1
|
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; 67:2603-2616. [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] [MESH Headings] [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
|
2
|
Rui W, Quanquan G, Meimei M, Xiaohong S. Targeting ROR1 Inhibits Glucocorticoid-induced Gastric Cancer Metastasis. Steroids 2023; 195:109239. [PMID: 37068700 DOI: 10.1016/j.steroids.2023.109239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/23/2023] [Accepted: 04/10/2023] [Indexed: 04/19/2023]
Abstract
Glucocorticoids are commonly used in clinic but are also a double-edged sword. While treating tumors, they are reported to promote tumor growth and metastasis. To explore the role and elucidate the mechanism of dexamethasone in promoting tumor growth and metastasis, we detected the levels of cortisol and adrenocorticotropic hormone (ACTH) in peripheral blood of patients with gastric cancer, and immunohistochemical staining was used to detect the expression of GR and ROR1 in the surgically resected gastric cancer samples. The levels of cortisol and ACTH in peripheral blood of patients with stage III and IV gastric cancer were higher than those of patients with stage I/II gastric cancer. Dexamethasone up-regulated the ROR1 level on gastric cancer cell lines in a concentration-dependent manner. Gastric cancer specimen with high ROR1 had higher rates of relapse and metastasis than gastric adenocarcinomas expressing low levels of ROR1.Gastric cancer patients with high expression of ROR1 had a short survival time. ROR1 was expressed by gastric cancer cell lines, but not on normal gastric epithelial cell line. Suppressing ROR1 in gastric cancer cell lines impaired their invasion, migration, scratch healing and clone formation ability in vitro and slowed down the tumor growth of MKN-45 cells in immunodeficient mice in vivo. Collectively, our study indicated that dexamethasone up-regulated ROR1 levels on gastric cancer cells. ROR1 participated in and mediated the role of dexamethasone in promoting gastric tumor growth, and blocking ROR1 can prevent the tumor growth.
Collapse
Affiliation(s)
- Wang Rui
- Department of Oncology, Suqian Affiliated Hospital of Xuzhou Medical University, No. 138, Huanghe Road, Su Cheng District, Suqian City, China; Department of Oncology, Suqian Affiliated Hospital of Xuzhou Medical University, No. 138, Huanghe Road, Su Cheng District, Suqian City, China; Department of Oncology, Suqian Affiliated Hospital of Xuzhou Medical University, No. 138, Huanghe Road, Su Cheng District, Suqian City, China.
| | - Guo Quanquan
- Department of Oncology, Suqian Affiliated Hospital of Xuzhou Medical University, No. 138, Huanghe Road, Su Cheng District, Suqian City, China; Department of Oncology, Suqian Affiliated Hospital of Xuzhou Medical University, No. 138, Huanghe Road, Su Cheng District, Suqian City, China; Department of Oncology, Suqian Affiliated Hospital of Xuzhou Medical University, No. 138, Huanghe Road, Su Cheng District, Suqian City, China
| | - Ma Meimei
- Department of Pathology, Suqian Affiliated Hospital of Xuzhou Medical University, No. 138, Huanghe Road, Su Cheng District, Suqian City, China
| | - Shi Xiaohong
- Department of Pathology, Suqian Affiliated Hospital of Xuzhou Medical University, No. 138, Huanghe Road, Su Cheng District, Suqian City, China
| |
Collapse
|
3
|
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
|
4
|
Fan Y, Zhang F, Xiong L, Su M, Luo F, Li M, Li Q, Zhong T, Yuan M, Xu Y, Mu S, Yang H. Design, synthesis, and biological evaluation of 6-(imidazo[1,2-a] pyridin-6-yl) quinazolin-4(3H)-one derivatives as potent anticancer agents by dual targeting Aurora kinase and ROR1. Bioorg Chem 2023; 135:106484. [PMID: 36963371 DOI: 10.1016/j.bioorg.2023.106484] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/26/2023]
Abstract
ROR1 and Aurora kinase were overexpressed in various cancers and essential for cell proliferation, survive and metastasis. Pharmaceutical inhibition of ROR1 and Aurora kinase abrogated the activation of downstream signaling and induced cancer cell apoptosis. Hence, ROR1 and Aurora kinase considered as attractive therapeutic targets for the development of anticancer drugs. In the present work, three series of novel 6-(imidazo[1,2-a] pyridin-6-yl)-quinazolin-4(3H)-one derivatives were designed and synthesized via bioisosterism and scaffold-hopping strategies guided by FLF-13, an Aurora kinase inhibitor we discovered earlier. Most of compounds in series 2 and series 3 showed submicromolar to nanomolar inhibitory activity against multiple cancer cell lines. More importantly, compounds 12d and 12f in series 3 showed nanomolar inhibitory activity against all test cancer cells. The most promising compound 12d exhibited potent inhibitory activity against Aurora A and Aurora B with IC50 values of 84.41 nM and 14.09 nM, respectively. Accordingly, compounds 12d induced G2/M phase cell cycle arrest at 24 h and polyploidy at 48 h. It's worth noting that 12d also displayed inhibitory activity against ROR1 and induce cell apoptosis. Furthermore, 12d could significantly inhibit the tumor growth in SH-SY5Y xenograft model with tumor growth inhibitory rate (IR) up to 46.31 % at 10 mg/kg and 52.66 % at 20 mg/kg. Overall, our data suggested that 12d might serve as a promising candidate for the development of therapeutic agents for cancers with aberrant expression of ROR1 and Aurora kinases by simultaneously targeting ROR1 and Aurora kinase.
Collapse
Affiliation(s)
- Yanhua Fan
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China.
| | - Feng Zhang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Liang Xiong
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Mingzhi Su
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Fang Luo
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Mei Li
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Qing Li
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Ting Zhong
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Meitao Yuan
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Yongnan Xu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shuzhen Mu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China.
| | - Huarong Yang
- Chongqing Liangping District People's Hospital, Chongqing 405200, China.
| |
Collapse
|
5
|
Ghaderi A, Zhong W, Okhovat MA, Aschan J, Svensson A, Sander B, Schultz J, Olin T, Österborg A, Hojjat-Farsangi M, Mellstedt H. A ROR1 Small Molecule Inhibitor (KAN0441571C) Induced Significant Apoptosis of Mantle Cell Lymphoma (MCL) Cells. Pharmaceutics 2022; 14:pharmaceutics14102238. [PMID: 36297673 PMCID: PMC9607197 DOI: 10.3390/pharmaceutics14102238] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/07/2022] [Accepted: 10/17/2022] [Indexed: 11/18/2022] Open
Abstract
The receptor tyrosine kinase orphan receptor 1 (ROR1) is absent in most normal adult tissues but overexpressed in various malignancies and is of importance for tumor cell survival, proliferation, and metastasis. In this study, we evaluated the apoptotic effects of a novel small molecule inhibitor of ROR1 (KAN0441571C) as well as venetoclax (BCL-2 inhibitor), bendamustine, idelalisib (PI3Kδ inhibitor), everolimus (mTOR inhibitor), and ibrutinib (BTK inhibitor) alone or in combination in human MCL primary cells and cell lines. ROR1 expression was evaluated by flow cytometry and Western blot (WB). Cytotoxicity was analyzed by MTT and apoptosis by Annexin V/PI staining as well as signaling and apoptotic proteins (WB). ROR1 was expressed both in patient-derived MCL cells and human MCL cell lines. KAN0441571C alone induced significant time- and dose-dependent apoptosis of MCL cells. Apoptosis was accompanied by decreased expression of MCL-1 and BCL-2 and cleavage of PARP and caspase 3. ROR1 was dephosphorylated as well as ROR1-associated signaling pathway molecules, including the non-canonical WNT signaling pathway (PI3Kδ/AKT/mTOR). The combination of KAN0441571C and ibrutinib, venetoclax, idelalisib, everolimus, or bendamustine had a synergistic apoptotic effect and significantly prevented phosphorylation of ROR1-associated signaling molecules as compared to KAN0441571C alone. Our results suggest that targeting ROR1 by a small molecule inhibitor, KAN0441571C, should be further evaluated particularly in combination with other targeting drugs as a new therapeutic approach for MCL.
Collapse
Affiliation(s)
- Amineh Ghaderi
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna and Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Wen Zhong
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna and Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Mohammad Ali Okhovat
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna and Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Johanna Aschan
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna and Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Ann Svensson
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna and Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Birgitta Sander
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, 171 77 Stockholm, 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 and Karolinska Institutet, 171 64 Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital Solna, 171 77 Stockholm, Sweden
| | - Mohammad Hojjat-Farsangi
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna and Karolinska Institutet, 171 64 Stockholm, Sweden
- Correspondence: ; Tel.: +46-735-234-706
| | - Håkan Mellstedt
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna and Karolinska Institutet, 171 64 Stockholm, Sweden
| |
Collapse
|
6
|
Katoh M, Katoh M. WNT signaling and cancer stemness. Essays Biochem 2022; 66:319-331. [PMID: 35837811 PMCID: PMC9484141 DOI: 10.1042/ebc20220016] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 12/11/2022]
Abstract
Cancer stemness, defined as the self-renewal and tumor-initiation potential of cancer stem cells (CSCs), is a cancer biology property featuring activation of CSC signaling networks. Canonical WNT signaling through Frizzled and LRP5/6 receptors is transmitted to the β-catenin-TCF/LEF-dependent transcription machinery to up-regulate MYC, CCND1, LGR5, SNAI1, IFNG, CCL28, CD274 (PD-L1) and other target genes. Canonical WNT signaling causes expansion of rapidly cycling CSCs and modulates both immune surveillance and immune tolerance. In contrast, noncanonical WNT signaling through Frizzled or the ROR1/2 receptors is transmitted to phospholipase C, Rac1 and RhoA to control transcriptional outputs mediated by NFAT, AP-1 and YAP-TEAD, respectively. Noncanonical WNT signaling supports maintenance of slowly cycling, quiescent or dormant CSCs and promotes epithelial-mesenchymal transition via crosstalk with TGFβ (transforming growth factor-β) signaling cascades, while the TGFβ signaling network induces immune evasion. The WNT signaling network orchestrates the functions of cancer-associated fibroblasts, endothelial cells and immune cells in the tumor microenvironment and fine-tunes stemness in human cancers, such as breast, colorectal, gastric and lung cancers. Here, WNT-related cancer stemness features, including proliferation/dormancy plasticity, epithelial-mesenchymal plasticity and immune-landscape plasticity, will be discussed. Porcupine inhibitors, β-catenin protein-protein interaction inhibitors, β-catenin proteolysis targeting chimeras, ROR1 inhibitors and ROR1-targeted biologics are investigational drugs targeting WNT signaling cascades. Mechanisms of cancer plasticity regulated by the WNT signaling network are promising targets for therapeutic intervention; however, further understanding of context-dependent reprogramming trajectories might be necessary to optimize the clinical benefits of WNT-targeted monotherapy and applied combination therapy for patients with cancer.
Collapse
Affiliation(s)
| | - Masaru Katoh
- M & M Precision Medicine
- Department of Omics Network, National Cancer Center, Japan
- Department of Clinical Genomics, National Cancer Center, Japan
| |
Collapse
|