1
|
Hughes K, Evans K, Earley EJ, Smith CM, Erickson SW, Stearns T, Philip VM, Neuhauser SB, Chuang JH, Jocoy EL, Bult CJ, Teicher BA, Smith MA, Lock RB. In vivo activity of the dual SYK/FLT3 inhibitor TAK-659 against pediatric acute lymphoblastic leukemia xenografts. Pediatr Blood Cancer 2023; 70:e30503. [PMID: 37339930 PMCID: PMC10730772 DOI: 10.1002/pbc.30503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/27/2023] [Accepted: 06/04/2023] [Indexed: 06/22/2023]
Abstract
BACKGROUND While children with acute lymphoblastic leukemia (ALL) experience close to a 90% likelihood of cure, the outcome for certain high-risk pediatric ALL subtypes remains dismal. Spleen tyrosine kinase (SYK) is a prominent cytosolic nonreceptor tyrosine kinase in pediatric B-lineage ALL (B-ALL). Activating mutations or overexpression of Fms-related receptor tyrosine kinase 3 (FLT3) are associated with poor outcome in hematological malignancies. TAK-659 (mivavotinib) is a dual SYK/FLT3 reversible inhibitor, which has been clinically evaluated in several other hematological malignancies. Here, we investigate the in vivo efficacy of TAK-659 against pediatric ALL patient-derived xenografts (PDXs). METHODS SYK and FLT3 mRNA expression was quantified by RNA-seq. PDX engraftment and drug responses in NSG mice were evaluated by enumerating the proportion of human CD45+ cells (%huCD45+ ) in the peripheral blood. TAK-659 was administered per oral at 60 mg/kg daily for 21 days. Events were defined as %huCD45+ ≥ 25%. In addition, mice were humanely killed to assess leukemia infiltration in the spleen and bone marrow (BM). Drug efficacy was assessed by event-free survival and stringent objective response measures. RESULTS FLT3 and SYK mRNA expression was significantly higher in B-lineage compared with T-lineage PDXs. TAK-659 was well tolerated and significantly prolonged the time to event in six out of eight PDXs tested. However, only one PDX achieved an objective response. The minimum mean %huCD45+ was significantly reduced in five out of eight PDXs in TAK-659-treated mice compared with vehicle controls. CONCLUSIONS TAK-659 exhibited low to moderate single-agent in vivo activity against pediatric ALL PDXs representative of diverse subtypes.
Collapse
Affiliation(s)
- Keira Hughes
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Kathryn Evans
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Eric J Earley
- RTI International, Research Triangle Park, North Carolina, USA
| | - Christopher M Smith
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | | | - Tim Stearns
- The Jackson Laboratory, Bar Harbor, Maine, USA
| | | | | | | | | | | | | | | | - Richard B Lock
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
2
|
Wang ZW, Zou FM, Wang AL, Yang J, Jin R, Wang BL, Shen LJ, Qi S, Liu J, Liu J, Wang WC, Liu QS. Repurposing of the FGFR inhibitor AZD4547 as a potent inhibitor of necroptosis by selectively targeting RIPK1. Acta Pharmacol Sin 2023; 44:801-810. [PMID: 36216899 PMCID: PMC10042809 DOI: 10.1038/s41401-022-00993-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/30/2022] [Indexed: 11/10/2022] Open
Abstract
Necroptosis is a form of regulated necrosis involved in various pathological diseases. The process of necroptosis is controlled by receptor-interacting kinase 1 (RIPK1), RIPK3, and pseudokinase mixed lineage kinase domain-like protein (MLKL), and pharmacological inhibition of these kinases has been shown to have therapeutic potentials in a variety of diseases. In this study, using drug repurposing strategy combined with high-throughput screening (HTS), we discovered that AZD4547, a previously reported FGFR inhibitor, is able to interfere with necroptosis through direct targeting of RIPK1 kinase. In both human and mouse cell models, AZD4547 blocked RIPK1-dependent necroptosis. In addition, AZD4547 rescued animals from TNF-induced lethal shock and inflammatory responses. Together, our study demonstrates that AZD4547 is a potent and selective inhibitor of RIPK1 with therapeutic potential for the treatment of inflammatory disorders that involve necroptosis.
Collapse
Affiliation(s)
- Zuo-Wei Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Feng-Ming Zou
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Ao-Li Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Jing Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Rui Jin
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Bei-Lei Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Li-Juan Shen
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Shuang Qi
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Juan Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Jing Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China.
| | - Wen-Chao Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China.
| | - Qing-Song Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- University of Science and Technology of China, Hefei, 230026, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China.
- Precision Medicine Research Laboratory of Anhui Province, Hefei, 230088, China.
| |
Collapse
|
3
|
Tang S, Yu Q, Ding C. Investigational spleen tyrosine kinase (SYK) inhibitors for the treatment of autoimmune diseases. Expert Opin Investig Drugs 2022; 31:291-303. [PMID: 35130124 DOI: 10.1080/13543784.2022.2040014] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Autoimmune diseases (ADs) are disorders induced by multiple inflammatory mediators, in which immune system attacks healthy tissues and triggers tissue injury. Targeted regulation of the activity of kinases that influence inflammation is one of the major therapies for ADs. Recently, investigational spleen tyrosine kinase (SYK) inhibitors have shown encouraging results in the ADs therapy. AREAS COVERED This article provides a background on autoimmune diseases and provides an update on investigational SYK inhibitors. This literature review was conducted by searching publications about investigational Spleen tyrosine kinase inhibitors in the treatment of ADs from experimental to clinical studies. The search terms used were SYK inhibitors, R406, fostamatinib (R788), P505-15 (PRT062607), entospletinib (GS-9973), R112, lanraplenib (GS-9876), cerdulatinib, R343, BAY-61-3606, GSK compound 143 (GSK143), R211, SKI-G-618, SKI-O-85, ER-27319, YM193306, RO9021 in conjunction with autoimmune disease using electronic databases including PubMed, EMBASE, MEDLINE and Google Scholar. EXPERT OPINION SYK inhibitors are promising drugs with unique advantages and acceptable tolerability and safety for the treatment of ADs. However, the difficulties in developing highly selective SYK inhibitors and the unknown effects are challenges. Long term and real-world data are essential to determine the risk-benefit ratio and true role of SYK inhibitors in the therapy of ADs.
Collapse
Affiliation(s)
- Su'an Tang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Centre of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qinghong Yu
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
4
|
Rajakumar SA, Papp E, Lee KK, Grandal I, Merico D, Liu CC, Allo B, Zhang L, Grynpas MD, Minden MD, Hitzler JK, Guidos CJ, Danska JS. B cell acute lymphoblastic leukemia cells mediate RANK-RANKL-dependent bone destruction. Sci Transl Med 2021; 12:12/561/eaba5942. [PMID: 32938796 DOI: 10.1126/scitranslmed.aba5942] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 06/05/2020] [Accepted: 07/21/2020] [Indexed: 12/20/2022]
Abstract
Although most children survive B cell acute lymphoblastic leukemia (B-ALL), they frequently experience long-term, treatment-related health problems, including osteopenia and osteonecrosis. Because some children present with fractures at ALL diagnosis, we considered the possibility that leukemic B cells contribute directly to bone pathology. To identify potential mechanisms of B-ALL-driven bone destruction, we examined the p53 -/-; Rag2 -/-; Prkdcscid/scid triple mutant (TM) mice and p53 -/-; Prkdcscid/scid double mutant (DM) mouse models of spontaneous B-ALL. In contrast to DM animals, leukemic TM mice displayed brittle bones, and the TM leukemic cells overexpressed Rankl, encoding receptor activator of nuclear factor κB ligand. RANKL is a key regulator of osteoclast differentiation and bone loss. Transfer of TM leukemic cells into immunodeficient recipient mice caused trabecular bone loss. To determine whether human B-ALL can exert similar effects, we evaluated primary human B-ALL blasts isolated at diagnosis for RANKL expression and their impact on bone pathology after their transplantation into NOD.Prkdcscid/scidIl2rgtm1Wjl /SzJ (NSG) recipient mice. Primary B-ALL cells conferred bone destruction evident in increased multinucleated osteoclasts, trabecular bone loss, destruction of the metaphyseal growth plate, and reduction in adipocyte mass in these patient-derived xenografts (PDXs). Treating PDX mice with the RANKL antagonist recombinant osteoprotegerin-Fc (rOPG-Fc) protected the bone from B-ALL-induced destruction even under conditions of heavy tumor burden. Our data demonstrate a critical role of the RANK-RANKL axis in causing B-ALL-mediated bone pathology and provide preclinical support for RANKL-targeted therapy trials to reduce acute and long-term bone destruction in these patients.
Collapse
Affiliation(s)
- Sujeetha A Rajakumar
- Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Eniko Papp
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Kathy K Lee
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5T 3H7, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Ildiko Grandal
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Daniele Merico
- Center for Applied Genomics, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Careesa C Liu
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5T 3H7, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Bedilu Allo
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5T 3H7, Canada
| | - Lucia Zhang
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5T 3H7, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Marc D Grynpas
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5T 3H7, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Mark D Minden
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Princess Margaret Cancer Center, University Health Network, Toronto, Ontario M5G 2M9, Canada
| | - Johann K Hitzler
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Pediatrics, Division of Hematology and Oncology, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Cynthia J Guidos
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jayne S Danska
- Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
5
|
Loftus JP, Yahiaoui A, Brown PA, Niswander LM, Bagashev A, Wang M, Schauf A, Tannheimer S, Tasian SK. Combinatorial efficacy of entospletinib and chemotherapy in patient-derived xenograft models of infant acute lymphoblastic leukemia. Haematologica 2021; 106:1067-1078. [PMID: 32414848 PMCID: PMC8018117 DOI: 10.3324/haematol.2019.241729] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Indexed: 12/21/2022] Open
Abstract
Survival of infants with KMT2A-rearranged acute lymphoblastic leukemia (ALL) remains dismal despite intensive chemotherapy. We observed constitutive phosphorylation of spleen tyrosine kinase (SYK) and associated signaling proteins in infant ALL patient-derived xenograft (PDX) model specimens and hypothesized that the SYK inhibitor entospletinib would inhibit signaling and cell growth in vitro and leukemia proliferation in vivo. We further predicted that combined entospletinib and chemotherapy could augment anti-leukemia effects. Basal kinase signaling activation and HOXA9/MEIS1 expression differed among KMT2Arearranged (KMT2A-AFF1 [n=4], KMT2A-MLLT3 [n=1], KMT2A-MLLT1 [n=4]) and non-KMT2A-rearranged [n=3] ALL specimens and stratified by genetic subgroup. Incubation of KMT2A-rearranged ALL cells in vitro with entospletinib inhibited methylcellulose colony formation and SYK pathway signaling in a dose-dependent manner. In vivo inhibition of leukemia proliferation with entospletinib monotherapy was observed in RAS-wild-type KMT2A-AFF1, KMT2A-MLLT3, and KMT2A-MLLT1 ALL PDX models with enhanced activity in combination with vincristine chemotherapy in several models. Surprisingly, entospletinib did not decrease leukemia burden in two KMT2A-AFF1 PDX models with NRAS or KRAS mutations, suggesting potential RAS-mediated resistance to SYK inhibition. As hypothesized, superior inhibition of ALL proliferation was observed in KMT2A-AFF1 PDX models treated with entospletinib and the MEK inhibitor selumetinib versus vehicle or inhibitor monotherapies (P<0.05). In summary, constitutive activation of SYK and associated signaling occurs in KMT2A-rearranged ALL with in vitro and in vivo sensitivity to entospletinib. Combination therapy with vincristine or selumetinib further enhanced treatment effects of SYK inhibition. Clinical study of entospletinib and chemotherapy or other kinase inhibitors in patients with KMT2A-rearranged leukemias may be warranted.
Collapse
Affiliation(s)
- Joseph P Loftus
- Div of Oncology, Children Hospital and Center for Childhood Cancer Research, Philadelphia, USA
| | | | - Patrick A Brown
- Johns Hopkins University and Sidney Kimmel Comprehensive Cancer Center, Baltimore, USA
| | - Lisa M Niswander
- Div of Oncology, Children Hospital and Center for Childhood Cancer Research, Philadelphia, USA
| | - Asen Bagashev
- Div of Oncology, Children Hospital and Center for Childhood Cancer Research, Philadelphia, USA
| | - Min Wang
- Gilead Sciences; Foster City, CA, USA
| | | | | | - Sarah K Tasian
- Div of Oncology, Children Hospital and Center for Childhood Cancer Research, Philadelphia, USA
| |
Collapse
|
6
|
Buffard M, Naldi A, Freiss G, Deckert M, Radulescu O, Coopman PJ, Larive RM. Comparison of SYK Signaling Networks Reveals the Potential Molecular Determinants of Its Tumor-Promoting and Suppressing Functions. Biomolecules 2021; 11:biom11020308. [PMID: 33670716 PMCID: PMC7923165 DOI: 10.3390/biom11020308] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/10/2021] [Accepted: 02/14/2021] [Indexed: 01/18/2023] Open
Abstract
Spleen tyrosine kinase (SYK) can behave as an oncogene or a tumor suppressor, depending on the cell and tissue type. As pharmacological SYK inhibitors are currently evaluated in clinical trials, it is important to gain more information on the molecular mechanisms underpinning these opposite roles. To this aim, we reconstructed and compared its signaling networks using phosphoproteomic data from breast cancer and Burkitt lymphoma cell lines where SYK behaves as a tumor suppressor and promoter. Bioinformatic analyses allowed for unveiling the main differences in signaling pathways, network topology and signal propagation from SYK to its potential effectors. In breast cancer cells, the SYK target-enriched signaling pathways included intercellular adhesion and Hippo signaling components that are often linked to tumor suppression. In Burkitt lymphoma cells, the SYK target-enriched signaling pathways included molecules that could play a role in SYK pro-oncogenic function in B-cell lymphomas. Several protein interactions were profoundly rewired in the breast cancer network compared with the Burkitt lymphoma network. These data demonstrate that proteomic profiling combined with mathematical network modeling allows untangling complex pathway interplays and revealing difficult to discern interactions among the SYK pathways that positively and negatively affect tumor formation and progression.
Collapse
Affiliation(s)
- Marion Buffard
- IRCM, Université de Montpellier, ICM, INSERM, F-34298 Montpellier, France; (M.B.); (G.F.); (P.J.C.)
- LPHI, Université de Montpellier, CNRS, F-34095 Montpellier, France;
| | - Aurélien Naldi
- Institut de Biologie de l'ENS (IBENS), Département de Biologie, École Normale Supérieure, CNRS, INSERM, Université PSL, F-75005 Paris, France;
- Lifeware Group, Inria Saclay-île de France, F-91120 Palaiseau, France
| | - Gilles Freiss
- IRCM, Université de Montpellier, ICM, INSERM, F-34298 Montpellier, France; (M.B.); (G.F.); (P.J.C.)
| | - Marcel Deckert
- C3M, Université Côte d'Azur, INSERM, équipe «Microenvironnement, Signalisation et Cancer», F-06204 Nice, France;
| | - Ovidiu Radulescu
- LPHI, Université de Montpellier, CNRS, F-34095 Montpellier, France;
| | - Peter J. Coopman
- IRCM, Université de Montpellier, ICM, INSERM, F-34298 Montpellier, France; (M.B.); (G.F.); (P.J.C.)
- CNRS—Centre National de la Recherche Scientifique, 1919 Route de Mende, F-34293 Montpellier, France
| | - Romain M. Larive
- IRCM, Université de Montpellier, ICM, INSERM, F-34298 Montpellier, France; (M.B.); (G.F.); (P.J.C.)
- IBMM, Université Montpellier, CNRS, ENSCM, F-34093 Montpellier, France
- Correspondence: ; Tel.: +33-467-61-24-30; Fax: +33-467-61-37-87
| |
Collapse
|
7
|
Sender S, Sekora A, Villa Perez S, Chabanovska O, Becker A, Ngezahayo A, Junghanss C, Murua Escobar H. Precursor B-ALL Cell Lines Differentially Respond to SYK Inhibition by Entospletinib. Int J Mol Sci 2021; 22:E592. [PMID: 33435587 PMCID: PMC7827334 DOI: 10.3390/ijms22020592] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Impaired B-cell receptor (BCR) function has been associated with the progress of several B-cell malignancies. The spleen tyrosine kinase (SYK) represents a potential therapeutic target in a subset of B-cell neoplasias. In precursor B-acute lymphoblastic leukemia (B-ALL), the pathogenic role and therapeutic potential of SYK is still controversially discussed. We evaluate the application of the SYK inhibitor entospletinib (Ento) in pre- and pro-B-ALL cell lines, characterizing the biologic and molecular effects. METHODS SYK expression was characterized in pre-B-ALL (NALM-6) and pro-B-ALL cell lines (SEM and RS4;11). The cell lines were exposed to different Ento concentrations and the cell biological response analyzed by proliferation, metabolic activity, apoptosis induction, cell-cycle distribution and morphology. BCR pathway gene expression and protein modulations were further characterized. RESULTS Ento significantly induced anti-proliferative and pro-apoptotic effects in NALM-6 and SEM, while barely affecting RS4;11. Targeted RNAseq revealed pronounced gene expression modulation only in NALM-6, while Western Blot analyses demonstrated that vital downstream effector proteins, such as pAKT, pERK, pGSK3β, p53 and BCL-6, were affected by Ento exposure in the inhibitor-sensitive cell lines. CONCLUSION Different acting modes of Ento, independent of pre-BCR dependency, were characterized, unexpected in SEM. Accordingly, SYK classifies as a potential target structure in a subset of pro-B-ALLs.
Collapse
Affiliation(s)
- Sina Sender
- Division of Medicine, Department of Hematology, Oncology and Palliative Medicine, University of Rostock, 18057 Rostock, Germany; (S.S.); (A.S.); (S.V.P.); (O.C.); (C.J.)
| | - Anett Sekora
- Division of Medicine, Department of Hematology, Oncology and Palliative Medicine, University of Rostock, 18057 Rostock, Germany; (S.S.); (A.S.); (S.V.P.); (O.C.); (C.J.)
| | - Simon Villa Perez
- Division of Medicine, Department of Hematology, Oncology and Palliative Medicine, University of Rostock, 18057 Rostock, Germany; (S.S.); (A.S.); (S.V.P.); (O.C.); (C.J.)
| | - Oleksandra Chabanovska
- Division of Medicine, Department of Hematology, Oncology and Palliative Medicine, University of Rostock, 18057 Rostock, Germany; (S.S.); (A.S.); (S.V.P.); (O.C.); (C.J.)
| | - Annegret Becker
- Department of Cell Physiology and Biophysics, Institute of Cell Biology and Biophysics, Leibniz University Hannover, 30419 Hannover, Germany; (A.B.); (A.N.)
| | - Anaclet Ngezahayo
- Department of Cell Physiology and Biophysics, Institute of Cell Biology and Biophysics, Leibniz University Hannover, 30419 Hannover, Germany; (A.B.); (A.N.)
| | - Christian Junghanss
- Division of Medicine, Department of Hematology, Oncology and Palliative Medicine, University of Rostock, 18057 Rostock, Germany; (S.S.); (A.S.); (S.V.P.); (O.C.); (C.J.)
| | - Hugo Murua Escobar
- Division of Medicine, Department of Hematology, Oncology and Palliative Medicine, University of Rostock, 18057 Rostock, Germany; (S.S.); (A.S.); (S.V.P.); (O.C.); (C.J.)
| |
Collapse
|
8
|
Davidzohn N, Biram A, Stoler-Barak L, Grenov A, Dassa B, Shulman Z. Syk degradation restrains plasma cell formation and promotes zonal transitions in germinal centers. J Exp Med 2020; 217:133542. [PMID: 31873727 PMCID: PMC7062533 DOI: 10.1084/jem.20191043] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/08/2019] [Accepted: 11/18/2019] [Indexed: 12/14/2022] Open
Abstract
In germinal centers, B cells interact with antigen in the light zone and clonally expand in the dark zone. Davidzohn et al. show that BCR-induced Syk degradation in the light zone attenuates signal transduction, impedes plasma cell formation, and promotes B cell transition to the dark zone. Germinal centers (GCs) are sites at which B cells proliferate and mutate their antibody-encoding genes in the dark zone (DZ), followed by affinity-based selection in the light zone (LZ). B cell antigen receptor (BCR) signals induce Syk activation followed by rapid phosphatase-mediated desensitization; however, how degradation events regulate BCR functions in GCs is unclear. Here, we found that Syk degradation restrains plasma cell (PC) formation in GCs and promotes B cell LZ to DZ transition. Using a mouse model defective in Cbl-mediated Syk degradation, we demonstrate that this machinery attenuates BCR signaling intensity by mitigating the Kras/Erk and PI3K/Foxo1 pathways, and restricting the expression of PC transcription factors in GC B cells. Inhibition of Syk degradation perturbed gene expression, specifically in the LZ, and enhanced the generation of PCs without affecting B cell proliferation. These findings reveal how long-lasting attenuation of signal transduction by degradation events regulates cell fate within specialized microanatomical sites.
Collapse
Affiliation(s)
- Natalia Davidzohn
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Adi Biram
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Liat Stoler-Barak
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Amalie Grenov
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Bareket Dassa
- Department of Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Ziv Shulman
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
9
|
Jin W. The Role of Tyrosine Kinases as a Critical Prognostic Parameter and Its Targeted Therapies in Ewing Sarcoma. Front Cell Dev Biol 2020; 8:613. [PMID: 32754598 PMCID: PMC7381324 DOI: 10.3389/fcell.2020.00613] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
Ewing sarcoma (ES) is a rare, highly aggressive, bone, or soft tissue-associated tumor. Although this sarcoma often responds well to initial chemotherapy, 40% of the patients develop a lethal recurrence of the disease, with death recorded in 75-80% of patients with metastatic ES within 5 years, despite receiving high-dose chemotherapy. ES is genetically well-characterized, as indicated by the EWS-FLI1 fusion protein encoded as a result of chromosomal translocation in 80-90% of patients with ES, as well as in ES-related cancer cell lines. Recently, tyrosine kinases have been identified in the pathogenesis of ES. These tyrosine kinases, acting as oncoproteins, are associated with the clinical pathogenesis, metastasis, acquisition of self-renewal traits, and chemoresistance of ES, through the activation of various intracellular signaling pathways. This review describes the recent progress related to cellular and molecular functional roles of tyrosine kinases in the progression of ES.
Collapse
Affiliation(s)
- Wook Jin
- Laboratory of Molecular Disease and Cell Regulation, Department of Biochemistry, School of Medicine, Gachon University, Incheon, South Korea
| |
Collapse
|
10
|
Alwithenani AI, Althubiti MA. Systematic Analysis of Spleen Tyrosine Kinase Expression and its Clinical Outcomes in Various Cancers. SAUDI JOURNAL OF MEDICINE & MEDICAL SCIENCES 2020; 8:95-104. [PMID: 32587490 PMCID: PMC7305679 DOI: 10.4103/sjmms.sjmms_300_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/28/2019] [Accepted: 02/06/2020] [Indexed: 12/04/2022]
Abstract
Background: Spleen tyrosine kinase (SYK) is an important enzyme in the proliferation and differentiation of all hematopoietic tissues. Its role as a cancer driver is well documented in liquid tumors; however, cumulative evidence has suggested an opposite role in other tumor types. Objectives: To systematically assess the expression of SYK, its prognostic value and epigenetic status in different cancers using bioinformatics tools. Methods: In this bioinformatics study, Oncomine database and cBioPortal were used to study the SYK gene expression, Kaplan–Meier plotter to study its prognostic value and MethHC to assess the SYK gene methylation in various cancers. Results: From 542 unique analyses of the SYK gene, it was found to be overexpressed in bladder, breast and colon cancers but downregulated in leukemia, lymphoma and myeloma. Compared with normal tissues, breast and brain tumors showed an overexpression of the SYK gene, whereas lymphoma and leukemia had lower expression. The Kaplan–Meier survival analysis revealed that SYK expression in pancreatic, gastric, liver and lung patients were correlated with better overall survival. Using cBioPortal, prostate cancer was found to have the highest SYK gene mutation frequency, and the mean expression was highest in diffuse large B-cell lymphoma, acute myeloid leukemia and thymoma. Using the MethHC database, SYK promoter hypermethylation was found to be significantly higher in breast, renal, liver, lung, pancreatic, prostatic, skin and stomach cancers compared with the normal tissue (P < 0.005). Conclusion: The results of this study indicate the potential use of SYK as a diagnostic and therapeutic target for different type of cancers. However, further experimental data are required to validate these results before use of SYK in clinical settings.
Collapse
Affiliation(s)
- Akram I Alwithenani
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mohammad A Althubiti
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
11
|
Inhibition of the deubiquitinase USP10 induces degradation of SYK. Br J Cancer 2020; 122:1175-1184. [PMID: 32015510 PMCID: PMC7156412 DOI: 10.1038/s41416-020-0731-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 10/29/2019] [Indexed: 12/13/2022] Open
Abstract
Background There is growing evidence that spleen tyrosine kinase (SYK) is critical for acute myeloid leukaemia (AML) transformation and maintenance of the leukemic clone in AML patients. It has also been found to be over-expressed in AML patients, with activating mutations in foetal liver tyrosine kinase 3 (FLT3), particularly those with internal tandem duplications (FLT3-ITD), where it transactivates FLT3-ITD and confers resistance to treatment with FLT3 tyrosine kinase inhibitors (TKIs). Methods We have previously described a pharmacological approach to treating FLT3-ITD-positive AML that relies on proteasome-mediated FLT3 degradation via inhibition of USP10, the deubiquitinating enzyme (DUB) responsible for cleaving ubiquitin from FLT3. Results Here, we show that USP10 is also a major DUB required for stabilisation of SYK. We further demonstrate that degradation of SYK can be induced by USP10-targeting inhibitors. USP10 inhibition leads to death of cells driven by active SYK or oncogenic FLT3 and potentiates the anti-leukemic effects of FLT3 inhibition in these cells. Conclusions We suggest that USP10 inhibition is a novel approach to inhibiting SYK and impeding its role in the pathology of AML, including oncogenic FLT3-positive AML. Also, given the significant transforming role SYK in other tumours, targeting USP10 may have broader applications in cancer.
Collapse
|
12
|
SYK Targeting Represents a Potential Therapeutic Option for Relapsed Resistant Pediatric ETV6-RUNX1 B-Acute Lymphoblastic Leukemia Patients. Int J Mol Sci 2019; 20:ijms20246175. [PMID: 31817853 PMCID: PMC6940898 DOI: 10.3390/ijms20246175] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 11/26/2019] [Accepted: 12/04/2019] [Indexed: 12/23/2022] Open
Abstract
The presence of the chromosomal rearrangement t(12;21)(ETV6-RUNX1) in childhood B-acute lymphoblastic leukemia (B-ALL) is an independent predictor of favorable prognosis, however relapses still occur many years later after stopping therapy, and patients often display resistance to current treatments. Since spleen tyrosine kinase (SYK), a cytosolic nonreceptor tyrosine kinase interacting with immune receptors, has been previously associated with malignant transformation and cancer cell proliferation, we aimed to assess its role in ETV6-RUNX1 cell survival and prognosis. We evaluated the effects on cell survival of three SYK inhibitors and showed that all of them, in particular entospletinib, are able to induce cell death and enhance the efficacy of conventional chemotherapeutics. By using reverse phase protein arrays we next revealed that activated SYK is upregulated at diagnosis in pediatric ETV6-RUNX1 patients who will experience relapse, and, importantly, hyperactivation is maintained at a high level also at relapse occurrence. We thus treated primary cells from patients both at diagnosis and relapse with the combination entospletinib + chemotherapeutics and observed that SYK inhibition is able to sensitize resistant primary cells to conventional drugs. Entospletinib could thus represent a new therapeutic option supporting conventional chemotherapy for relapsed ETV6-RUNX1 patients, and these evidences encourage further studies on SYK for treatment of other relapsed resistant acute lymphoblastic leukemia (ALL) subgroups.
Collapse
|
13
|
Qu C, Zheng D, Li S, Liu Y, Lidofsky A, Holmes JA, Chen J, He L, Wei L, Liao Y, Yuan H, Jin Q, Lin Z, Hu Q, Jiang Y, Tu M, Chen X, Li W, Lin W, Fuchs BC, Chung RT, Hong A. Tyrosine kinase SYK is a potential therapeutic target for liver fibrosis. Hepatology 2018; 68. [PMID: 29537660 PMCID: PMC6138581 DOI: 10.1002/hep.29881] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Spleen tyrosine kinase (SYK) plays a critical role in immune cell signaling pathways and has been reported as a biomarker for human hepatocellular carcinoma (HCC). We sought to investigate the mechanism by which SYK promotes liver fibrosis and to evaluate SYK as a therapeutic target for liver fibrosis. We evaluated the cellular localization of SYK and the association between SYK expression and liver fibrogenesis in normal, hepatitis B virus (HBV)-infected, hepatitis C virus (HCV)-infected and non-alcoholic steatohepatitis (NASH) liver tissue (n=36, 127, 22 and 30, respectively). A polymerase chain reaction (PCR) array was used to detect the changes in transcription factor (TF) expression in hepatic stellate cells (HSCs) with SYK knockdown. The effects of SYK antagonism on liver fibrogenesis were studied in LX-2 cells, TWNT-4 cells, primary human HSCs, and three progressive fibrosis/cirrhosis animal models, including a CCL4 mouse model, and diethylnitrosamine (DEN) and bile duct ligation (BDL) rat models. We found that SYK protein in HSCs and hepatocytes correlated positively with liver fibrosis stage in human liver tissue. HBV or HCV infection significantly increased SYK and cytokine expression in hepatocytes. Increasing cytokine production further induced SYK expression and fibrosis-related gene transcription in HSCs. Up-regulated SYK in HSCs promoted HSC activation by increasing the expression of specific TFs related to activation of HSCs. SYK antagonism effectively suppressed liver fibrosis via inhibition of HSC activation, and decreased obstructive jaundice and reduced HCC development in animal models. Conclusion: SYK promotes liver fibrosis via activation of HSCs and is an attractive potential therapeutic target for liver fibrosis and prevention of HCC development. (Hepatology 2018).
Collapse
Affiliation(s)
- Chen Qu
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China,Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Dandan Zheng
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China,Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Sai Li
- Department of Pharmacy, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Yingjun Liu
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, Henan 450008, China
| | - Anna Lidofsky
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jacinta A. Holmes
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jianning Chen
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Lu He
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China,Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Lan Wei
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA
| | - Yadi Liao
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China,Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Hui Yuan
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China,Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Qimeng Jin
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China,Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Zelong Lin
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China,Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Qiaoting Hu
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China,Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Yuchuan Jiang
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China,Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Mengxian Tu
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China,Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Xijun Chen
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China,Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Weiming Li
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China,Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Wenyu Lin
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Bryan C. Fuchs
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA
| | - Raymond T. Chung
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - andJian Hong
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510315, China,Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, China,Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Corresponding author. Contact Information. Dr. Jian Hong, Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510135, China. Phone & Fax: (+86 20) 6165 0514;
| |
Collapse
|
14
|
Lee SJ, Choi JS, Bong SM, Hwang HJ, Lee J, Song HJ, Lee J, Kim JH, Koh JS, Lee BI. Crystal Structures of Spleen Tyrosine Kinase in Complex with Two Novel 4-Aminopyrido[4,3-d] Pyrimidine Derivative Inhibitors. Mol Cells 2018; 41:545-552. [PMID: 29890824 PMCID: PMC6030240 DOI: 10.14348/molcells.2018.2219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 03/01/2018] [Accepted: 04/06/2018] [Indexed: 11/27/2022] Open
Abstract
Spleen tyrosine kinase (SYK) is a cytosolic non-receptor protein tyrosine kinase. Because SYK mediates key receptor signaling pathways involving the B cell receptor and Fc receptors, SYK is an attractive target for autoimmune disease and cancer treatments. To date, representative oral SYK inhibitors, including fostamatinib (R406 or R788), entospletinib (GS-9973), cerdulatinib (PRT062070), and TAK-659, have been assessed in clinical trials. Here, we report the crystal structures of SYK in complex with two newly developed inhibitors possessing 4-aminopyrido[4,3-D]pyrimidine moieties (SKI-G-618 and SKI-O-85). One SYK inhibitor (SKI-G-618) exhibited moderate inhibitory activity against SYK, whereas the other inhibitor (SKI-O-85) exhibited a low inhibitory profile against SYK. Binding mode analysis indicates that a highly potent SYK inhibitor might be developed by modifying and optimizing the functional groups that interact with Leu377, Gly378, and Val385 in the G-loop and the nearby region in SYK. In agreement with our structural analysis, one of our SYK inhibitor (SKI-G-618) shows strong inhibitory activities on the β-hexosaminidase release and phosphorylation of SYK/Vav in RBL-2H3 cells. Taken together, our findings have important implications for the design of high affinity SYK inhibitors.
Collapse
Affiliation(s)
- Sang Jae Lee
- Research Institute, National Cancer Center, Goyang, Gyeonggi 10408,
Korea
- The Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826,
Korea
| | | | - Seoung Min Bong
- Research Institute, National Cancer Center, Goyang, Gyeonggi 10408,
Korea
| | | | | | - Ho-Juhn Song
- Genosco, 767C Concord Ave, 2nd Floor, Cambridge, MA 02138,
USA
| | - Jaekyoo Lee
- Genosco, 767C Concord Ave, 2nd Floor, Cambridge, MA 02138,
USA
| | | | - Jong Sung Koh
- Genosco, 767C Concord Ave, 2nd Floor, Cambridge, MA 02138,
USA
| | - Byung Il Lee
- Research Institute, National Cancer Center, Goyang, Gyeonggi 10408,
Korea
| |
Collapse
|
15
|
McKinnon T, Venier R, Yohe M, Sindiri S, Gryder BE, Shern JF, Kabaroff L, Dickson B, Schleicher K, Chouinard-Pelletier G, Menezes S, Gupta A, Zhang X, Guha R, Ferrer M, Thomas CJ, Wei Y, Davani D, Guidos CJ, Khan J, Gladdy RA. Functional screening of FGFR4-driven tumorigenesis identifies PI3K/mTOR inhibition as a therapeutic strategy in rhabdomyosarcoma. Oncogene 2018; 37:2630-2644. [PMID: 29487419 PMCID: PMC8054765 DOI: 10.1038/s41388-017-0122-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/03/2017] [Accepted: 12/05/2017] [Indexed: 11/08/2022]
Abstract
Rhabdomyosarcoma (RMS) is the most common pediatric soft tissue sarcoma and outcomes have stagnated, highlighting a need for novel therapies. Genomic analysis of RMS has revealed that alterations in the receptor tyrosine kinase (RTK)/RAS/PI3K axis are common and that FGFR4 is frequently mutated or overexpressed. Although FGFR4 is a potentially druggable receptor tyrosine kinase, its functions in RMS are undefined. This study tested FGFR4-activating mutations and overexpression for the ability to generate RMS in mice. Murine tumor models were subsequently used to discover potential therapeutic targets and to test a dual PI3K/mTOR inhibitor in a preclinical setting. Specifically, we provide the first mechanistic evidence of differential potency in the most common human RMS mutations, V550E or N535K, compared to FGFR4wt overexpression as murine myoblasts expressing FGFR4V550E undergo higher rates of cellular transformation, engraftment into mice, and rapidly form sarcomas that highly resemble human RMS. Murine tumor cells overexpressing FGFR4V550E were tested in an in vitro dose-response drug screen along with human RMS cell lines. Compounds were grouped by target class, and potency was determined using average percentage of area under the dose-response curve (AUC). RMS cells were highly sensitive to PI3K/mTOR inhibitors, in particular, GSK2126458 (omipalisib) was a potent inhibitor of FGFR4V550E tumor-derived cell and human RMS cell viability. FGFR4V550E-overexpressing myoblasts and tumor cells had low nanomolar GSK2126458 EC50 values. Mass cytometry using mouse and human RMS cell lines validated GSK2126458 specificity at single-cell resolution, decreasing the abundance of phosphorylated Akt as well as decreasing phosphorylation of the downstream mTOR effectors 4ebp1, Eif4e, and S6. Moreover, PI3K/mTOR inhibition also robustly decreased the growth of RMS tumors in vivo. Thus, by developing a preclinical platform for testing novel therapies, we identified PI3K/mTOR inhibition as a promising new therapy for this devastating pediatric cancer.
Collapse
Affiliation(s)
- Timothy McKinnon
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Rosemarie Venier
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Marielle Yohe
- Genetics Branch, Oncogenomics Section, Center for Cancer Research, National Cancer Institute, Gaithersburg, MD, USA
| | - Sivasish Sindiri
- Genetics Branch, Oncogenomics Section, Center for Cancer Research, National Cancer Institute, Gaithersburg, MD, USA
| | - Berkley E Gryder
- Genetics Branch, Oncogenomics Section, Center for Cancer Research, National Cancer Institute, Gaithersburg, MD, USA
| | - Jack F Shern
- Genetics Branch, Oncogenomics Section, Center for Cancer Research, National Cancer Institute, Gaithersburg, MD, USA
| | - Leah Kabaroff
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Brendan Dickson
- Department of Pathology, Mount Sinai Hospital, Toronto, ON, Canada
| | - Krista Schleicher
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | | | - Serena Menezes
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Abha Gupta
- Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Xiaohu Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Rajarashi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Marc Ferrer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yuhong Wei
- Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Dariush Davani
- Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Cynthia J Guidos
- Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Javed Khan
- Genetics Branch, Oncogenomics Section, Center for Cancer Research, National Cancer Institute, Gaithersburg, MD, USA
| | - Rebecca A Gladdy
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.
- Ontario Institute for Cancer Research, Toronto, ON, Canada.
- Department of Surgery, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
16
|
Barbhuiya MA, Kashyap MK, Puttamallesh VN, Kumar RV, Wu X, Pandey A, Gowda H. Identification of spleen tyrosine kinase as a potential therapeutic target for esophageal squamous cell carcinoma using reverse phase protein arrays. Oncotarget 2018; 9:18422-18434. [PMID: 29719615 PMCID: PMC5915082 DOI: 10.18632/oncotarget.24853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 03/06/2018] [Indexed: 02/07/2023] Open
Abstract
The vast majority of esophageal cancers in China, India and Iran are esophageal squamous cell carcinomas (ESCC). A timely diagnosis provides surgical removal as the main therapeutic option for patients with ESCC. Currently, there are no targeted therapies available for ESCC. We carried out reverse phase protein array-based protein expression profiling of seven ESCC-derivedcell lines and a non-neoplastic esophageal epithelial cell line (Het-1A) to identify differentially expressed proteins in ESCC. SYK non-receptortyrosine kinase was overexpressed in six out of seven ESCC cell lines that were used in the study. We evaluated the role of SYK in ESCC using the pharmacological inhibitor entospletinib (GS-9973) and siRNA-based knock down studies. Entospletinib is a selective inhibitor of SYK, which is currently being evaluated in phase II clinical trials for hematological malignancies. Using in vivo subcutaneous tumor xenografts in mice, we demonstrate that treatment with entospletinib significantly inhibits tumor growth. Further clinical studies are needed to prove the efficacy of entospletinib as a targeted therapeutic agent for treating ESCC.
Collapse
Affiliation(s)
- Mustafa A. Barbhuiya
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Centre, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Manoj K. Kashyap
- School of Life and Allied Health Sciences, Glocal University, Saharanpur, India
| | - Vinuth N. Puttamallesh
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, India
| | - Rekha Vijay Kumar
- Department of Pathology, Kidwai Memorial Institute of Oncology, Bangalore, India
| | - Xinyan Wu
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Akhilesh Pandey
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Centre, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Harsha Gowda
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, India
| |
Collapse
|
17
|
Barbhuiya MA, Kashyap MK, Puttamallesh VN, Kumar RV, Wu X, Pandey A, Gowda H. Identification of spleen tyrosine kinase as a potential therapeutic target for esophageal squamous cell carcinoma using reverse phase protein arrays. Oncotarget 2018. [PMID: 29719615 DOI: 10.18632/oncotarget.24853,] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The vast majority of esophageal cancers in China, India and Iran are esophageal squamous cell carcinomas (ESCC). A timely diagnosis provides surgical removal as the main therapeutic option for patients with ESCC. Currently, there are no targeted therapies available for ESCC. We carried out reverse phase protein array-based protein expression profiling of seven ESCC-derivedcell lines and a non-neoplastic esophageal epithelial cell line (Het-1A) to identify differentially expressed proteins in ESCC. SYK non-receptortyrosine kinase was overexpressed in six out of seven ESCC cell lines that were used in the study. We evaluated the role of SYK in ESCC using the pharmacological inhibitor entospletinib (GS-9973) and siRNA-based knock down studies. Entospletinib is a selective inhibitor of SYK, which is currently being evaluated in phase II clinical trials for hematological malignancies. Using in vivo subcutaneous tumor xenografts in mice, we demonstrate that treatment with entospletinib significantly inhibits tumor growth. Further clinical studies are needed to prove the efficacy of entospletinib as a targeted therapeutic agent for treating ESCC.
Collapse
Affiliation(s)
- Mustafa A Barbhuiya
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Centre, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Manoj K Kashyap
- School of Life and Allied Health Sciences, Glocal University, Saharanpur, India
| | - Vinuth N Puttamallesh
- Institute of Bioinformatics, International Technology Park, Bangalore, India.,Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, India
| | - Rekha Vijay Kumar
- Department of Pathology, Kidwai Memorial Institute of Oncology, Bangalore, India
| | - Xinyan Wu
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Akhilesh Pandey
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Centre, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Institute of Bioinformatics, International Technology Park, Bangalore, India.,Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Harsha Gowda
- Institute of Bioinformatics, International Technology Park, Bangalore, India.,Manipal Academy of Higher Education (MAHE), Manipal, India
| |
Collapse
|
18
|
Liu D, Mamorska-Dyga A. Syk inhibitors in clinical development for hematological malignancies. J Hematol Oncol 2017; 10:145. [PMID: 28754125 PMCID: PMC5534090 DOI: 10.1186/s13045-017-0512-1] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Accepted: 07/20/2017] [Indexed: 01/03/2023] Open
Abstract
Spleen tyrosine kinase (Syk) is a cytosolic non-receptor protein tyrosine kinase (PTK) and is mainly expressed in hematopoietic cells. Syk was recognized as a critical element in the B-cell receptor signaling pathway. Syk is also a key component in signal transduction from other immune receptors like Fc receptors and adhesion receptors. Several oral Syk inhibitors including fostamatinib (R788), entospletinib (GS-9973), cerdulatinib (PRT062070), and TAK-659 are being assessed in clinical trials. The second generation compound, entospletinib, showed promising results in clinical trials against B-cell malignancies, mainly chronic lymphoid leukemia. Syk inhibitors are being evaluated in combination regimens in multiple malignancies.
Collapse
Affiliation(s)
- Delong Liu
- Department of Oncology, The first Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Aleksandra Mamorska-Dyga
- Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY, 10595, USA
| |
Collapse
|
19
|
Matsumaru T, Inai M, Ishigami K, Iwamatsu T, Maita H, Otsuguro S, Nomura T, Matsuda A, Ichikawa S, Sakaitani M, Shuto S, Maenaka K, Kan T. Divergent synthesis of kinase inhibitor derivatives, leading to discovery of selective Gck inhibitors. Bioorg Med Chem Lett 2017; 27:2144-2147. [DOI: 10.1016/j.bmcl.2017.03.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/11/2017] [Accepted: 03/22/2017] [Indexed: 12/01/2022]
|
20
|
Erasmus MF, Matlawska-Wasowska K, Kinjyo I, Mahajan A, Winter SS, Xu L, Horowitz M, Lidke DS, Wilson BS. Dynamic pre-BCR homodimers fine-tune autonomous survival signals in B cell precursor acute lymphoblastic leukemia. Sci Signal 2016; 9:ra116. [PMID: 27899526 DOI: 10.1126/scisignal.aaf3949] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The pre-B cell receptor (pre-BCR) is an immature form of the BCR critical for early B lymphocyte development. It is composed of the membrane-bound immunoglobulin (Ig) heavy chain, surrogate light chain components, and the signaling subunits Igα and Igβ. We developed monovalent quantum dot (QD)-labeled probes specific for Igβ to study the behavior of pre-BCRs engaged in autonomous, ligand-independent signaling in live B cells. Single-particle tracking revealed that QD-labeled pre-BCRs engaged in transient, but frequent, homotypic interactions. Receptor motion was correlated at short separation distances, consistent with the formation of dimers and higher-order oligomers. Repeated encounters between diffusing pre-BCRs appeared to reflect transient co-confinement in plasma membrane domains. In human B cell precursor acute lymphoblastic leukemia (BCP-ALL) cells, we showed that frequent, short-lived, homotypic pre-BCR interactions stimulated survival signals, including expression of BCL6, which encodes a transcriptional repressor. These survival signals were blocked by inhibitory monovalent antigen-binding antibody fragments (Fabs) specific for the surrogate light chain components of the pre-BCR or by inhibitors of the tyrosine kinases Lyn and Syk. For comparison, we evaluated pre-BCR aggregation mediated by dimeric galectin-1, which has binding sites for carbohydrate and for the surrogate light chain λ5 component. Galectin-1 binding resulted in the formation of large, highly immobile pre-BCR aggregates, which was partially relieved by the addition of lactose to prevent the cross-linking of galectin-BCR complexes to other glycosylated membrane components. Analysis of the pre-BCR and its signaling partners suggested that they could be potential targets for combination therapy in BCP-ALL.
Collapse
Affiliation(s)
- M Frank Erasmus
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.,UNM Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Ksenia Matlawska-Wasowska
- UNM Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.,Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Ichiko Kinjyo
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.,UNM Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Avanika Mahajan
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.,UNM Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Stuart S Winter
- UNM Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.,Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Li Xu
- Sea Lane Biotechnologies, 2450 Bayshore Parkway, Mountain View, CA 94043, USA
| | - Michael Horowitz
- Sea Lane Biotechnologies, 2450 Bayshore Parkway, Mountain View, CA 94043, USA
| | - Diane S Lidke
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.,UNM Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Bridget S Wilson
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA. .,UNM Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| |
Collapse
|
21
|
Lee SJ, Choi JS, Han BG, Kim HS, Song HJ, Lee J, Nam S, Goh SH, Kim JH, Koh JS, Lee BI. Crystal structures of spleen tyrosine kinase in complex with novel inhibitors: structural insights for design of anticancer drugs. FEBS J 2016; 283:3613-3625. [DOI: 10.1111/febs.13831] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 07/04/2016] [Accepted: 08/08/2016] [Indexed: 11/27/2022]
Affiliation(s)
- Sang Jae Lee
- Research Institute; National Cancer Center; Goyang Gyeonggi Korea
- The Research Institute of Pharmaceutical Sciences; College of Pharmacy; Seoul National University; Korea
| | | | - Byeong-Gu Han
- Research Institute; National Cancer Center; Goyang Gyeonggi Korea
| | - Hyoun Sook Kim
- Research Institute; National Cancer Center; Goyang Gyeonggi Korea
| | | | | | - Seungyoon Nam
- Department of Life Sciences; College of BioNano Technology; Gachon University; Sungnam Korea
- Department of Genome Medicine and Science; Graduate School of Medicine; Gachon University; Incheon Korea
| | - Sung-Ho Goh
- Research Institute; National Cancer Center; Goyang Gyeonggi Korea
| | | | | | - Byung Il Lee
- Research Institute; National Cancer Center; Goyang Gyeonggi Korea
| |
Collapse
|
22
|
Dasgupta N, Thakur BK, Ta A, Dutta P, Das S. Suppression of Spleen Tyrosine Kinase (Syk) by Histone Deacetylation Promotes, Whereas BAY61-3606, a Synthetic Syk Inhibitor Abrogates Colonocyte Apoptosis by ERK Activation. J Cell Biochem 2016; 118:191-203. [PMID: 27293079 DOI: 10.1002/jcb.25625] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 06/10/2016] [Indexed: 01/10/2023]
Abstract
Spleen tyrosine kinase (Syk), a non-receptor tyrosine kinase, regulates tumor progression, either negatively or positively, depending on the tissue lineage. Information about the role of Syk in colorectal cancers (CRC) is limited, and conflicting reports have been published. We studied Syk expression and its role in differentiation and apoptosis of the colonocytes. Here, we reported for the first time that expression of two transcript variants of Syk is suppressed in colonocytes during butyrate-induced differentiation, which mediates apoptosis of HT-29 cells. Despite being a known HDAC inhibitor, butyrate deacetylates histone3/4 around the transcription start site (TSS) of Syk. Histone deacetylation precludes the binding of RNA Polymerase II to the promoter and inhibits transcription. Since butyrate is a colonic metabolite derived from undigested fibers, our study offers a plausible explanation of the underlying mechanisms of the protective role of butyrate as well as the dietary fibers against CRC through the regulation of Syk. We also report that combined use of butyrate and highly specific Syk inhibitor BAY61-3606 does not enhance differentiation and apoptosis of colonocytes. Instead, BAY completely abolishes butyrate-induced differentiation and apoptosis in a Syk- and ERK1/2-dependent manner. While butyrate dephosphorylates ERK1/2 in HT-29 cells, BAY re-phosphorylates it, leading to its activation. This study describes a novel mechanism of butyrate action in CRC and explores the role of Syk in butyrate-induced differentiation and apoptosis. In addition, our study highlights those commercial small molecule inhibitors, although attractive drug candidates should be used with concern because of their frequent off-target effects. J. Cell. Biochem. 118: 191-203, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nirmalya Dasgupta
- Department of Clinical Medicine, National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata 700010, India
| | - Bhupesh Kumar Thakur
- Department of Clinical Medicine, National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata 700010, India
| | - Atri Ta
- Department of Clinical Medicine, National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata 700010, India
| | - Pujarini Dutta
- Department of Clinical Medicine, National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata 700010, India
| | - Santasabuj Das
- Department of Clinical Medicine, National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme XM, Beliaghata, Kolkata 700010, India
| |
Collapse
|
23
|
Köhrer S, Havranek O, Seyfried F, Hurtz C, Coffey GP, Kim E, Hacken ET, Jäger U, Vanura K, O’Brien S, Thomas DA, Kantarjian H, Ghosh D, Wang Z, Zhang M, Ma W, Jumaa H, Debatin KM, Müschen M, Meyer LH, Davis RE, Burger JA. Pre-BCR signaling in precursor B-cell acute lymphoblastic leukemia regulates PI3K/AKT, FOXO1 and MYC, and can be targeted by SYK inhibition. Leukemia 2016; 30:1246-54. [PMID: 26847027 PMCID: PMC5459356 DOI: 10.1038/leu.2016.9] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/20/2015] [Accepted: 12/23/2015] [Indexed: 12/11/2022]
Abstract
Precursor-B-cell receptor (pre-BCR) signaling and spleen tyrosine kinase (SYK) recently were introduced as therapeutic targets for patients with B-cell acute lymphoblastic leukemia (B-ALL), but the importance of this pathway in B-ALL subsets and mechanism of downstream signaling have not fully been elucidated. Here, we provide new detailed insight into the mechanism of pre-BCR signaling in B-ALL. We compared the effects of pharmacological and genetic disruption of pre-BCR signaling in vitro and in mouse models for B-ALL, demonstrating exquisite dependency of pre-BCR(+) B-ALL, but not other B-ALL subsets, on this signaling pathway. We demonstrate that SYK, PI3K/AKT, FOXO1 and MYC are important downstream mediators of pre-BCR signaling in B-ALL. Furthermore, we define a characteristic immune phenotype and gene expression signature of pre-BCR(+) ALL to distinguish them from other B-ALL subsets. These data provide comprehensive new insight into pre-BCR signaling in B-ALL and corroborate pre-BCR signaling and SYK as promising new therapeutic targets in pre-BCR(+) B-ALL.
Collapse
Affiliation(s)
- Stefan Köhrer
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States, 77030
| | - Ondrej Havranek
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States, 77030
| | - Felix Seyfried
- Ulm University Medical Center, Department of Pediatrics and Adolescent Medicine, Ulm, Germany
| | - Christian Hurtz
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143
| | | | - Ekaterina Kim
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States, 77030
| | - Elisa ten Hacken
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States, 77030
| | - Ulrich Jäger
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Katrina Vanura
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Susan O’Brien
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States, 77030
| | - Deborah A. Thomas
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States, 77030
| | - Hagop Kantarjian
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States, 77030
| | - Dipanjan Ghosh
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States, 77030
| | - Zhiqiang Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States, 77030
| | - Min Zhang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States, 77030
| | - Wencai Ma
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States, 77030
| | - Hassan Jumaa
- Ulm University, Department of Immunology, Ulm, Germany
| | - Klaus-Michael Debatin
- Ulm University Medical Center, Department of Pediatrics and Adolescent Medicine, Ulm, Germany
| | - Markus Müschen
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143
| | - Lüder H. Meyer
- Ulm University Medical Center, Department of Pediatrics and Adolescent Medicine, Ulm, Germany
| | - R. Eric Davis
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States, 77030
| | - Jan A. Burger
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States, 77030
| |
Collapse
|
24
|
Bednarski JJ, Pandey R, Schulte E, White LS, Chen BR, Sandoval GJ, Kohyama M, Haldar M, Nickless A, Trott A, Cheng G, Murphy KM, Bassing CH, Payton JE, Sleckman BP. RAG-mediated DNA double-strand breaks activate a cell type-specific checkpoint to inhibit pre-B cell receptor signals. J Exp Med 2016; 213:209-23. [PMID: 26834154 PMCID: PMC4749927 DOI: 10.1084/jem.20151048] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 12/03/2015] [Indexed: 01/17/2023] Open
Abstract
DNA double-strand breaks (DSBs) activate a canonical DNA damage response, including highly conserved cell cycle checkpoint pathways that prevent cells with DSBs from progressing through the cell cycle. In developing B cells, pre-B cell receptor (pre-BCR) signals initiate immunoglobulin light (Igl) chain gene assembly, leading to RAG-mediated DNA DSBs. The pre-BCR also promotes cell cycle entry, which could cause aberrant DSB repair and genome instability in pre-B cells. Here, we show that RAG DSBs inhibit pre-BCR signals through the ATM- and NF-κB2-dependent induction of SPIC, a hematopoietic-specific transcriptional repressor. SPIC inhibits expression of the SYK tyrosine kinase and BLNK adaptor, resulting in suppression of pre-BCR signaling. This regulatory circuit prevents the pre-BCR from inducing additional Igl chain gene rearrangements and driving pre-B cells with RAG DSBs into cycle. We propose that pre-B cells toggle between pre-BCR signals and a RAG DSB-dependent checkpoint to maintain genome stability while iteratively assembling Igl chain genes.
Collapse
Affiliation(s)
- Jeffrey J Bednarski
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110
| | - Ruchi Pandey
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Emily Schulte
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110
| | - Lynn S White
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110
| | - Bo-Ruei Chen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Gabriel J Sandoval
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Masako Kohyama
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Malay Haldar
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Andrew Nickless
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110
| | - Amanda Trott
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110
| | - Genhong Cheng
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Craig H Bassing
- Division of Cancer Pathobiology, Department of Pathology and Laboratory Medicine, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Jacqueline E Payton
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Barry P Sleckman
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
25
|
Chen WC, Yuan JS, Xing Y, Mitchell A, Mbong N, Popescu AC, McLeod J, Gerhard G, Kennedy JA, Bogdanoski G, Lauriault S, Perdu S, Merkulova Y, Minden MD, Hogge DE, Guidos C, Dick JE, Wang JCY. An Integrated Analysis of Heterogeneous Drug Responses in Acute Myeloid Leukemia That Enables the Discovery of Predictive Biomarkers. Cancer Res 2016; 76:1214-24. [PMID: 26833125 DOI: 10.1158/0008-5472.can-15-2743] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/17/2015] [Indexed: 11/16/2022]
Abstract
Many promising new cancer drugs proceed through preclinical testing and early-phase trials only to fail in late-stage clinical testing. Thus, improved models that better predict survival outcomes and enable the development of biomarkers are needed to identify patients most likely to respond to and benefit from therapy. Here, we describe a comprehensive approach in which we incorporated biobanking, xenografting, and multiplexed phospho-flow (PF) cytometric profiling to study drug response and identify predictive biomarkers in acute myeloid leukemia (AML) patients. To test the efficacy of our approach, we evaluated the investigational JAK2 inhibitor fedratinib (FED) in 64 patient samples. FED robustly reduced leukemia in mouse xenograft models in 59% of cases and was also effective in limiting the protumorigenic activity of leukemia stem cells as shown by serial transplantation assays. In parallel, PF profiling identified FED-mediated reduction in phospho-STAT5 (pSTAT5) levels as a predictive biomarker of in vivo drug response with high specificity (92%) and strong positive predictive value (93%). Unexpectedly, another JAK inhibitor, ruxolitinib (RUX), was ineffective in 8 of 10 FED-responsive samples. Notably, this outcome could be predicted by the status of pSTAT5 signaling, which was unaffected by RUX treatment. Consistent with this observed discrepancy, PF analysis revealed that FED exerted its effects through multiple JAK2-independent mechanisms. Collectively, this work establishes an integrated approach for testing novel anticancer agents that captures the inherent variability of response caused by disease heterogeneity and in parallel, facilitates the identification of predictive biomarkers that can help stratify patients into appropriate clinical trials.
Collapse
Affiliation(s)
- Weihsu C Chen
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Julie S Yuan
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Yan Xing
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Amanda Mitchell
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Nathan Mbong
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Andreea C Popescu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Jessica McLeod
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Gitte Gerhard
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - James A Kennedy
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada. Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Goce Bogdanoski
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Stevan Lauriault
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Sofie Perdu
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Yulia Merkulova
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada. Department of Medicine, University of Toronto, Toronto, Ontario, Canada. Division of Medical Oncology and Hematology, University Health Network, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Donna E Hogge
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Cynthia Guidos
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada. Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada. Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Jean C Y Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada. Department of Medicine, University of Toronto, Toronto, Ontario, Canada. Division of Medical Oncology and Hematology, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
26
|
Krisenko MO, Geahlen RL. Calling in SYK: SYK's dual role as a tumor promoter and tumor suppressor in cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:254-63. [PMID: 25447675 DOI: 10.1016/j.bbamcr.2014.10.022] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 10/21/2014] [Accepted: 10/27/2014] [Indexed: 12/18/2022]
Abstract
SYK (spleen tyrosine kinase) is well-characterized in the immune system as an essential enzyme required for signaling through multiple classes of immune recognition receptors. As a modulator of tumorigenesis, SYK has a bit of a schizophrenic reputation, acting in some cells as a tumor promoter and in others as a tumor suppressor. In many hematopoietic malignancies, SYK provides an important survival function and its inhibition or silencing frequently leads to apoptosis. In cancers of non-immune cells, SYK provides a pro-survival signal, but can also suppress tumorigenesis by restricting epithelial-mesenchymal transition, enhancing cell-cell interactions and inhibiting migration.
Collapse
Affiliation(s)
- Mariya O Krisenko
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, United States
| | - Robert L Geahlen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
27
|
A PREVIOUSLY UNKNOWN UNIQUE CHALLENGE FOR INHIBITORS OF SYK ATP-BINDING SITE: ROLE OF SYK AS A CELL CYCLE CHECKPOINT REGULATOR. EBioMedicine 2014; 1:16-28. [PMID: 25506060 PMCID: PMC4259291 DOI: 10.1016/j.ebiom.2014.10.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The identification of SYK as a molecular target in B-lineage leukemia/lymphoma cells prompted the development of SYK inhibitors as a new class of anti-cancer drug candidates. Here we report that induction of the SYK gene expression in human cells causes a significant down-regulation of evolutionarily conserved genes associated with mitosis and cell cycle progression providing unprecedented evidence that SYK is a master regulator of cell cycle regulatory checkpoint genes in human cells. We further show that SYK regulates the G2 checkpoint by physically associating with and inhibiting the dual-specificity phosphatase CDC25C via phosphorylation of its S216 residue. SYK depletion by RNA interference or treatment with the chemical SYK inhibitor prevented nocodazole-treated human cell lines from activating the G2 checkpoint via CDC25C S216-phosphorylation and resulted in polyploidy. Our study provides genetic and biochemical evidence that spleen tyrosine kinase (SYK) has a unique role in the activation of the G2 checkpoint in both non-lymphohematopoietic and B-lineage lymphoid cells. This previously unknown role of SYK as a cell cycle checkpoint regulator represents an unforeseen and significant challenge for inhibitors of SYK ATP binding site. SYK is a cell cycle regulatory kinase that phosphorylates CDC25C at S216 SYK is a master regulator of cell cycle regulatory checkpoint genes in human cells Inhibitors of SYK ATP binding site may increase the risk for secondary cancer
Collapse
|
28
|
Wei MC, Cleary ML. Novel methods and approaches to acute lymphoblastic leukemia drug discovery. Expert Opin Drug Discov 2014; 9:1435-46. [DOI: 10.1517/17460441.2014.956720] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
29
|
Anticancer drugs: Hitting SYK in B-ALL leukaemia. Nat Rev Drug Discov 2014; 13:494. [PMID: 24948121 DOI: 10.1038/nrd4371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|