstream components of RTK-activated transduction pathways, in this review we analyze the potential utility of different kinase inhibitors for colorectal cancer treatment.
Collapse
Affiliation(s)
- Marilina García-Aranda
- Research Unit, Hospital Costa del Sol. Autovía A7, km 187. 29603 Marbella, Málaga, Spain.
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC), 28029 Madrid, Spain.
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Málaga, Spain.
| | - Maximino Redondo
- Research Unit, Hospital Costa del Sol. Autovía A7, km 187. 29603 Marbella, Málaga, Spain.
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC), 28029 Madrid, Spain.
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Málaga, Spain.
- Facultad de Medicina, Campus Universitario de Teatinos, Universidad de Málaga, 29010 Málaga, Spain.
| |
Collapse
|
36
|
Zhang R, Wang Z, Yu Q, Shen J, He W, Zhou D, Yu Q, Fan J, Gao S, Duan L. Atractylenolide II reverses the influence of lncRNA XIST/miR-30a-5p/ROR1 axis on chemo-resistance of colorectal cancer cells. J Cell Mol Med 2019; 23:3151-3165. [PMID: 30907503 PMCID: PMC6484310 DOI: 10.1111/jcmm.14148] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/14/2018] [Accepted: 12/20/2018] [Indexed: 01/04/2023] Open
Abstract
This investigation was conducted to elucidate whether atractylenolide II could reverse the role of lncRNA XIST/miR-30a-5p/ROR1 axis in modulating chemosensitivity of colorectal cancer cells. We totally collected 294 pairs of colorectal cancer tissues and adjacent normal tissues and also purchased colorectal cancer cell lines and human embryonic kidney cell line. 5-fluorouracil, cisplatin, mitomycin and adriamycin were designated as the chemotherapies for colorectal cell lines, and atractylenolides were arranged as the Chinese drug. The expressions of XIST, miR-30a-5p and ROR1 were quantified with aid of qRT-PCR or Western blot, and luciferase reporter gene assay was implemented to determine the relationships among XIST, miR-30a-5p and ROR1. Our results demonstrated that XIST and ROR1 expressions were dramatically up-regulated, yet miR-30a-5p expression was down-regulated within colorectal cancer tissues (P < 0.05). The overexpressed XIST and ROR1, as well as under-expressed miR-30a-5p, were inclined to promote viability and proliferation of colorectal cells under the influence of chemo drugs (P < 0.05). In addition, XIST could directly target miR-30a-5p, and ROR1 acted as the targeted molecule of miR-30a-5p. Interestingly, atractylenolides not only switched the expressions of XIST, miR-30a-5p and ROR1 within colorectal cancer cells but also significantly intensified the chemosensitivity of colorectal cancer cells (P < 0.05). Finally, atractylenolide II was discovered to slow down the viability and proliferation of colorectal cancer cells (P < 0.05). In conclusion, the XIST/miR-30a-5p/ROR1 axis could be deemed as pivotal markers underlying colorectal cancer, and administration of atractylenolide II might improve the chemotherapeutic efficacy for colorectal cancer.
Collapse
Affiliation(s)
- Ruijuan Zhang
- Department of Traditional Chinese Medicine, Putuo People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Zhijun Wang
- Department of Traditional Chinese Medicine, Putuo People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Qianyun Yu
- Department of Traditional Chinese Medicine, Wuliqiao Community Health Center of Huangpu District, Shanghai, China
| | - Jun Shen
- Department of Traditional Chinese Medicine, Putuo People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Wenji He
- Department of Traditional Chinese Medicine, Putuo People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Dongqing Zhou
- Department of Traditional Chinese Medicine, Putuo People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Qingqing Yu
- Department of Traditional Chinese Medicine, Putuo People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Jiawei Fan
- Department of Traditional Chinese Medicine, Putuo People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Shurong Gao
- Department of Traditional Chinese Medicine, Putuo People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Lihong Duan
- Department of Rheumatology, Putuo People's Hospital Affiliated to Tongji University, Shanghai, China
| |
Collapse
|
37
|
Townsend MH, Shrestha G, Robison RA, O’Neill KL. The expansion of targetable biomarkers for CAR T cell therapy. J Exp Clin Cancer Res 2018; 37:163. [PMID: 30031396 PMCID: PMC6054736 DOI: 10.1186/s13046-018-0817-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 06/28/2018] [Indexed: 12/13/2022] Open
Abstract
Biomarkers are an integral part of cancer management due to their use in risk assessment, screening, differential diagnosis, prognosis, prediction of response to treatment, and monitoring progress of disease. Recently, with the advent of Chimeric Antigen Receptor (CAR) T cell therapy, a new category of targetable biomarkers has emerged. These biomarkers are associated with the surface of malignant cells and serve as targets for directing cytotoxic T cells. The first biomarker target used for CAR T cell therapy was CD19, a B cell marker expressed highly on malignant B cells. With the success of CD19, the last decade has shown an explosion of new targetable biomarkers on a range of human malignancies. These surface targets have made it possible to provide directed, specific therapy that reduces healthy tissue destruction and preserves the patient's immune system during treatment. As of May 2018, there are over 100 clinical trials underway that target over 25 different surface biomarkers in almost every human tissue. This expansion has led to not only promising results in terms of patient outcome, but has also led to an exponential growth in the investigation of new biomarkers that could potentially be utilized in CAR T cell therapy for treating patients. In this review, we discuss the biomarkers currently under investigation and point out several promising biomarkers in the preclinical stage of development that may be useful as targets.
Collapse
Affiliation(s)
- Michelle H. Townsend
- Department of Microbiology and Molecular Biology, Brigham Young University, 3142 LSB, Provo, UT 84602 USA
| | - Gajendra Shrestha
- Department of Microbiology and Molecular Biology, Brigham Young University, 3142 LSB, Provo, UT 84602 USA
- Thunder Biotech, Highland, UT USA
| | - Richard A. Robison
- Department of Microbiology and Molecular Biology, Brigham Young University, 3142 LSB, Provo, UT 84602 USA
| | - Kim L. O’Neill
- Department of Microbiology and Molecular Biology, Brigham Young University, 3142 LSB, Provo, UT 84602 USA
| |
Collapse
|
38
|
Wang X, Qiu H, Tang R, Song H, Pan H, Feng Z, Chen L. miR‑30a inhibits epithelial‑mesenchymal transition and metastasis in triple‑negative breast cancer by targeting ROR1. Oncol Rep 2018; 39:2635-2643. [PMID: 29693179 PMCID: PMC5983935 DOI: 10.3892/or.2018.6379] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 03/30/2018] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer subtype that lacks effective targeted therapies. In the present study, we revealed that the expression of miR-30a was significantly decreased in TNBC, and TNBC patients with low expression of miR-30a were associated with high histological grade and more lymph node metastasis. Moreover, we found that miR-30a suppressed TNBC cell epithelial-mesenchymal transition (EMT), as demonstrated by the overexpression of miR-30a which increased the expression of epithelial marker E-cadherin but decreased the expression of mesenchymal markers N-cadherin and vimentin. Furthermore, we demonstrated that overexpression of miR-30a significantly suppressed TNBC cell invasion and migration, as well as inhibited tumor growth and metastasis in vivo. More importantly, RTK-like orphan receptor 1 (ROR1) was predicted as the direct target of miR-30a, which was subsequently confirmed by luciferase assays. Forced expression of miR-30a in TNBC cells decreased ROR1 expression, whereas the overexpression of ROR1 reversed the suppressive effects of miR-30a in TNBC cell migration and invasion. Collectively, this study indicated that miR-30a functions as a tumor-metastasis suppressor miRNA in TNBC by directly targeting ROR1 and that miR-30a may serve as a novel therapeutic target for TNBC.
Collapse
Affiliation(s)
- Xin Wang
- Department of Radiation Oncology, Nan Fang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Huisi Qiu
- Department of Radiation Oncology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong 511500, P.R. China
| | - Ruiming Tang
- Department of Radiation Oncology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong 511500, P.R. China
| | - Huisheng Song
- Department of Radiation Oncology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong 511500, P.R. China
| | - Huilin Pan
- Department of Radiation Oncology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong 511500, P.R. China
| | - Zhengfu Feng
- Department of Radiation Oncology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong 511500, P.R. China
| | - Longhua Chen
- Department of Radiation Oncology, Nan Fang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
39
|
Zhong A, Tian Y, Zhang H, Lai M. DNA hydroxymethylation of colorectal primary carcinoma and its association with survival. J Surg Oncol 2018. [PMID: 29529342 DOI: 10.1002/jso.24978] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
METHODS A total of 71 cases of colorectal carcinoma with hepatic metastasis were enrolled from the Department of Pathology of SIR RUN RUN SHAW Hospital. Paired primary tumors, hepatic metastases, and normal mucosa samples were collected from formalin-fixed paraffin-embedded tissues by manual macrodissection. And global levels of DNA methylation and hydroxymethylation in these tissues, measured by an ELISA-like microplate-based colorimetric methods. The immunohistochemical expression of 5-methylcytosine and 5-hydroxymethylcytosine were analyzed also. RESULTS The levels of DNA methylation in both primary and metastatic tumors were elevated when compared with normal mucosa, while DNA hydroxymethylation decreased slightly in those tissues. Similar results were observed in immunohistochemical staining. DNA methylation in hepatic metastases differed significantly in lymph node metastases (P = 0.037). And DNA hydroxymethylation in colorectal primary carcinoma was significantly different between tumor grade group (P = 0.018) and gender group (P = 0.048) respectively. And survival analyzes revealed that higher levels DNA hydroxymethylation were associated with better prognosis in colorectal primary carcinoma (P < 0.05). CONCLUSION DNA hydroxymethylation correlated with less aggressive tumor behavior in colorectal cancer and were identified as an independent prognostic factor in patients' overall survival, and downregulation of DNA hydroxymethylation may serve as a useful biomarker for colorectal cancer prognosis evaluation.
Collapse
Affiliation(s)
- Anjing Zhong
- Department of Pathology and Pathophysiology, Cancer Research Center, School of Medicine, Hangzhou, Zhejiang University, Zhejiang, P. R. China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, Zhejiang, P. R. China.,Pathology Department of the Affiliated Hospital of Jiangsu University, Jiangsu, P. R. China
| | - Yiping Tian
- Department of Pathology and Pathophysiology, Cancer Research Center, School of Medicine, Hangzhou, Zhejiang University, Zhejiang, P. R. China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, Zhejiang, P. R. China.,Pathology Department, Zhejiang Cancer Hospital, Hangzhou, P. R. China
| | - Honghe Zhang
- Department of Pathology and Pathophysiology, Cancer Research Center, School of Medicine, Hangzhou, Zhejiang University, Zhejiang, P. R. China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, Zhejiang, P. R. China
| | - Maode Lai
- Department of Pathology and Pathophysiology, Cancer Research Center, School of Medicine, Hangzhou, Zhejiang University, Zhejiang, P. R. China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, Zhejiang, P. R. China
| |
Collapse
|
40
|
Hemmati A, Hassannia H, Milani S, Hadavi R, Ghaemimanesh F, Rabbani H. Detecting Receptor Tyrosine Kinase ROR1 Using a Developed Anti-ROR1 Polyclonal Antibody. Monoclon Antib Immunodiagn Immunother 2018; 37:38-44. [PMID: 29474159 DOI: 10.1089/mab.2017.0059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Receptor tyrosine kinase ROR1 has been introduced as an interesting prognostic cancer marker in histopathology. The aim of this study was to produce a polyclonal antibody (PAb) against recombinant human ROR1 protein to be used as a tool for investigation of ROR1 expression in human cancer tissue blocks. The extracellular part of human ROR1 recombinant protein was expressed using pET-28b(+) plasmid in Escherichia coli Bl21(DE3) host. The recombinant ROR1, as a candidate immunogen, was purified and injected to a New Zealand rabbit. Followed by raising the titration of antibody, polyclonal anti-ROR1 antibody was purified through affinity chromatography column. After determining the purity of PAb anti-ROR1, its specific reactivity was assessed through various assessments. Flow cytometry analysis showed that PAb anti-ROR1 specifically recognizes ROR1 molecule in a number of positive and negative cell lines. Results obtained from detection of ROR1 in paraffin-embedded breast adenocarcinoma tissue blocks (n = 11) also demonstrated that PAb anti-ROR1 can effectively be used in immunohistochemistry. In conclusion, the developed anti-ROR1 PAb can be used as a tool for determining the prognostic value of ROR1 in histopathology of cancer tissues.
Collapse
Affiliation(s)
- Azam Hemmati
- Antibody-Antigen Engineering Department, Monoclonal Antibody Research Center, Avicenna Research Institute , ACECR, Tehran, Iran
| | - Hadi Hassannia
- Antibody-Antigen Engineering Department, Monoclonal Antibody Research Center, Avicenna Research Institute , ACECR, Tehran, Iran
| | - Saeideh Milani
- Antibody-Antigen Engineering Department, Monoclonal Antibody Research Center, Avicenna Research Institute , ACECR, Tehran, Iran
| | - Reza Hadavi
- Antibody-Antigen Engineering Department, Monoclonal Antibody Research Center, Avicenna Research Institute , ACECR, Tehran, Iran
| | - Fatemeh Ghaemimanesh
- Antibody-Antigen Engineering Department, Monoclonal Antibody Research Center, Avicenna Research Institute , ACECR, Tehran, Iran
| | - Hodjattallah Rabbani
- Antibody-Antigen Engineering Department, Monoclonal Antibody Research Center, Avicenna Research Institute , ACECR, Tehran, Iran
| |
Collapse
|
41
|
Milani S, Ghaemimanesh F, Salimi A, Hadavi R, Bayat AA, Alirezapour B, Rabbani H. Production and evaluation of a 67Ga-labeled anti-Ror1 monoclonal antibody in a mouse model of breast cancer. J Radioanal Nucl Chem 2018. [DOI: 10.1007/s10967-018-5755-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|