1
|
Neale DA, Morris JC, Verrills NM, Ammit AJ. Understanding the regulatory landscape of protein phosphatase 2A (PP2A): Pharmacological modulators and potential therapeutics. Pharmacol Ther 2025; 269:108834. [PMID: 40023321 DOI: 10.1016/j.pharmthera.2025.108834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/20/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Protein phosphatase 2A (PP2A) is a ubiquitously expressed serine/threonine phosphatase with a diverse and integral role in cellular signalling pathways. Consequently, its dysfunction is frequently observed in disease states such as cancer, inflammation and Alzheimer's disease. A growing understanding of both PP2A and its endogenous regulatory proteins has presented numerous targets for therapeutic intervention. This provides important context for the dynamic control and dysregulation of PP2A function in disease states. Understanding the intricate regulation of PP2A signalling in disease has resulted in the development of novel pharmacological agents aimed at restoring cellular homeostasis. Herein we review the structure and function of PP2A together with pharmacological modulators, both endogenous (proteins) and exogenous (small molecules and peptides), with relevance to targeting PP2A as a future pharmacotherapeutic strategy.
Collapse
Affiliation(s)
- David A Neale
- School of Chemistry, UNSW Sydney, NSW 2052, Australia
| | | | - Nicole M Verrills
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, NSW 2308, Australia; Precision Medicine Program, Hunter Medical Research Institute, New Lambton, NSW 2305, Australia
| | - Alaina J Ammit
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, Macquarie University, NSW, Australia; School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW, Australia.
| |
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; 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
|
3
|
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
|
4
|
Raso MG, Barrientos Toro E, Evans K, Rizvi Y, Lazcano R, Akcakanat A, Sini P, Trapani F, Madlener EJ, Waldmeier L, Lazar A, Meric-Bernstam F. Heterogeneous Profile of ROR1 Protein Expression across Tumor Types. Cancers (Basel) 2024; 16:1874. [PMID: 38791952 PMCID: PMC11119314 DOI: 10.3390/cancers16101874] [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: 04/12/2024] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
The Wnt receptor ROR1 has generated increased interest as a cancer therapeutic target. Research on several therapeutic approaches involving this receptor is ongoing; however, ROR1 tissue expression remains understudied. We performed an immunohistochemistry analysis of ROR1 protein expression in a large cohort of multiple tumor and histologic types. We analyzed 12 anonymized multi-tumor tissue microarrays (TMAs), including mesothelioma, esophageal and upper gastrointestinal carcinomas, and uterine endometrioid carcinoma, among other tumor types. Additionally, we studied 5 different sarcoma types of TMAs and 6 patient-derived xenografts (PDX) TMAs developed from 19 different anatomic sites and tumor histologic types. A total of 1142 patient cases from different histologic types and 140 PDXs placed in TMAs were evaluated. Pathologists assessed the percentage of tumor cells in each case that were positive for ROR1 and the intensity of staining. For determining the prevalence of staining for each tumor type, a case was considered positive if >1% of its tumor cells showed ROR1 staining. Our immunohistochemistry assays revealed a heterogeneous ROR1 expression profile. A high prevalence of ROR1 expression was found in mesothelioma (84.6%), liposarcoma (36.1%), gastrointestinal stromal tumors (33.3%), and uterine endometrioid carcinoma (28.9%). Other histologic types such as breast, lung, renal cell, hepatocellular, urothelial carcinoma, and colon carcinomas; glioblastoma; cholangiocarcinoma; and leiomyosarcoma showed less ROR1 overall expression, ranging between 0.9 and 13%. No ROR1 expression was seen in mesenchymal chondrosarcoma, rhabdomyosarcoma, or gastric adenocarcinoma cases. Overall, ROR1 expression was relatively infrequent and low in most tumor types investigated; however, ROR1 expression was infrequent but high in selected tumor types, such as gastroesophageal GIST, suggesting that ROR1 prescreening may be preferable for those indications. Further, mesothelioma exhibited frequent and high levels of ROR1 expression, which represents a previously unrecognized therapeutic opportunity. These findings can contribute to the development of ROR1-targeted therapies.
Collapse
Affiliation(s)
- Maria Gabriela Raso
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (E.B.T.); (R.L.)
| | - Elizve Barrientos Toro
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (E.B.T.); (R.L.)
| | - Kurt Evans
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (K.E.); (Y.R.); (A.A.); (F.M.-B.)
| | - Yasmeen Rizvi
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (K.E.); (Y.R.); (A.A.); (F.M.-B.)
| | - Rossana Lazcano
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (E.B.T.); (R.L.)
| | - Argun Akcakanat
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (K.E.); (Y.R.); (A.A.); (F.M.-B.)
| | - Patrizia Sini
- Boehringer Ingelheim RCV, 1121 Vienna, Austria (F.T.)
| | | | | | | | - Alexander Lazar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (K.E.); (Y.R.); (A.A.); (F.M.-B.)
| |
Collapse
|
5
|
Han M, Liu X, Hailati S, Nurahmat N, Dilimulati D, Baishan A, Aikebaier A, Zhou W. Evaluation of the Efficacy of OSU-2S in the Treatment of Non-Small-Cell Lung Cancer and Screening of Potential Targets of Action. Pharmaceuticals (Basel) 2024; 17:582. [PMID: 38794152 PMCID: PMC11124116 DOI: 10.3390/ph17050582] [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: 02/25/2024] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 05/26/2024] Open
Abstract
(1) Background: OSU-2S is a derivative of FTY720 and exhibits significant inhibitory effects on various cancer cells. There is currently no research on the mechanism of the impact of OSU-2S on NSCLC development. We analysed and validated the hub genes and pharmacodynamic effects of OSU-2S to treat NSCLC. (2) Methods: The hub genes of OSU-2S for the treatment of NSCLC were screened in PharmMapper, genecard, and KM Plotter database by survival and expression analysis. The effect of OSU-2S on hub gene expression was verified by Western blot analysis. The ex vivo and in vivo efficacy of OSU-2S on tumour growth was verified using A549 cells and a xenografted animal model. (3) Results: A total of 7 marker genes for OSU-2S treatment of NSCLC were obtained. AURKA and S1PR1 were screened as hub genes. Significant differences in the expression of AURKA and S1PR1 between normal and lung adenocarcinoma (LUAD) tissues were found in the GEPIA2 database; Western blot showed that OSU-2S could affect p-AURKA and S1PR1 protein expression. OSU-2S significantly inhibited tumour growth in A549 cells and xenografted animal models. (4) Conclusions: Our study confirms the inhibitory effect of OSU-2S on NSCLC, screens and demonstrates its potential targets AURKA(p-AURKA) and S1PR1, and provides a research basis for treating NSCLC with OSU-2S.
Collapse
Affiliation(s)
- Mengyuan Han
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (M.H.); (X.L.); (S.H.); (N.N.); (D.D.); (A.B.); (A.A.)
- Xinjiang Key Laboratory of Active Components and Drug Release Technology of Natural Medicines, Urumqi 830017, China
| | - Xiangran Liu
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (M.H.); (X.L.); (S.H.); (N.N.); (D.D.); (A.B.); (A.A.)
| | - Sendaer Hailati
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (M.H.); (X.L.); (S.H.); (N.N.); (D.D.); (A.B.); (A.A.)
- Xinjiang Key Laboratory of Active Components and Drug Release Technology of Natural Medicines, Urumqi 830017, China
| | - Nurbiya Nurahmat
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (M.H.); (X.L.); (S.H.); (N.N.); (D.D.); (A.B.); (A.A.)
- Xinjiang Key Laboratory of Active Components and Drug Release Technology of Natural Medicines, Urumqi 830017, China
| | - Dilihuma Dilimulati
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (M.H.); (X.L.); (S.H.); (N.N.); (D.D.); (A.B.); (A.A.)
- Xinjiang Key Laboratory of Active Components and Drug Release Technology of Natural Medicines, Urumqi 830017, China
| | - Alhar Baishan
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (M.H.); (X.L.); (S.H.); (N.N.); (D.D.); (A.B.); (A.A.)
- Xinjiang Key Laboratory of Active Components and Drug Release Technology of Natural Medicines, Urumqi 830017, China
| | - Alifeiye Aikebaier
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (M.H.); (X.L.); (S.H.); (N.N.); (D.D.); (A.B.); (A.A.)
- Xinjiang Key Laboratory of Active Components and Drug Release Technology of Natural Medicines, Urumqi 830017, China
| | - Wenting Zhou
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (M.H.); (X.L.); (S.H.); (N.N.); (D.D.); (A.B.); (A.A.)
- Xinjiang Key Laboratory of Active Components and Drug Release Technology of Natural Medicines, Urumqi 830017, China
| |
Collapse
|
6
|
Mouawad N, Ruggeri E, Capasso G, Martinello L, Visentin A, Frezzato F, Trentin L. How receptor tyrosine kinase-like orphan receptor 1 meets its partners in chronic lymphocytic leukemia. Hematol Oncol 2024; 42:e3250. [PMID: 38949887 DOI: 10.1002/hon.3250] [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: 10/12/2023] [Revised: 12/29/2023] [Accepted: 01/02/2024] [Indexed: 07/03/2024]
Abstract
Chronic lymphocytic leukemia (CLL) is the most common leukemia in western societies, recognized by clinical and molecular heterogeneity. Despite the success of targeted therapies, acquired resistance remains a challenge for relapsed and refractory CLL, as a consequence of mutations in the target or the upregulation of other survival pathways leading to the progression of the disease. Research on proteins that can trigger such pathways may define novel therapies for a successful outcome in CLL such as the receptor tyrosine kinase-like orphan receptor 1 (ROR1). ROR1 is a signaling receptor for Wnt5a, with an important role during embryogenesis. The aberrant expression on CLL cells and several types of tumors, is involved in cell proliferation, survival, migration as well as drug resistance. Antibody-based immunotherapies and small-molecule compounds emerged to target ROR1 in preclinical and clinical studies. Efforts have been made to identify new prognostic markers having predictive value to refine and increase the detection and management of CLL. ROR1 can be considered as an attractive target for CLL diagnosis, prognosis, and treatment. It can be clinically effective alone and/or in combination with current approved agents. In this review, we summarize the scientific achievements in targeting ROR1 for CLL diagnosis, prognosis, and treatment.
Collapse
MESH Headings
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Humans
- Receptor Tyrosine Kinase-like Orphan Receptors/metabolism
- Prognosis
- Molecular Targeted Therapy
- Animals
- Biomarkers, Tumor/metabolism
Collapse
Affiliation(s)
- Nayla Mouawad
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Edoardo Ruggeri
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Guido Capasso
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Leonardo Martinello
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Andrea Visentin
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Federica Frezzato
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Livio Trentin
- Hematology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| |
Collapse
|
7
|
Johnson H, Narayan S, Sharma AK. Altering phosphorylation in cancer through PP2A modifiers. Cancer Cell Int 2024; 24:11. [PMID: 38184584 PMCID: PMC10770906 DOI: 10.1186/s12935-023-03193-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 12/25/2023] [Indexed: 01/08/2024] Open
Abstract
Protein phosphatase 2A (PP2A) is a serine/threonine phosphatase integral to the regulation of many cellular processes. Due to the deregulation of PP2A in cancer, many of these processes are turned toward promoting tumor progression. Considerable research has been undertaken to discover molecules capable of modulating PP2A activity in cancer. Because PP2A is capable of immense substrate specificity across many cellular processes, the therapeutic targeting of PP2A in cancer can be completed through either enzyme inhibitors or activators. PP2A modulators likewise tend to be effective in drug-resistant cancers and work synergistically with other known cancer therapeutics. In this review, we will discuss the patterns of PP2A deregulation in cancer, and its known downstream signaling pathways important for cancer regulation, along with many activators and inhibitors of PP2A known to inhibit cancer progression.
Collapse
Affiliation(s)
- Hannah Johnson
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Satya Narayan
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL, 32610, USA
| | - Arun K Sharma
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
8
|
Yikilmaz AŞ, Bakanay ŞM, Avcı DN, Akinci S, Falay M, Özet G, Dilek İ. Prognostic Value of the Expression of Receptor Tyrosine Kinase-Like Orphan Receptor 1 (ROR-1) in Chronic Lymphocytic Leukemia. Int J Hematol Oncol Stem Cell Res 2023; 17:39-47. [PMID: 37638287 PMCID: PMC10448922 DOI: 10.18502/ijhoscr.v17i1.11712] [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: 06/08/2021] [Accepted: 09/06/2021] [Indexed: 08/29/2023] Open
Abstract
Background: The transmembrane receptor tyrosine kinase-like orphan receptor 1 (ROR1) has acted on the causation and sustentation of mature B-cell lymphomagenesis for chronic lymphocytic leukemia (CLL) cells. The study attempted to show whether there is a relationship between the level of ROR1 surface expression in CLL cells and disease findings. Materials and Methods: The level of ROR1 cell surface expression was determined in accordance with the flow cytometric analysis of CLL patients at the first diagnosis time. Two groups were formed according to the high and low ROR1 levels. The cut-off point for the ROR1 level was calculated for advanced-stage disease using receiver operating characteristic (ROC) curves. A two-sided p-value <0,05 was considered statistically significant. Results: 108 CLL cases with a median age of 60 were enrolled. The median percentage of ROR1 cell surface marker positivity in the CD5/CD19 positive leukemic cell was 62%. The CLL cases with high ROR1 levels have thrombocytopenia (p=0.042), anemia (p=0.028), and high beta-2 microglobulin value ≥3 mg/dL (p=0.002) and the need for first-line treatment (p=0.043). Conclusion: The poor prognostic parameters such as splenomegaly, anemia, higher beta-2 microglobulin levels, intermediate/advanced RAİ stage disease, and need for first-line treatment had associated high-level ROR 1 expression of our CLL patients. It needs to be investigated for its effect on predicting disease burden and aggressiveness with more comprehensive studies on ROR1 expression levels in CLL cases.
Collapse
Affiliation(s)
- Aysun Şentürk Yikilmaz
- Department of Hematology, Bilkent City Hospital, Yıldırım Beyazıt University, Ankara, Turkey
| | - Şule Mine Bakanay
- Department of Hematology, Bilkent City Hospital, Yıldırım Beyazıt University, Ankara, Turkey
| | - Duygu Nurdan Avcı
- Department of Hematology, Ankara Numune Training and Research Hospital, Ankara, Turkey
| | - Sema Akinci
- Department of Hematology, Atatürk Training and Research Hospital, Ankara, Turkey
| | - Mesude Falay
- Department of Hematology, Ankara Numune Training and Research Hospital, Ankara, Turkey
| | - Gülsüm Özet
- Department of Hematology, Ankara Numune Training and Research Hospital, Ankara, Turkey
| | - İmdat Dilek
- Department of Hematology, Bilkent City Hospital, Yıldırım Beyazıt University, Ankara, Turkey
| |
Collapse
|
9
|
Abstract
Since its initial identification in 1992 as a possible class 1 cell-surface receptor without a known parent ligand, receptor tyrosine kinase-like orphan receptor 1 (ROR1) has stimulated research, which has made apparent its significance in embryonic development and cancer. Chronic lymphocytic leukemia (CLL) was the first malignancy found to have distinctive expression of ROR1, which can help distinguish leukemia cells from most noncancer cells. Aside from its potential utility as a diagnostic marker or target for therapy, ROR1 also factors in the pathophysiology of CLL. This review is a report of the studies that have elucidated the expression, biology, and evolving strategies for targeting ROR1 that hold promise for improving the therapy of patients with CLL or other ROR1-expressing malignancies.
Collapse
Affiliation(s)
- Thomas J. Kipps
- Center for Novel Therapeutics, Moores Cancer Center, Department of Medicine, University of California, San Diego, La Jolla, CA
| |
Collapse
|
10
|
Developing ROR1 Targeting CAR-T Cells against Solid Tumors in Preclinical Studies. Cancers (Basel) 2022; 14:cancers14153618. [PMID: 35892876 PMCID: PMC9331269 DOI: 10.3390/cancers14153618] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022] Open
Abstract
Chimeric antigen receptor (CAR)-modified T-cells (CAR-T) have demonstrated promising clinical benefits against B-cell malignancies. Yet, its application for solid tumors is still facing challenges. Unlike haematological cancers, solid tumors often lack good targets, which are ideally expressed on the tumor cells, but not by the normal healthy cells. Fortunately, receptor tyrosine kinase-like orphan receptor 1 (ROR1) is among a few good cancer targets that is aberrantly expressed on various tumors but has a low expression on normal tissue, suggesting it as a good candidate for CAR-T therapy. Here, we constructed two ROR1 CARs with the same antigen recognition domain that was derived from Zilovertamab but differing in hinge regions. Both CARs target ROR1+ cancer cells specifically, but CAR with a shorter IgG4 hinge exhibits a higher surface expression and better in vitro functionality. We further tested the ROR1 CAR-T in three human solid tumor xenografted mouse models. Our ROR1 CAR-T cells controlled the solid tumor growth without causing any severe toxicity. Our results demonstrated that ROR1 CAR-T derived from Zilovertamab is efficacious and safe to suppress ROR1+ solid tumors in vitro and in vivo, providing a promising therapeutic option for future clinical application.
Collapse
|
11
|
Collard JP, McKenna MK, Noothi SK, Alhakeem SS, Rivas JR, Rangnekar VM, Muthusamy N, Bondada S. Role of the splenic microenvironment in chronic lymphocytic leukemia development in Eµ-TCL1 transgenic mice. Leuk Lymphoma 2022; 63:1810-1822. [DOI: 10.1080/10428194.2022.2045596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- James P. Collard
- Department of Microbiology, Immunology and Molecular Genetics and Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Mary K. McKenna
- Department of Microbiology, Immunology and Molecular Genetics and Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Sunil K. Noothi
- Department of Microbiology, Immunology and Molecular Genetics and Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Sara S. Alhakeem
- Department of Microbiology, Immunology and Molecular Genetics and Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Jacqueline R. Rivas
- Department of Microbiology, Immunology and Molecular Genetics and Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Vivek M. Rangnekar
- Department of Radiation Medicine and Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Natarajan Muthusamy
- Division of Hematology, James Cancer Center, Ohio State University, Columbus, OH, USA
| | - Subbarao Bondada
- Department of Microbiology, Immunology and Molecular Genetics and Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
12
|
Goswami S, Chiang CL, Zapolnik K, Nunes J, Ventura A, Mo X, Xie Z, Lee LJ, Baskar S, Rader C, Byrd JC, Phelps M, Bhatnagar B, Muthusamy N. ROR1 targeted immunoliposomal delivery of OSU-2S shows selective cytotoxicity in t(1;19)(q23;p13) translocated B-cell acute lymphoblastic leukemia. Leuk Res 2022; 118:106872. [PMID: 35640397 PMCID: PMC10029232 DOI: 10.1016/j.leukres.2022.106872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 05/02/2022] [Accepted: 05/19/2022] [Indexed: 10/18/2022]
Affiliation(s)
- Swagata Goswami
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Chi-Ling Chiang
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Kevan Zapolnik
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Jessica Nunes
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Ann Ventura
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Xiaokui Mo
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Zhiliang Xie
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - L James Lee
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Sivasubramanian Baskar
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - John C Byrd
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Mitch Phelps
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Bhavana Bhatnagar
- Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Natarajan Muthusamy
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
13
|
Goswami S, Mani R, Nunes J, Chiang CL, Zapolnik K, Hu E, Frissora F, Mo X, Walker LA, Yan P, Bundschuh R, Beaver L, Devine R, Tsai YT, Ventura A, Xie Z, Chen M, Lapalombella R, Walker A, Mims A, Larkin K, Grieselhuber N, Bennett C, Phelps M, Hertlein E, Behbehani G, Vasu S, Byrd JC, Muthusamy N. PP2A is a therapeutically targetable driver of cell fate decisions via a c-Myc/p21 axis in human and murine acute myeloid leukemia. Blood 2022; 139:1340-1358. [PMID: 34788382 PMCID: PMC8900275 DOI: 10.1182/blood.2020010344] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 10/30/2021] [Indexed: 11/20/2022] Open
Abstract
Dysregulated cellular differentiation is a hallmark of acute leukemogenesis. Phosphatases are widely suppressed in cancers but have not been traditionally associated with differentiation. In this study, we found that the silencing of protein phosphatase 2A (PP2A) directly blocks differentiation in acute myeloid leukemia (AML). Gene expression and mass cytometric profiling revealed that PP2A activation modulates cell cycle and transcriptional regulators that program terminal myeloid differentiation. Using a novel pharmacological agent, OSU-2S, in parallel with genetic approaches, we discovered that PP2A enforced c-Myc and p21 dependent terminal differentiation, proliferation arrest, and apoptosis in AML. Finally, we demonstrated that PP2A activation decreased leukemia-initiating stem cells, increased leukemic blast maturation, and improved overall survival in murine Tet2-/-Flt3ITD/WT and human cell-line derived xenograft AML models in vivo. Our findings identify the PP2A/c-Myc/p21 axis as a critical regulator of the differentiation/proliferation switch in AML that can be therapeutically targeted in malignancies with dysregulated maturation fate.
Collapse
Affiliation(s)
- Swagata Goswami
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH
| | | | - Jessica Nunes
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH
| | - Chi-Ling Chiang
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Kevan Zapolnik
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Eileen Hu
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Frank Frissora
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Xiaokui Mo
- Center for Biostatistics, The Ohio State University, Columbus, OH
| | - Logan A Walker
- Biophysics Graduate Program, University of Michigan, Ann Arbor, MI
| | - Pearlly Yan
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Ralf Bundschuh
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH
- Department of Physics, The Ohio State University, Columbus, OH; and
| | - Larry Beaver
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Raymond Devine
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Yo-Ting Tsai
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Ann Ventura
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Zhiliang Xie
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Min Chen
- College of Pharmacy, The Ohio State University, Columbus, OH
| | - Rosa Lapalombella
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Alison Walker
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Alice Mims
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Karilyn Larkin
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Nicole Grieselhuber
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Chad Bennett
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Mitch Phelps
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- College of Pharmacy, The Ohio State University, Columbus, OH
| | - Erin Hertlein
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Gregory Behbehani
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Sumithira Vasu
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - John C Byrd
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
- College of Pharmacy, The Ohio State University, Columbus, OH
| | - Natarajan Muthusamy
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
| |
Collapse
|
14
|
Ci T, Zhang W, Qiao Y, Li H, Zang J, Li H, Feng N, Gu Z. Delivery strategies in treatments of leukemia. Chem Soc Rev 2022; 51:2121-2144. [PMID: 35188506 DOI: 10.1039/d1cs00755f] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Leukemia is a hematological malignancy associated with the uncontrolled proliferation of mutant progenitors, suppressing the production of normal blood cells. Current treatments, including chemotherapy, radiotherapy, and immunotherapy, still lead to unsatisfactory results with a 5 year survival rate of only 30-50%. The poor prognosis is related to both disease relapse and treatment-associated toxicity. Delivery strategies can improve the in vivo pharmacokinetics of drugs, navigating the therapeutics to target cells or the tumor microenvironment and reversing drug resistance, which maximizes tumor elimination and alleviates systematic adverse effects. This review discusses available FDA-approved anti-leukemia drugs and therapies with a focus on the advances in the development of anti-leukemia drug delivery systems. Additionally, challenges in clinical translation of the delivery strategies and future research opportunities in leukemia treatment are also included.
Collapse
Affiliation(s)
- Tianyuan Ci
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Wentao Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Yingyu Qiao
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, China
| | - Huangjuan Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, China
| | - Jing Zang
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Hongjun Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Nianping Feng
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Zhen Gu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China. .,Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China.,MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
15
|
The ROR1 antibody-drug conjugate huXBR1-402-G5-PNU effectively targets ROR1+ leukemia. Blood Adv 2021; 5:3152-3162. [PMID: 34424320 DOI: 10.1182/bloodadvances.2020003276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 04/18/2021] [Indexed: 11/20/2022] Open
Abstract
Antibody-drug conjugates directed against tumor-specific targets have allowed targeted delivery of highly potent chemotherapy to malignant cells while sparing normal cells. Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is an oncofetal protein with limited expression on normal adult tissues and is overexpressed on the surface of malignant cells in mantle cell lymphoma, acute lymphocytic leukemia with t(1;19)(q23;p13) translocation, and chronic lymphocytic leukemia. This differential expression makes ROR1 an attractive target for antibody-drug conjugate therapy, especially in malignancies such as mantle cell lymphoma and acute lymphocytic leukemia, in which systemic chemotherapy remains the gold standard. Several preclinical and phase 1 clinical studies have established the safety and effectiveness of anti-ROR1 monoclonal antibody-based therapies. Herein we describe a humanized, first-in-class anti-ROR1 antibody-drug conjugate, huXBR1-402-G5-PNU, which links a novel anti-ROR1 antibody (huXBR1-402) to a highly potent anthracycline derivative (PNU). We found that huXBR1-402-G5-PNU is cytotoxic to proliferating ROR1+ malignant cells in vitro and suppressed leukemia proliferation and extended survival in multiple models of mice engrafted with human ROR1+ leukemia. Lastly, we show that the B-cell lymphoma 2 (BCL2)-dependent cytotoxicity of huXBR1-402-G5-PNU can be leveraged by combined treatment strategies with the BCL2 inhibitor venetoclax. Together, our data present compelling preclinical evidence for the efficacy of huXBR1-402-G5-PNU in treating ROR1+ hematologic malignancies.
Collapse
|
16
|
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: 45] [Impact Index Per Article: 11.3] [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
|
17
|
Xie Z, Chen M, Goswami S, Mani R, Wang D, Kulp SK, Coss CC, Schaaf LJ, Cui F, Byrd JC, Jennings RN, Schober KK, Freed C, Lewis S, Malbrue R, Muthusamy N, Bennett C, Kisseberth WC, Phelps MA. Pharmacokinetics and Tolerability of the Novel Non-immunosuppressive Fingolimod Derivative, OSU-2S, in Dogs and Comparisons with Data in Mice and Rats. AAPS JOURNAL 2020; 22:92. [PMID: 32676788 DOI: 10.1208/s12248-020-00474-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/24/2020] [Indexed: 12/29/2022]
Abstract
In this study, we characterized the pharmacokinetics of OSU-2S, a fingolimod-derived, non-immunosuppressive phosphatase activator, in mice, rats, and dogs, as well as tolerability and food effects in dogs. Across all species tested, plasma protein binding for OSU-2S was > 99.5%, and metabolic stability and hepatic intrinsic clearance were in the moderate range. OSU-2S did not significantly modulate CYP enzyme activity up until 50 μM, and Caco-2 data suggested low permeability with active efflux at 2 μM. Apparent oral bioavailability in mice was 16% and 69% at 10 and 50 mg/kg, respectively. In rats, bioavailability was 24%, 35%, and 28% at 10, 30, and 100 mg/kg, respectively, while brain/plasma ratio was 36 at 6-h post-dose at 30 mg/kg. In dogs, OSU-2S was well tolerated with oral capsule bioavailability of 27.5%. Plasma OSU-2S exposures increased proportionally over a 2.5-20 mg/kg dose range. After 4 weeks of 3 times weekly, oral administration (20 mg/kg), plasma AUClast (26.1 μM*h), and Cmax (0.899 μM) were nearly 2-fold greater than those after 1 week of dosing, and no food effects were observed. The elimination half-life (29.7 h), clearance (22.9 mL/min/kg), and plasma concentrations of repeated oral doses support a 3-times weekly dosing schedule in dogs. No significant CBC, serum biochemical, or histopathological changes were observed. OSU-2S has favorable oral PK properties similar to fingolimod in rodents and dogs and is well tolerated in healthy animals. This work supports establishing trials of OSU-2S efficacy in dogs with spontaneous tumors to guide its clinical development as a cancer therapeutic for human patients.
Collapse
Affiliation(s)
- Zhiliang Xie
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 506 Riffe Building, 496 W. 12th Ave., Columbus, Ohio, 43210, USA
| | - Min Chen
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 506 Riffe Building, 496 W. 12th Ave., Columbus, Ohio, 43210, USA
| | - Swagata Goswami
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Rajes Mani
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Dasheng Wang
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Samuel K Kulp
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 506 Riffe Building, 496 W. 12th Ave., Columbus, Ohio, 43210, USA
| | - Chris C Coss
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 506 Riffe Building, 496 W. 12th Ave., Columbus, Ohio, 43210, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Larry J Schaaf
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | | | - John C Byrd
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 506 Riffe Building, 496 W. 12th Ave., Columbus, Ohio, 43210, USA.,Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Ryan N Jennings
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Karsten K Schober
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, 448 VMAB, 1900 Coffey Rd., Columbus, Ohio, 43210, USA
| | - Carrie Freed
- University Laboratory Animal Resources, The Ohio State University, Columbus, Ohio, USA
| | - Stephanie Lewis
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Raphael Malbrue
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Natarajan Muthusamy
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Chad Bennett
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - William C Kisseberth
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio, USA. .,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, 448 VMAB, 1900 Coffey Rd., Columbus, Ohio, 43210, USA.
| | - Mitch A Phelps
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 506 Riffe Building, 496 W. 12th Ave., Columbus, Ohio, 43210, USA. .,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
18
|
Javadpour P, Dargahi L, Ahmadiani A, Ghasemi R. To be or not to be: PP2A as a dual player in CNS functions, its role in neurodegeneration, and its interaction with brain insulin signaling. Cell Mol Life Sci 2019; 76:2277-2297. [PMID: 30874837 PMCID: PMC11105459 DOI: 10.1007/s00018-019-03063-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 02/16/2019] [Accepted: 03/07/2019] [Indexed: 12/26/2022]
Abstract
Accumulating evidence has reached the consensus that the balance of phosphorylation state of signaling molecules is a pivotal point in the regulation of cell signaling. Therefore, characterizing elements (kinases-phosphatases) in the phosphorylation balance are at great importance. However, the role of phosphatase enzymes is less investigated than kinase enzymes. PP2A is a member of serine/threonine protein phosphatase that its imbalance has been reported in neurodegenerative diseases. Therefore, we reviewed the superfamily of phosphatases and more specifically PP2A, its regulation, and physiological functions participate in CNS. Thereafter, we discussed the latest findings about PP2A dysregulation in Alzheimer and Parkinson diseases and possible interplay between this phosphatase and insulin signaling pathways. Finally, activating/inhibitory modulators for PP2A activity as well as experimental methods for PP2A study have been reviewed.
Collapse
Affiliation(s)
- Pegah Javadpour
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Ghasemi
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
ROR1-targeted delivery of miR-29b induces cell cycle arrest and therapeutic benefit in vivo in a CLL mouse model. Blood 2019; 134:432-444. [PMID: 31151986 DOI: 10.1182/blood.2018882290] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 04/29/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) occurs in 2 major forms: aggressive and indolent. Low miR-29b expression in aggressive CLL is associated with poor prognosis. Indiscriminate miR-29b overexpression in the B-lineage of mice causes aberrance, thus warranting the need for selective introduction of miR-29b into B-CLL cells for therapeutic benefit. The oncofetal antigen receptor tyrosine kinase orphan receptor 1 (ROR1) is expressed on malignant B-CLL cells, but not normal B cells, encouraging us with ROR1-targeted delivery for therapeutic miRs. Here, we describe targeted delivery of miR-29b to ROR1+ CLL cells leading to downregulation of DNMT1 and DNMT3A, modulation of global DNA methylation, decreased SP1, and increased p21 expression in cell lines and primary CLL cells in vitro. Furthermore, using an Eμ-TCL1 mouse model expressing human ROR1, we report the therapeutic benefit of enhanced survival via cellular reprograming by downregulation of DNMT1 and DNMT3A in vivo. Gene expression profiling of engrafted murine leukemia identified reprogramming of cell cycle regulators with decreased SP1 and increased p21 expression after targeted miR-29b treatment. This finding was confirmed by protein modulation, leading to cell cycle arrest and survival benefit in vivo. Importantly, SP1 knockdown results in p21-dependent compensation of the miR-29b effect on cell cycle arrest. These studies form a basis for leukemic cell-targeted delivery of miR-29b as a promising therapeutic approach for CLL and other ROR1+ B-cell malignancies.
Collapse
|
20
|
Li X, Wu N, Li B. A high mutation rate of immunoglobulin heavy chain variable region gene associates with a poor survival and chemotherapy response of mantle cell lymphoma patients. Medicine (Baltimore) 2019; 98:e15811. [PMID: 31145313 PMCID: PMC6708879 DOI: 10.1097/md.0000000000015811] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Immunoglobulin heavy chain variable region (IGHV) gene mutation status is a biomarker for the prognosis of chronic lymphocytic leukemia, whether it is associated with the diagnosis, staging, and prognosis of patients with mantle cell lymphoma (MCL) remains to be determined.The IGHV gene mutations of 52 MCL patients were determined by DNA sequencing and compared with published IGHV germline sequences.DNA sequence alignment of IGHV variable regions with published IGHV germline sequences showed that the coincidence rate was 94% to 100%. Ten cases (21%) were significantly mutated with the rate of 96.9% to 94.0%. The overall survival time of patients was negatively correlated with the degree of IGHV gene mutation. Further survival analysis with log-rank test demonstrated that the patients with significant IGHV gene mutations showed a trend towards poor survival.The mutation rate of the IGHV variant region may be determined to assess the prognosis and overall survival time of MCL patients.
Collapse
Affiliation(s)
- Xianqian Li
- Clinical Laboratory, Shanghai Yangpu District Psychiatric Hospital
| | | | - Bin Li
- Department of Pathology, Shanghai Xuhui Central Hospital, Shanghai, China
| |
Collapse
|
21
|
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: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [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
|
22
|
Greene JT, Mani R, Ramaswamy R, Frissora F, Yano M, Zapolnik K, Harrington B, Wasmuth R, Tran M, Mo X, McKenna M, Rangnekar VM, Byrd JC, Bondada S, Muthusamy N. Par-4 overexpression impedes leukemogenesis in the Eµ-TCL1 leukemia model through downregulation of NF-κB signaling. Blood Adv 2019; 3:1255-1266. [PMID: 30987970 PMCID: PMC6482354 DOI: 10.1182/bloodadvances.2018025973] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 02/23/2019] [Indexed: 01/25/2023] Open
Abstract
Prostate apoptosis response 4 (Par-4) is a tumor suppressor that prevents proliferation and induces cell death in several solid tumors. However, its role in B-cell malignancies has not been elucidated. To describe the role of Par-4 in chronic lymphocytic leukemia (CLL) pathogenesis, we developed a B-cell-specific human Par-4-overexpressing mouse model of CLL using the TCL1 leukemia model. While Par-4 transgenic mice did not display any obvious defects in B-cell development or function, disease burden as evidenced by abundance of CD19+CD5+ B cells in the peripheral blood was significantly reduced in Par-4 × TCL1 mice compared with TCL1 littermates. This conferred a survival advantage on the Par-4-overexpressing mice. In addition, a B-cell-specific knockout model displayed the opposite effect, where lack of Par-4 expression resulted in accelerated disease progression and abbreviated survival in the TCL1 model. Histological and flow cytometry-based analysis of spleen and bone marrow upon euthanasia revealed comparable levels of malignant B-cell infiltration in Par-4 × TCL1 and TCL1 individuals, indicating delayed but pathologically normal disease progression in Par-4 × TCL1 mice. In vivo analysis of splenic B-cell proliferation by 5-ethynyl-2-deoxyuridine incorporation indicated >50% decreased expansion of CD19+CD5+ cells in Par-4 × TCL1 mice compared with TCL1 littermates. Moreover, reduced nuclear p65 levels were observed in Par-4 × TCL1 splenic B cells compared with TCL1, suggesting suppressed NF-κB signaling. These findings have identified an in vivo antileukemic role for Par-4 through an NF-κB-dependent mechanism in TCL1-mediated CLL-like disease progression.
Collapse
MESH Headings
- Animals
- Apoptosis Regulatory Proteins/biosynthesis
- Carcinogenesis/genetics
- Carcinogenesis/metabolism
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Mice
- Mice, Transgenic
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Signal Transduction
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
Collapse
Affiliation(s)
- J T Greene
- The James Comprehensive Cancer Center and
| | | | | | | | - Max Yano
- The James Comprehensive Cancer Center and
| | | | | | | | - Minh Tran
- The James Comprehensive Cancer Center and
| | - Xiaokui Mo
- Center for Biostatistics, The Ohio State University, Columbus, OH; and
| | - Mary McKenna
- Markey Cancer Center, University of Kentucky, Lexington, KY
| | | | | | | | | |
Collapse
|
23
|
Karvonen H, Perttilä R, Niininen W, Barker H, Ungureanu D. Targeting Wnt signaling pseudokinases in hematological cancers. Eur J Haematol 2018; 101:457-465. [DOI: 10.1111/ejh.13137] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/05/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Hanna Karvonen
- BioMediTech Institute; University of Tampere; Tampere Finland
- Faculty of Medicine and Life Sciences; University of Tampere; Tampere Finland
| | - Robert Perttilä
- BioMediTech Institute; University of Tampere; Tampere Finland
- Faculty of Medicine and Life Sciences; University of Tampere; Tampere Finland
| | - Wilhelmiina Niininen
- BioMediTech Institute; University of Tampere; Tampere Finland
- Faculty of Medicine and Life Sciences; University of Tampere; Tampere Finland
| | - Harlan Barker
- Faculty of Medicine and Life Sciences; University of Tampere; Tampere Finland
| | - Daniela Ungureanu
- BioMediTech Institute; University of Tampere; Tampere Finland
- Faculty of Medicine and Life Sciences; University of Tampere; Tampere Finland
| |
Collapse
|
24
|
Chow M, Gao L, MacManiman JD, Bicocca VT, Chang BH, Alumkal JJ, Tyner JW. Maintenance and pharmacologic targeting of ROR1 protein levels via UHRF1 in t(1;19) pre-B-ALL. Oncogene 2018; 37:5221-5232. [PMID: 29849118 PMCID: PMC6150818 DOI: 10.1038/s41388-018-0299-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 04/04/2018] [Accepted: 04/13/2018] [Indexed: 12/23/2022]
Abstract
Expression of the transmembrane pseudokinase ROR1 is required for survival of t(1;19)-pre-B-cell acute lymphoblastic leukemia (t(1;19) pre-B-ALL), chronic lymphocytic leukemia, and many solid tumors. However, targeting ROR1 with small-molecules has been challenging due to the absence of ROR1 kinase activity. To identify genes that regulate ROR1 expression and may, therefore, serve as surrogate drug targets, we employed an siRNA screening approach and determined that the epigenetic regulator and E3 ubiquitin ligase, UHRF1, is required for t(1;19) pre-B-ALL cell viability in a ROR1-dependent manner. Upon UHRF1 silencing, ROR1 protein is reduced without altering ROR1 mRNA, and ectopically expressed UHRF1 is sufficient to increase ROR1 levels. Additionally, proteasome inhibition rescues loss of ROR1 protein after UHRF1 silencing, suggesting a role for the proteasome in the UHRF1-ROR1 axis. Finally, we show that ROR1-positive cells are twice as sensitive to the UHRF1-targeting drug, naphthazarin, and undergo increased apoptosis compared to ROR1-negative cells. Naphthazarin elicits reduced expression of UHRF1 and ROR1, and combination of naphthazarin with inhibitors of pre-B cell receptor signaling results in further reduction of cell survival compared with either inhibitor alone. Therefore, our work reveals a mechanism by which UHRF1 stabilizes ROR1, suggesting a potential targeting strategy to inhibit ROR1 in t(1;19) pre-B-ALL and other malignancies.
Collapse
MESH Headings
- CCAAT-Enhancer-Binding Proteins/deficiency
- CCAAT-Enhancer-Binding Proteins/genetics
- CCAAT-Enhancer-Binding Proteins/metabolism
- Cell Line, Tumor
- Cell Survival/drug effects
- Down-Regulation/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Silencing
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Molecular Targeted Therapy
- Naphthoquinones/pharmacology
- Naphthoquinones/therapeutic use
- Receptor Tyrosine Kinase-like Orphan Receptors/metabolism
- Ubiquitin-Protein Ligases
Collapse
Affiliation(s)
- Marilynn Chow
- Department of Cell, Developmental, and Cancer Biology, Oregon Health and Science University, Portland, USA
| | - Lina Gao
- Knight Cancer Institute, Oregon Health and Science University, Portland, USA
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, USA
| | - Jason D MacManiman
- Knight Cancer Institute, Oregon Health and Science University, Portland, USA
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, USA
| | - Vincent T Bicocca
- Knight Cancer Institute, Oregon Health and Science University, Portland, USA
| | - Bill H Chang
- Knight Cancer Institute, Oregon Health and Science University, Portland, USA
- Division of Pediatric Hematology and Oncology at Doernbecher Children's Hospital, Oregon Health and Science University, Portland, USA
| | - Joshi J Alumkal
- Knight Cancer Institute, Oregon Health and Science University, Portland, USA
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, USA
| | - Jeffrey W Tyner
- Department of Cell, Developmental, and Cancer Biology, Oregon Health and Science University, Portland, USA.
- Knight Cancer Institute, Oregon Health and Science University, Portland, USA.
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, USA.
| |
Collapse
|
25
|
Mani R, Goswami S, Gopalakrishnan B, Ramaswamy R, Wasmuth R, Tran M, Mo X, Gordon A, Bucci D, Lucas DM, Mims A, Brooks C, Dorrance A, Walker A, Blum W, Byrd JC, Lozanski G, Vasu S, Muthusamy N. The interleukin-3 receptor CD123 targeted SL-401 mediates potent cytotoxic activity against CD34 +CD123 + cells from acute myeloid leukemia/myelodysplastic syndrome patients and healthy donors. Haematologica 2018; 103:1288-1297. [PMID: 29773600 PMCID: PMC6068035 DOI: 10.3324/haematol.2018.188193] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/15/2018] [Indexed: 11/24/2022] Open
Abstract
Diseases with clonal hematopoiesis such as myelodysplastic syndrome and acute myeloid leukemia have high rates of relapse. Only a small subset of acute myeloid leukemia patients are cured with chemotherapy alone. Relapse in these diseases occurs at least in part due to the failure to eradicate leukemic stem cells or hematopoietic stem cells in myelodysplastic syndrome. CD123, the alpha chain of the interleukin-3 receptor heterodimer, is expressed on the majority of leukemic stem cells and myelodysplastic syndrome hematopoietic stem cells and in 80% of acute myeloid leukemia. Here, we report indiscriminate killing of CD123+ normal and acute myeloid leukemia / myelodysplastic syndrome cells by SL-401, a diphtheria toxin interleukin-3 fusion protein. SL-401 induced cytotoxicity of CD123+ primary cells/blasts from acute myeloid leukemia and myelodysplastic syndrome patients but not CD123− lymphoid cells. Importantly, SL-401 was highly active even in cells expressing low levels of CD123, with minimal effect on modulation of the CD123 target in acute myeloid leukemia. SL-401 significantly prolonged survival of leukemic mice in acute myeloid leukemia patient-derived xenograft mouse models. In addition to primary samples, studies on normal cord blood and healthy marrow show that SL-401 has activity against normal hematopoietic progenitors. These findings indicate potential use of SL-401 as a “bridge-to-transplant” before allogeneic hematopoietic cell transplantation in acute myeloid leukemia / myelodysplastic syndrome patients.
Collapse
Affiliation(s)
- Rajeswaran Mani
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Swagata Goswami
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | | | - Rahul Ramaswamy
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Ronni Wasmuth
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Minh Tran
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Xiaokui Mo
- Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - Amber Gordon
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Donna Bucci
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - David M Lucas
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Alice Mims
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | | | - Adrienne Dorrance
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Alison Walker
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - William Blum
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - John C Byrd
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Gerard Lozanski
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Sumithira Vasu
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Natarajan Muthusamy
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA .,Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
26
|
White C, Alshaker H, Cooper C, Winkler M, Pchejetski D. The emerging role of FTY720 (Fingolimod) in cancer treatment. Oncotarget 2018; 7:23106-27. [PMID: 27036015 PMCID: PMC5029614 DOI: 10.18632/oncotarget.7145] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 01/19/2016] [Indexed: 02/07/2023] Open
Abstract
FTY720 (Fingolimod) is a clinically approved immunomodulating therapy for multiple sclerosis that sequesters T-cells to lymph nodes through functional antagonism of sphingosine-1-phosphate 1 receptor. FTY720 also demonstrates a proven efficacy in multiple in vitro and in vivo cancer models, suggesting a potential therapeutic role in cancer patients. A potential anticancer mechanism of FTY720 is through the inhibition of sphingosine kinase 1, a proto-oncogene with in vitro and clinical cancer association. In addition, FTY720's anticancer properties may be attributable to actions on several other molecular targets. This study focuses on reviewing the emerging evidence regarding the anticancer properties and molecular targets of FTY720. While the clinical transition of FTY720 is currently limited by its immune suppression effects, studies aiming at FTY720 delivery and release together with identifying its key synergetic combinations and relevant patient subsets may lead to its rapid introduction into the clinic.
Collapse
Affiliation(s)
| | - Heba Alshaker
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan.,School of Medicine, University of East Anglia, Norwich, UK
| | - Colin Cooper
- School of Medicine, University of East Anglia, Norwich, UK
| | - Matthias Winkler
- Department of Surgery and Cancer, Imperial College London, London, UK
| | | |
Collapse
|
27
|
A systematic evaluation of the safety and toxicity of fingolimod for its potential use in the treatment of acute myeloid leukaemia. Anticancer Drugs 2017; 27:560-8. [PMID: 26967515 PMCID: PMC4881728 DOI: 10.1097/cad.0000000000000358] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Treatment of acute myeloid leukaemia (AML) is challenging and emerging treatment options include protein phosphatase 2A (PP2A) activators. Fingolimod is a known PP2A activator that inhibits multiple signalling pathways and has been used extensively in patients with multiple sclerosis and other indications. The initial positive results of PP2A activators in vitro and mouse models of AML are promising; however, its safety for use in AML has not been assessed. From human studies of fingolimod in other indications, it is possible to evaluate whether the safety and toxicity profile of the PP2A activators will allow their use in treating AML. A literature review was carried out to assess safety before the commencement of Phase I trials of the PP2A activator Fingolimod in AML. From human studies of fingolimod in other indications, it is possible to evaluate whether the safety and toxicity profile of the PP2A activators will allow their use in treating AML. A systematic review of published literature in Medline, EMBASE and the Cochrane Library of critical reviews was carried out. International standards for the design and reporting of search strategies were followed. Search terms and medical subject headings used in trials involving PP2A activators as well as a specific search were performed for ‘adverse events’, ‘serious adverse events’, ‘delays in treatment’, ‘ side effects’ and ‘toxicity’ for primary objectives. Database searches were limited to papers published in the last 12 years and available in English. The search yielded 677 articles. A total of 69 journal articles were identified as relevant and included 30 clinical trials, 24 review articles and 15 case reports. The most frequently reported adverse events were nausea, diarrhoea, fatigue, back pain, influenza viral infections, nasopharyngitis and bronchitis. Specific safety concerns include monitoring of the heart rate and conduction at commencement of treatment as cardiotoxicity has been reported. There is little evidence to suggest specific bone marrow toxicity. Lymophopenia is a desired effect in the management of multiple sclerosis, but may have implications in patients with acute leukaemia as it may potentially increase susceptibility to viral infections such as influenza. Fingolimod is a potential treatment option for AML with an acceptable risk to benefit ratio, given its lack of bone marrow toxicity and the relatively low rate of serious side effects. As most patients with AML are elderly, specific monitoring for cardiac toxicity as well as infection would be required.
Collapse
|
28
|
Therapeutic targeting of PP2A. Int J Biochem Cell Biol 2017; 96:182-193. [PMID: 29107183 DOI: 10.1016/j.biocel.2017.10.008] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/13/2017] [Accepted: 10/16/2017] [Indexed: 12/19/2022]
Abstract
Protein phosphatase 2A (PP2A) is a major serine/threonine phosphatase that regulates many cellular processes. Given the central role of PP2A in regulating diverse biological functions and its dysregulation in many diseases, including cancer, PP2A directed therapeutics have become of great interest. The main approaches leveraged thus far can be categorized as follows: 1) inhibiting endogenous inhibitors of PP2A, 2) targeted disruption of post translational modifications on PP2A subunits, or 3) direct targeting of PP2A. Additional insight into the structural, molecular, and biological framework driving the efficacy of these therapeutic strategies will provide a foundation for the refinement and development of novel and clinically tractable PP2A targeted therapies.
Collapse
|
29
|
Genetic evolution in chronic lymphocytic leukaemia. Best Pract Res Clin Haematol 2016; 29:67-78. [PMID: 27742073 DOI: 10.1016/j.beha.2016.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 06/30/2016] [Accepted: 08/04/2016] [Indexed: 11/21/2022]
Abstract
Next-generation sequencing provides a comprehensive understanding of the genomic, epigenomic and transcriptomic underpinnings of chronic lymphocytic leukaemia. Recent studies have uncovered new drivers, including mutations in non-coding regions, and signalling pathways whose role in cancer was previously unknown or poorly understood. Moreover, massive scale epigenomics and transcriptomics have supplied the foundations for the cellular origin of the disease. Some drivers could be targeted pharmacologically, and the ability to detect mutations present in minority subclones might even allow treatment before clonal selection occurs, thus preventing disease refractoriness. As our understanding broadens and ongoing technological innovation propels new achievements, we will certainly learn how to apply it in our daily practice.
Collapse
|
30
|
Poppova L, Janovska P, Plevova K, Radova L, Plesingerova H, Borsky M, Kotaskova J, Kantorova B, Hlozkova M, Figulova J, Brychtova Y, Machalova M, Urik M, Doubek M, Kozubik A, Pospisilova S, Pavlova S, Bryja V. DecreasedWNT3expression in chronic lymphocytic leukaemia is a hallmark of disease progression and identifies patients with worse prognosis in the subgroup with mutatedIGHV. Br J Haematol 2016; 175:851-859. [DOI: 10.1111/bjh.14312] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/13/2016] [Indexed: 10/21/2022]
Affiliation(s)
- Lucie Poppova
- Department of Internal Medicine-Hematology and Oncology; Center of Molecular Biology and Gene Therapy; University Hospital Brno and Medical Faculty; Masaryk University; Brno Czech Republic
- CEITEC - Central European Institute of Technology; Masaryk University; Brno Czech Republic
| | - Pavlina Janovska
- Institute of Experimental Biology; Faculty of Science; Masaryk University; Brno Czech Republic
| | - Karla Plevova
- Department of Internal Medicine-Hematology and Oncology; Center of Molecular Biology and Gene Therapy; University Hospital Brno and Medical Faculty; Masaryk University; Brno Czech Republic
- CEITEC - Central European Institute of Technology; Masaryk University; Brno Czech Republic
| | - Lenka Radova
- CEITEC - Central European Institute of Technology; Masaryk University; Brno Czech Republic
| | - Hana Plesingerova
- Department of Internal Medicine-Hematology and Oncology; Center of Molecular Biology and Gene Therapy; University Hospital Brno and Medical Faculty; Masaryk University; Brno Czech Republic
- CEITEC - Central European Institute of Technology; Masaryk University; Brno Czech Republic
| | - Marek Borsky
- Department of Internal Medicine-Hematology and Oncology; Center of Molecular Biology and Gene Therapy; University Hospital Brno and Medical Faculty; Masaryk University; Brno Czech Republic
| | - Jana Kotaskova
- Department of Internal Medicine-Hematology and Oncology; Center of Molecular Biology and Gene Therapy; University Hospital Brno and Medical Faculty; Masaryk University; Brno Czech Republic
- CEITEC - Central European Institute of Technology; Masaryk University; Brno Czech Republic
| | - Barbara Kantorova
- Department of Internal Medicine-Hematology and Oncology; Center of Molecular Biology and Gene Therapy; University Hospital Brno and Medical Faculty; Masaryk University; Brno Czech Republic
- CEITEC - Central European Institute of Technology; Masaryk University; Brno Czech Republic
| | - Michaela Hlozkova
- Department of Internal Medicine-Hematology and Oncology; Center of Molecular Biology and Gene Therapy; University Hospital Brno and Medical Faculty; Masaryk University; Brno Czech Republic
| | - Jana Figulova
- Department of Internal Medicine-Hematology and Oncology; Center of Molecular Biology and Gene Therapy; University Hospital Brno and Medical Faculty; Masaryk University; Brno Czech Republic
| | - Yvona Brychtova
- Department of Internal Medicine-Hematology and Oncology; Center of Molecular Biology and Gene Therapy; University Hospital Brno and Medical Faculty; Masaryk University; Brno Czech Republic
| | - Michaela Machalova
- Department of Paediatric Otorhinolaryngology; University Hospital Brno and Medical Faculty, MU; Brno Czech Republic
| | - Milan Urik
- Department of Paediatric Otorhinolaryngology; University Hospital Brno and Medical Faculty, MU; Brno Czech Republic
| | - Michael Doubek
- Department of Internal Medicine-Hematology and Oncology; Center of Molecular Biology and Gene Therapy; University Hospital Brno and Medical Faculty; Masaryk University; Brno Czech Republic
- CEITEC - Central European Institute of Technology; Masaryk University; Brno Czech Republic
| | - Alois Kozubik
- Institute of Experimental Biology; Faculty of Science; Masaryk University; Brno Czech Republic
- Department of Cytokinetics; Institute of Biophysics; Academy of Sciences of the Czech Republic; Brno Czech Republic
| | - Sarka Pospisilova
- Department of Internal Medicine-Hematology and Oncology; Center of Molecular Biology and Gene Therapy; University Hospital Brno and Medical Faculty; Masaryk University; Brno Czech Republic
- CEITEC - Central European Institute of Technology; Masaryk University; Brno Czech Republic
| | - Sarka Pavlova
- Department of Internal Medicine-Hematology and Oncology; Center of Molecular Biology and Gene Therapy; University Hospital Brno and Medical Faculty; Masaryk University; Brno Czech Republic
- CEITEC - Central European Institute of Technology; Masaryk University; Brno Czech Republic
| | - Vitezslav Bryja
- Institute of Experimental Biology; Faculty of Science; Masaryk University; Brno Czech Republic
- Department of Cytokinetics; Institute of Biophysics; Academy of Sciences of the Czech Republic; Brno Czech Republic
| |
Collapse
|
31
|
Chao K, Zhang S, Qiu Y, Chen X, Zhang X, Cai C, Peng Y, Mao R, Pevsner-Fischer M, Ben-horin S, Elinav E, Zeng Z, Chen B, He Y, Xiang AP, Chen M. Human umbilical cord-derived mesenchymal stem cells protect against experimental colitis via CD5(+) B regulatory cells. Stem Cell Res Ther 2016; 7:109. [PMID: 27515534 PMCID: PMC4981968 DOI: 10.1186/s13287-016-0376-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 07/13/2016] [Accepted: 07/26/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND To clarify the effect of human umbilical cord-derived mesenchymal stem cell (hUC-MSCs) treatment on colitis and to explore the role of CD5(+) B cells in MSC therapy. METHODS The trinitrobenzenesulfonic acid (TNBS)-induced colitis mouse model was used. HUC-MSCs were transferred peritoneally. Survival rates, colitis symptoms, and macroscopic and histologic scores were evaluated. CD4(+) T helper (Th) cell subgroups and CD5(+) regulatory B cell (Bregs) in lymphocytes were quantitated by flow cytometry. Cytokine levels were detected by ELISA and Bio-plex. CD5(+) B cells were isolated for in vitro co-culture and adaptive transfer. RESULTS HUC-MSC treatment alleviated TNBS-induced colitis by increasing survival rates, relieving symptoms, and improving macroscopic and histologic scores. Labeled hUC-MSCs were located in the inflamed areas of colitis mice. Increases in regulatory T cells (Tregs) and CD5(+) B cells and decreases in Th1 cells, Th17 cells, and several pro-inflammatory cytokines were observed with hUC-MSC treatment. After adaptive transfer, CD5(+) B cells, which were located mainly in the peritoneal lavage fluid, improved TNBS-induced colitis by correcting Treg/Th1/Th17 imbalances. CD5(+) B cells also inhibited T-cell proliferation and produced interleukin (IL)-10. CONCLUSIONS HUC-MSCs protected against experimental colitis by boosting the numbers of CD5(+) B cells and IL-10-producing CD5(+) Bregs, and correcting Treg/Th17/Th1 imbalances.
Collapse
Affiliation(s)
- Kang Chao
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080 People’s Republic of China
- Division of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655 People’s Republic of China
| | - Shenghong Zhang
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080 People’s Republic of China
| | - Yun Qiu
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080 People’s Republic of China
| | - Xiaoyong Chen
- Center for Stem Cell Biology and Tissue Engineering, The Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080 People’s Republic of China
| | - Xiaoran Zhang
- Center for Stem Cell Biology and Tissue Engineering, The Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080 People’s Republic of China
| | - Chuang Cai
- Center for Stem Cell Biology and Tissue Engineering, The Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080 People’s Republic of China
| | - Yanwen Peng
- Center for Stem Cell Biology and Tissue Engineering, The Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080 People’s Republic of China
| | - Ren Mao
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080 People’s Republic of China
| | | | - Shomron Ben-horin
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080 People’s Republic of China
| | - Eran Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot, 7610001 Israel
| | - Zhirong Zeng
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080 People’s Republic of China
| | - Baili Chen
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080 People’s Republic of China
| | - Yao He
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080 People’s Republic of China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, The Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080 People’s Republic of China
| | - Minhu Chen
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080 People’s Republic of China
| |
Collapse
|
32
|
Disruption and inactivation of the PP2A complex promotes the proliferation and angiogenesis of hemangioma endothelial cells through activating AKT and ERK. Oncotarget 2016; 6:25660-76. [PMID: 26308070 PMCID: PMC4694857 DOI: 10.18632/oncotarget.4705] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/15/2015] [Indexed: 12/31/2022] Open
Abstract
Hemangioma is a benign vascular neoplasm of unknown etiology. In this study, we generated an endothelial-specific PyMT gene-expressing transgenic mouse model that spontaneously develops hemangioma. Based on this transgenic model, a specific binding between PyMT and the core AC dimer of protein phosphatase 2A (PP2A) was verified in hemangioma vascular endothelial cells. The binding between PyMT and the PP2A AC dimer resulted in dissociation of the B subunit from the PP2A complex and inactivation of PP2A phosphatases, which in turn activated AKT and ERK signaling and promoted cell proliferation, migration and angiogenesis in vitro and tumorigenesis in vivo. Consistent with the in vitro findings, decreased PP2A phosphatase activity and disruption of the PP2A heterotrimeric complex were also observed in both primary transgene-positive TG(+) mouse hemangioma endothelial cells (TG(+) HEC cells) and human proliferating phase hemangioma endothelial (human HEC-P) cells, but not in transgene-negative TG(-) mouse normal vascular endothelial cells (TG(-) NEC cells) and human involuting phase hemangioma endothelial (human HEC-I) cells. Further, it was observed that in human hemangioma cells, endoglin could compete with the PP2A/A, C subunits for binding to the PP2A/B subunit, thereby resulting in dissociation of the B subunit from the PP2A complex. Treatment of Tie2/PyMT transgenic mice with the PP2A activator FTY720 significantly delayed the occurrence of hemangioma. Our data provide evidence of a previously unreported anti-proliferation and anti-angiogenesis effect of PP2A in vascular endothelial cells, and show the therapeutic value of PP2A activators in hemangioma.
Collapse
|
33
|
Pre-clinical Specificity and Safety of UC-961, a First-In-Class Monoclonal Antibody Targeting ROR1. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2016; 15 Suppl:S167-9. [PMID: 26297272 DOI: 10.1016/j.clml.2015.02.010] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 02/03/2015] [Indexed: 01/18/2023]
Abstract
Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is an oncoembryonic antigen. Because of its expression on the cell surface of leukemia cells from patients with chronic lymphocytic leukemia (CLL), but not on normal B-cells or other postpartum tissues, ROR1 is an attractive candidate for targeted therapies. UC-961 is a first-in-class humanized monoclonal antibody that binds the extracellular domain of ROR1. In this article we outline some of the preclinical studies leading to an investigational new drug designation, enabling clinical studies in patients with CLL.
Collapse
|
34
|
Mani R, Yan R, Mo X, Chen CS, Phelps MA, Klisovic R, Byrd JC, Kisseberth WC, London CA, Muthusamy N. Non-immunosuppressive FTY720-derivative OSU-2S mediates reactive oxygen species-mediated cytotoxicity in canine B-cell lymphoma. Vet Comp Oncol 2016; 15:1115-1118. [PMID: 27136276 DOI: 10.1111/vco.12221] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 12/29/2015] [Accepted: 01/19/2016] [Indexed: 02/03/2023]
Abstract
OSU-2S is a FTY720 (Fingolimod) derivative that lacks immunosuppressive properties but exhibits strong anti-tumour activity in several haematological and solid tumour models. We have recently shown OSU-2S to mediate potent cytotoxicity in human mantle cell lymphoma cell lines and primary cells. We report here the pre-clinical activity of OSU-2S in spontaneous B-cell lymphoma of dogs which shares many characteristics of human lymphoma. OSU-2S mediated apoptosis in canine B-cell lines and primary B-cell lymphoma cells obtained from spontaneous lymphoma bearing dogs. OSU-2S induced reactive oxygen species (ROS) in canine lymphoma cells and inhibition of ROS partially rescued OSU-2S-mediated cell death. These studies provide a rational basis for the use of spontaneous lymphoma in pet dogs as a preclinical large animal model for the development of OSU-2S as small molecule for treating people and dogs with lymphoma.
Collapse
Affiliation(s)
- R Mani
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - R Yan
- Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - X Mo
- Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - C-S Chen
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA.,Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - M A Phelps
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - R Klisovic
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - J C Byrd
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA.,Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, USA.,Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - W C Kisseberth
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - C A London
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - N Muthusamy
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA.,Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
35
|
Hong D, Zhang G, Zhang X, Liang X. Pulmonary Toxicities of Gefitinib in Patients With Advanced Non-Small-Cell Lung Cancer: A Meta-Analysis of Randomized Controlled Trials. Medicine (Baltimore) 2016; 95:e3008. [PMID: 26945426 PMCID: PMC4782910 DOI: 10.1097/md.0000000000003008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Gefitinib is a selective tyrosine kinase inhibitor of the epidermal growth factor receptor (EGFR) used to treat adults with EGFR mutation-positive non-small-cell lung cancer (NSCLC). Clinical benefits of gefitinib administration in NSCLC patients have been observed in clinical practice, but the extent of the pulmonary toxicity of gefitinib in patients with advanced NSCLC remains unclear. The aim of this systematic review was to evaluate the overall incidence and risk of gefitinib-related pulmonary toxicity in advanced NSCLC patients. Relevant trials were identified from the databases of Pubmed, Embase, Cochrane Library, and the clinicaltrials.gov of the U.S. National Institutes of Health. The outcomes included the overall incidence, odds ratios (ORs), and 95% confidence intervals (CIs). Fixed-effects models were used in the statistical analyses according to the heterogeneity of the included studies. According to the data from the included trials, the overall incidence of high-grade hemoptysis, pneumonia, pneumonitis, and interstitial lung disease (ILD) was 0.49% (95% CI: 0.24%-0.99%), 2.33% (95% CI: 1.47%-3.66%), 2.24% (95% CI: 1.34%-3.72%), and 1.43% (95% CI: 0.98%-2.09%), respectively. The pooled ORs of high-grade hemoptysis, pneumonia, pneumonitis, and ILD were 1.73 (95% CI: 0.46-6.52; P = 0.42), 0.99 (95% CI: 0.66-1.49; P = 0.95), 4.70 (95% CI: 1.48-14.95; P = 0.0087), and 2.64 (95% CI: 1.22-5.69; P = 0.01), respectively. Gefitinib was associated with a significantly increased risk of high-grade/fatal ILD and pneumonitis compared with the controls, whereas the risk of other high-grade pulmonary events (pneumonia and hemoptysis) was not significant. Careful surveillance of gefitinib-related pulmonary toxicity is critical for the safe use of this drug.
Collapse
Affiliation(s)
- Dongsheng Hong
- From the Department of Pharmacy (DH, GZ, XZ), The First Affiliated Hospital of College of Medicine, Zhejiang University; and Central Laboratory (XL), The First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou, China
| | | | | | | |
Collapse
|
36
|
Kutsch N, Marks R, Ratei R, Held TK, Schmidt-Hieber M. Role of Tyrosine Kinase Inhibitors in Indolent and Other Mature B-Cell Neoplasms. Biomark Insights 2015; 10:15-23. [PMID: 26327780 PMCID: PMC4539014 DOI: 10.4137/bmi.s22434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/30/2015] [Accepted: 07/02/2015] [Indexed: 12/26/2022] Open
Abstract
Targeting tyrosine kinases represents a highly specific treatment approach for different malignancies. This also includes non-Hodgkin lymphoma since it is well known that these enzymes are frequently involved in the lymphomagenesis. Hereby, tyrosine kinases might either be dysregulated intrinsically or be activated within signal transduction pathways leading to tumor survival and growth. Among others, Bruton's tyrosine kinase (Btk) is of particular interest as a potential therapeutic target. Btk is stimulated by B-cell receptor signaling and activates different transcription factors such as nuclear factor κB. The Btk inhibitor ibrutinib has been approved for the treatment of chronic lymphocytic leukemia and mantle-cell lymphoma recently. Numerous clinical trials evaluating this agent in different combinations (eg, with rituximab or classical chemotherapeutic agents) as a treatment option for aggressive and indolent lymphoma are under way. Here, we summarize the role of tyrosine kinase inhibitors in the treatment of indolent and other non-Hodgkin lymphomas (eg, mantle-cell lymphoma).
Collapse
Affiliation(s)
- Nadine Kutsch
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn, University of Cologne, Cologne, Germany
| | - Reinhard Marks
- Clinic for Hematology, Oncology and Stem Cell Transplantation, University Hospital of Freiburg, Freiburg, Germany
| | - Richard Ratei
- Department of Hematology, Oncology and Tumor Immunology, HELIOS Clinic Berlin-Buch, Berlin, Germany
| | - Thomas K Held
- Department of Hematology, Oncology and Tumor Immunology, HELIOS Clinic Berlin-Buch, Berlin, Germany
| | - Martin Schmidt-Hieber
- Department of Hematology, Oncology and Tumor Immunology, HELIOS Clinic Berlin-Buch, Berlin, Germany
| |
Collapse
|
37
|
Lin B, Gao A, Zhang R, Ma H, Shen H, Hu Q, Zhang H, Zhao M, Lan X, Liu K. Use of a Novel Integrase-Deficient Lentivirus for Targeted Anti-Cancer Therapy With Survivin Promoter-Driven Diphtheria Toxin A. Medicine (Baltimore) 2015; 94:e1301. [PMID: 26252309 PMCID: PMC4616595 DOI: 10.1097/md.0000000000001301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
As an immunotoxin, diphtheria toxin has been widely used in gene therapy and gene function assays for its roles in protein synthesis inhibition, and the aim of our study is to set up a nonintegrating lentiviral system for specific expression of diphtheria toxin A (DTA) used in cancer gene therapy.Here, we established a lentiviral system that could coordinately express fluorescent protein and DTA driven by the cytomegalovirus (CMV) promoter, which is convenient for us to precisely trace the expression of DTA and monitor the process of lentivirus packaging. To achieve safer cancer therapy, we replaced the CMV promoter with the Survivin promoter, a specific promoter that is dramatically activated in cancer tissues and cells, but not in normal tissues and cells, and that will impose greater therapeutic potential because a significant expression difference occurred between these 2 groups. Meanwhile, we obtained integrase-deficient lentivirus (IDLV) after packaging with the integrase mutant, which expresses defective integrase RRK262263264AAH, to minimize the side effects that derived from the insertional mutagenesis of the host genome.Our results suggest that the IDLV system that we generated possesses therapeutic potential in cancers in vitro and in vivo.
Collapse
Affiliation(s)
- Baoshun Lin
- From the Institute for Laboratory Medicine, Fuzhou General Hospital, PLA, Fuzhou, Fujian, P.R. China (BL, AG, RZ, HM, MZ, XL, KL); Dong Fang Hospital, Xiamen University, Fuzhou, Fujian, P.R. China (BL, AG, QH, MZ, XL, KL); Institute for Laboratory Medicine, Fuzhou General Hospital, Second Military Medical University, Fuzhou, Fujian, P.R. China (HM); School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian, P.R. China (HS); Agriculture and Animal Husbandry College, Tibet University, Nyingchi, P.R. China (HZ); and Fuzhou General Hospital Clinical Medical School, Fujian Medical University, Fuzhou, P.R. China (RZ, XL, KL)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Patmanathan SN, Yap LF, Murray PG, Paterson IC. The antineoplastic properties of FTY720: evidence for the repurposing of fingolimod. J Cell Mol Med 2015; 19:2329-40. [PMID: 26171944 PMCID: PMC4594675 DOI: 10.1111/jcmm.12635] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 05/20/2015] [Indexed: 12/20/2022] Open
Abstract
Almost all drugs approved for use in humans possess potentially beneficial 'off-target' effects in addition to their principal activity. In some cases this has allowed for the relatively rapid repurposing of drugs for other indications. In this review we focus on the potential for re-purposing FTY720 (also known as fingolimod, Gilenya(™)), an immunomodulatory drug recently approved for the treatment of multiple sclerosis (MS). The therapeutic benefit of FTY720 in MS is largely attributed to the immunosuppressive effects that result from its modulation of sphingosine 1-phosphate receptor signalling. However, this drug has also been shown to inhibit other cancer-associated signal transduction pathways in part because of its structural similarity to sphingosine, and consequently shows efficacy as an anti-cancer agent both in vitro and in vivo. Here, we review the effects of FTY720 on signal transduction pathways and cancer-related cellular processes, and discuss its potential use as an anti-cancer drug.
Collapse
Affiliation(s)
- Sathya Narayanan Patmanathan
- Department of Oral Biology and Biomedical Sciences and Oral Cancer Research & Coordinating Centre, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Lee Fah Yap
- Department of Oral Biology and Biomedical Sciences and Oral Cancer Research & Coordinating Centre, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Paul G Murray
- School of Cancer Sciences, University of Birmingham, Birmingham, UK
| | - Ian C Paterson
- Department of Oral Biology and Biomedical Sciences and Oral Cancer Research & Coordinating Centre, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
39
|
Lyn sustains oncogenic signaling in chronic lymphocytic leukemia by strengthening SET-mediated inhibition of PP2A. Blood 2015; 125:3747-55. [DOI: 10.1182/blood-2014-12-619155] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 04/21/2015] [Indexed: 01/12/2023] Open
Abstract
Key Points
Cytosolic HSP90-bound Lyn mediates resistance to apoptosis by strengthening PP2A/SET interaction in CLL cells. FTY720-analogues antagonizing the PP2A/SET interaction and Lyn inhibitors may provide a therapeutic approach of CLL.
Collapse
|
40
|
Mani R, Chiang CL, Frissora FW, Yan R, Mo X, Baskar S, Rader C, Klisovic R, Phelps MA, Chen CS, Lee RJ, Byrd JC, Baiocchi R, Lee LJ, Muthusamy N. ROR1-targeted delivery of OSU-2S, a nonimmunosuppressive FTY720 derivative, exerts potent cytotoxicity in mantle-cell lymphoma in vitro and in vivo. Exp Hematol 2015; 43:770-4.e2. [PMID: 25937048 DOI: 10.1016/j.exphem.2015.04.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 04/03/2015] [Accepted: 04/21/2015] [Indexed: 01/09/2023]
Abstract
Mantle-cell lymphoma (MCL) remains incurable despite numerous therapeutic advances. OSU-2S, a novel nonimmunosuppressive FTY720 (Fingolimod) derivative, exhibits potent cytotoxicity in MCL cell lines and primary cells. OSU-2S increased the surface expression of CD74, a therapeutic antibody target in MCL cells. OSU-2S, in combination with anti-CD74 antibody milatuzumab, enhanced cytotoxicity in MCL. Moreover, MCL tumor antigen receptor tyrosine kinase-like orphan receptor 1 (ROR1) targeted immunonanoparticle-carrying OSU-2S (2A2-OSU-2S-ILP)-mediated selective cytotoxicity of MCL in vitro, as well as activity in a xenografted mouse model of MCL in vivo. The newly developed OSU-2S delivery using ROR1-directed immunonanoparticles provide selective targeting of OSU-2S to MCL and other ROR1(+) malignancies, sparing normal B cells.
Collapse
Affiliation(s)
- Rajeswaran Mani
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Chi-Ling Chiang
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Center for Affordable Nanoengineering of Polymeric Biomedical Devices, The Ohio State University, Columbus, OH, USA
| | - Frank W Frissora
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Ribai Yan
- Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Xiaokui Mo
- Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - Sivasubramanian Baskar
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Christoph Rader
- Department of Cancer Biology and Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida, Jupiter, FL, USA
| | - Rebecca Klisovic
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Mitch A Phelps
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Ching-Shih Chen
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA; Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Robert J Lee
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Center for Affordable Nanoengineering of Polymeric Biomedical Devices, The Ohio State University, Columbus, OH, USA; Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - John C Byrd
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA; Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Robert Baiocchi
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - L James Lee
- Center for Affordable Nanoengineering of Polymeric Biomedical Devices, The Ohio State University, Columbus, OH, USA; Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Natarajan Muthusamy
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
41
|
Switching the sphingolipid rheostat in the treatment of diabetes and cancer comorbidity from a problem to an advantage. BIOMED RESEARCH INTERNATIONAL 2015; 2015:165105. [PMID: 25866760 PMCID: PMC4383402 DOI: 10.1155/2015/165105] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/16/2014] [Indexed: 12/11/2022]
Abstract
Cancer and diabetes are among the most common diseases in western societies. Epidemiological studies have shown that diabetic patients have a significantly higher risk of developing a number of different types of cancers and that individuals with comorbidity (cancer and diabetes/prediabetes) have a poorer prognosis relative to nondiabetic cancer patients. The increasing frequency of comorbidity of cancer and diabetes mellitus, mainly type 2 diabetes, has driven the development of therapeutic interventions that target both disease states. There is strong evidence to suggest that balancing the sphingolipid rheostat, ceramide--sphingosine--sphingosine-1-phosphate (S1P) is crucial in the prevention of diabetes and cancer and sphingosine kinase/S1P modulators are currently under development for the treatment of cancer and diabetes. This paper will highlight some of the complexities inherent in the use of the emerging sphingosine kinase/S1P modulators in the treatment of comorbidity of diabetes and cancer.
Collapse
|