51
|
Xiang X, Yuan D, Liu Y, Li J, Wen Q, Kong P, Gao L, Zhang C, Gao L, Peng X, Zhang X. PIM1 overexpression in T-cell lymphomas protects tumor cells from apoptosis and confers doxorubicin resistance by upregulating c-myc expression. Acta Biochim Biophys Sin (Shanghai) 2018; 50:800-806. [PMID: 30020405 DOI: 10.1093/abbs/gmy076] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 06/08/2018] [Indexed: 12/17/2022] Open
Abstract
T-cell lymphomas (TCLs) are a malignancy characterized by tumor aggression and resistance to traditional chemotherapy. Disruption of the extrinsic cell death pathway is essential for resistance to chemotherapy. PIM1 serves as a crucial modulator in cancers. However, the role of PIM1 in TCLs remains unclear. In this study, we studied the roles of PIM1 in established T-lymphoma cell lines Jurkat and HUT-78. CCK-8 assay was conducted to evaluate cell survival and flow cytometry was performed to evaluate cell death of TCL cells. siRNAs were used to knockdown the expression of PIM1 and c-myc. qRT-PCR was used to evaluate the mRNA expression levels of c-myc and PIM1. Western blot analysis was used to evaluate the protein expression levels of PIM1, c-myc, STAT3, and phospho-STAT3. Doxorubicin was used to determine the effect of PIM1 on apoptosis. Our results showed that PIM1 expression was markedly enhanced and induced c-myc expression in TCL cells. Doxorubicin inhibited the expressions of c-myc and PIM1, and triggered the extrinsic cell death of TCLs by suppressing the JAK-STAT3 signaling pathway. Moreover, PIM1 silencing via siRNA suppressed c-myc expression, promoted the cell death of TCLs, and increased doxorubicin sensitivity. Conversely, PIM1 overexpression in TCL cells induced c-myc expression, suppressed TCL cell death, and promoted doxorubicin resistance. Collectively, our results demonstrate that PIM1 overexpression in TCLs participates in cancer cell protection from apoptosis and leads to doxorubicin resistance by inducing c-myc expression, indicating that PIM1 may be a promising target in TCL treatment.
Collapse
Affiliation(s)
- Xixi Xiang
- Center of Hematology, the Second Affiliated Hospital of Army Military Medical University, Chongqing, China
| | - Di Yuan
- Department of Educational Technology, Army Medical University, Chongqing, China
| | - Yao Liu
- Center of Hematology, the Second Affiliated Hospital of Army Military Medical University, Chongqing, China
| | - Jiali Li
- Center of Hematology, the Second Affiliated Hospital of Army Military Medical University, Chongqing, China
| | - Qin Wen
- Center of Hematology, the Second Affiliated Hospital of Army Military Medical University, Chongqing, China
| | - Peiyan Kong
- Center of Hematology, the Second Affiliated Hospital of Army Military Medical University, Chongqing, China
| | - Lei Gao
- Center of Hematology, the Second Affiliated Hospital of Army Military Medical University, Chongqing, China
| | - Cheng Zhang
- Center of Hematology, the Second Affiliated Hospital of Army Military Medical University, Chongqing, China
| | - Li Gao
- Center of Hematology, the Second Affiliated Hospital of Army Military Medical University, Chongqing, China
| | - Xiangui Peng
- Center of Hematology, the Second Affiliated Hospital of Army Military Medical University, Chongqing, China
| | - Xi Zhang
- Center of Hematology, the Second Affiliated Hospital of Army Military Medical University, Chongqing, China
| |
Collapse
|
52
|
Chojnacki K, Wińska P, Wielechowska M, Łukowska-Chojnacka E, Tölzer C, Niefind K, Bretner M. Biological properties and structural study of new aminoalkyl derivatives of benzimidazole and benzotriazole, dual inhibitors of CK2 and PIM1 kinases. Bioorg Chem 2018; 80:266-275. [PMID: 29966873 DOI: 10.1016/j.bioorg.2018.06.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/15/2018] [Accepted: 06/18/2018] [Indexed: 11/30/2022]
Abstract
The new aminoalkyl-substituted derivatives of known CK2 inhibitors 4,5,6,7-tetrabromo-1H-benzimidazole (TBBi) and 4,5,6,7-tetrabromo-1H-benzotriazole (TBBt) were synthesized, and their influence on the activity of recombinant human CK2 α, CK2 holoenzyme and PIM1 kinases was evaluated. All derivatives inhibited the activity of studied kinases and the most efficient were aminopropyl-derivatives 8b and 14b. These compounds also exerted inhibition of cancer cell lines - CCRF-CEM (acute lymphoblastoid leukemia), MCF-7 (human breast cancer), and PC-3 (prostate cancer) proliferation and their EC50 is comparable with the value for clinically studied CK2 inhibitor CX-4945. Preliminary structure activity relationship analysis indicated that the spacer length affected antitumor potency, and two to three methylene units were more favorable. The complex of CK2 α1-335/8b was crystallized, both under high-salt conditions and under low-salt conditions giving crystals which diffracted X-rays to about 2.4 Å resolution, what enabled the determination of the corresponding 3D-structures.
Collapse
Affiliation(s)
- K Chojnacki
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - P Wińska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - M Wielechowska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - E Łukowska-Chojnacka
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - C Tölzer
- Department für Chemie, Institut für Biochemie, Universtät zu Köln, Zülpicher Straße 47, D-50674 Köln, Germany
| | - K Niefind
- Department für Chemie, Institut für Biochemie, Universtät zu Köln, Zülpicher Straße 47, D-50674 Köln, Germany
| | - M Bretner
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland.
| |
Collapse
|
53
|
Koblish H, Li YL, Shin N, Hall L, Wang Q, Wang K, Covington M, Marando C, Bowman K, Boer J, Burke K, Wynn R, Margulis A, Reuther GW, Lambert QT, Dostalik Roman V, Zhang K, Feng H, Xue CB, Diamond S, Hollis G, Yeleswaram S, Yao W, Huber R, Vaddi K, Scherle P. Preclinical characterization of INCB053914, a novel pan-PIM kinase inhibitor, alone and in combination with anticancer agents, in models of hematologic malignancies. PLoS One 2018; 13:e0199108. [PMID: 29927999 PMCID: PMC6013247 DOI: 10.1371/journal.pone.0199108] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/31/2018] [Indexed: 12/17/2022] Open
Abstract
The Proviral Integration site of Moloney murine leukemia virus (PIM) serine/threonine protein kinases are overexpressed in many hematologic and solid tumor malignancies and play central roles in intracellular signaling networks important in tumorigenesis, including the Janus kinase-signal transducer and activator of transcription (JAK/STAT) and phosphatidylinositol 3-kinase (PI3K)/AKT pathways. The three PIM kinase isozymes (PIM1, PIM2, and PIM3) share similar downstream substrates with other key oncogenic kinases and have differing but mutually compensatory functions across tumors. This supports the therapeutic potential of pan-PIM kinase inhibitors, especially in combination with other anticancer agents chosen based on their role in overlapping signaling networks. Reported here is a preclinical characterization of INCB053914, a novel, potent, and selective adenosine triphosphate-competitive pan-PIM kinase inhibitor. In vitro, INCB053914 inhibited proliferation and the phosphorylation of downstream substrates in cell lines from multiple hematologic malignancies. Effects were confirmed in primary bone marrow blasts from patients with acute myeloid leukemia treated ex vivo and in blood samples from patients receiving INCB053914 in an ongoing phase 1 dose-escalation study. In vivo, single-agent INCB053914 inhibited Bcl-2-associated death promoter protein phosphorylation and dose-dependently inhibited tumor growth in acute myeloid leukemia and multiple myeloma xenografts. Additive or synergistic inhibition of tumor growth was observed when INCB053914 was combined with selective PI3Kδ inhibition, selective JAK1 or JAK1/2 inhibition, or cytarabine. Based on these data, pan-PIM kinase inhibitors, including INCB053914, may have therapeutic utility in hematologic malignancies when combined with other inhibitors of oncogenic kinases or standard chemotherapeutics.
Collapse
Affiliation(s)
- Holly Koblish
- Incyte Corporation, Wilmington, Delaware, United States of America
- * E-mail:
| | - Yun-long Li
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Niu Shin
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Leslie Hall
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Qian Wang
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Kathy Wang
- Incyte Corporation, Wilmington, Delaware, United States of America
| | | | - Cindy Marando
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Kevin Bowman
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Jason Boer
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Krista Burke
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Richard Wynn
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Alex Margulis
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Gary W. Reuther
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Que T. Lambert
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida, United States of America
| | | | - Ke Zhang
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Hao Feng
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Chu-Biao Xue
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Sharon Diamond
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Greg Hollis
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Swamy Yeleswaram
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Wenqing Yao
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Reid Huber
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Kris Vaddi
- Incyte Corporation, Wilmington, Delaware, United States of America
| | - Peggy Scherle
- Incyte Corporation, Wilmington, Delaware, United States of America
| |
Collapse
|
54
|
|
55
|
de Bock CE, Demeyer S, Degryse S, Verbeke D, Sweron B, Gielen O, Vandepoel R, Vicente C, Vanden Bempt M, Dagklis A, Geerdens E, Bornschein S, Gijsbers R, Soulier J, Meijerink JP, Heinäniemi M, Teppo S, Bouvy-Liivrand M, Lohi O, Radaelli E, Cools J. HOXA9 Cooperates with Activated JAK/STAT Signaling to Drive Leukemia Development. Cancer Discov 2018; 8:616-631. [PMID: 29496663 DOI: 10.1158/2159-8290.cd-17-0583] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 01/26/2018] [Accepted: 02/22/2018] [Indexed: 11/16/2022]
Abstract
Leukemia is caused by the accumulation of multiple genomic lesions in hematopoietic precursor cells. However, how these events cooperate during oncogenic transformation remains poorly understood. We studied the cooperation between activated JAK3/STAT5 signaling and HOXA9 overexpression, two events identified as significantly co-occurring in T-cell acute lymphoblastic leukemia. Expression of mutant JAK3 and HOXA9 led to a rapid development of leukemia originating from multipotent or lymphoid-committed progenitors, with a significant decrease in disease latency compared with JAK3 or HOXA9 alone. Integrated RNA sequencing, chromatin immunoprecipitation sequencing, and Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq) revealed that STAT5 and HOXA9 have co-occupancy across the genome, resulting in enhanced STAT5 transcriptional activity and ectopic activation of FOS/JUN (AP1). Our data suggest that oncogenic transcription factors such as HOXA9 provide a fertile ground for specific signaling pathways to thrive, explaining why JAK/STAT pathway mutations accumulate in HOXA9-expressing cells.Significance: The mechanism of oncogene cooperation in cancer development remains poorly characterized. In this study, we model the cooperation between activated JAK/STAT signaling and ectopic HOXA9 expression during T-cell leukemia development. We identify a direct cooperation between STAT5 and HOXA9 at the transcriptional level and identify PIM1 kinase as a possible drug target in mutant JAK/STAT/HOXA9-positive leukemia cases. Cancer Discov; 8(5); 616-31. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 517.
Collapse
Affiliation(s)
- Charles E de Bock
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Sofie Demeyer
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Sandrine Degryse
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Delphine Verbeke
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Bram Sweron
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Olga Gielen
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Roel Vandepoel
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Carmen Vicente
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Marlies Vanden Bempt
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Antonis Dagklis
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Ellen Geerdens
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Simon Bornschein
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Rik Gijsbers
- Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Jean Soulier
- U944 INSERM and Hematology Laboratory, St-Louis Hospital, APHP, Hematology University Institute, University Paris-Diderot, Paris, France
| | - Jules P Meijerink
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Merja Heinäniemi
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Susanna Teppo
- Tampere Centre for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Maria Bouvy-Liivrand
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Olli Lohi
- Tampere Centre for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Enrico Radaelli
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Jan Cools
- KU Leuven, Center for Human Genetics, Leuven, Belgium. .,VIB, Center for Cancer Biology, Leuven, Belgium
| |
Collapse
|
56
|
Abstract
Pim kinases are being implicated in oncogenic process in various human cancers. Pim kinases primarily deal with three broad categories of functions such as tumorigenesis, protecting cells from apoptotic signals and evading immune attacks. Here in this review, we discuss the regulation of Pim kinases and their expression, and how these kinases defend cancer cells from therapeutic and immune attacks with special emphasis on how Pim kinases maintain their own expression during apoptosis and cellular transformation, defend mitochondria during apoptosis, defend cancer cells from immune attack, defend cancer cells from therapeutic attack, choose localization, self-regulation, activation of oncogenic transcription, metabolic regulation and so on. In addition, we also discuss how Pim kinases contribute to tumorigenesis by regulating cellular transformation and glycolysis to reinforce the importance of Pim kinases in cancer and cancer stem cells.
Collapse
|
57
|
Hasegawa T, Sugita M, Kikuchi T, Hirata F. A Systematic Analysis of the Binding Affinity between the Pim-1 Kinase and Its Inhibitors Based on the MM/3D-RISM/KH Method. J Chem Inf Model 2017; 57:2789-2798. [PMID: 29019402 DOI: 10.1021/acs.jcim.7b00158] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A systematic study of the binding affinities of 16 lead compounds targeting the Pim-1 kinase based on the 3D-RISM/KH theory and MD simulations is reported. The results show a correlation coefficient R = 0.69 between the theoretical and experimental values of the binding free energy. This demonstrates that the method is applicable to the problem of compound screening and lead optimization, for which relative values of the free energy among the compounds have significance. We elucidate the contribution of the ligand fragments to the binding free energy. Our results indicate that the interactions between the residues and the triazolo[4,3-b]pyridazine scaffold as well as the phenyl ring of the ligand molecule make significant contributions to stabilization of the complex. Using the 3D-RISM/KH theory, we further analyze the probability distribution of a ligand fragment around the protein-ligand complex in which the substituent around the phenyl ring is removed from the ligand. The results demonstrate that the 3D-RISM/KH theory is capable of predicting the position of substitution on a ligand that has a higher affinity to a target protein.
Collapse
Affiliation(s)
- Takeshi Hasegawa
- Department of Bioinformatics, College of Life Science, Ritsumeikan University , 1-1-1 Noji-higashi, Kusatsu, Shiga 525-8577, Japan
| | - Masatake Sugita
- Department of Bioinformatics, College of Life Science, Ritsumeikan University , 1-1-1 Noji-higashi, Kusatsu, Shiga 525-8577, Japan
| | - Takeshi Kikuchi
- Department of Bioinformatics, College of Life Science, Ritsumeikan University , 1-1-1 Noji-higashi, Kusatsu, Shiga 525-8577, Japan
| | - Fumio Hirata
- Toyota Physical and Chemical Research Institute , 41-1 Yokomichi, Nagakute, Aichi 480-1192, Japan
| |
Collapse
|
58
|
Huang PS, Lin YH, Chi HC, Chen PY, Huang YH, Yeh CT, Wang CS, Lin KH. Thyroid hormone inhibits growth of hepatoma cells through induction of miR-214. Sci Rep 2017; 7:14868. [PMID: 29093516 PMCID: PMC5665905 DOI: 10.1038/s41598-017-14864-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 10/05/2017] [Indexed: 12/17/2022] Open
Abstract
Thyroid hormone (TH) plays a role in regulating the metabolic rate, heart functions, muscle control and maintenance of bones. 3,3′5-tri-iodo-L-thyronine (T3) displays high affinity to nuclear thyroid hormone receptors (TRs), which mediate most TH actions. Recent studies have shown hypothyroidism in patients with an increased risk of hepatocellular carcinoma (HCC). MicroRNAs (miRNAs), a class of non-protein-coding RNA, are suggested to control tumor growth by interacting with target genes. However, the clinical significance of T3/TR-regulated miRNAs in tumors has yet to be established. In the current study, miRNA expression profile screening was performed using SYBR Green-Based qRT-PCR array in TR-overexpressing HepG2 cells. miR-214-3p, which is expressed at low levels in HCC, was stimulated upon T3 application. The 3′UTR luciferase reporter assay confirmed that the proto-oncogene serine/threonine-protein kinase, PIM-1, is a miR-214-3p target. PIM-1 was decreased upon treatment with miR-214-3p or T3 stimulation. PIM-1 was highly expressed in HCC, and the effect of PIM-1 on cell proliferation might be mediated by the inhibition of p21. Furthermore, the T3-induced suppression of cell proliferation was partially rescued upon miR-214-3p knockdown. Our data demonstrate that T3 induces miR-214-3p expression and suppresses cell proliferation through PIM-1, thus contributing to the inhibition of HCC tumor formation.
Collapse
Affiliation(s)
- Po-Shuan Huang
- Department of Biochemistry, College of Medicine, Chang-Gung University, 333, Taoyuan, Taiwan
| | - Yang-Hsiang Lin
- Department of Biochemistry, College of Medicine, Chang-Gung University, 333, Taoyuan, Taiwan
| | - Hsiang-Cheng Chi
- Radiation Biology Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, 333, Linkou, Taoyuan, Taiwan
| | - Pei-Yu Chen
- Department of Biochemistry, College of Medicine, Chang-Gung University, 333, Taoyuan, Taiwan
| | - Ya-Hui Huang
- Liver Research Center, Chang Gung Memorial Hospital, 333, Linko, Taoyuan, Taiwan
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, 333, Linko, Taoyuan, Taiwan
| | - Chia-Siu Wang
- Department of General Surgery, Chang Gung Memorial Hospital, Chiayi, 613, Taiwan.
| | - Kwang-Huei Lin
- Department of Biochemistry, College of Medicine, Chang-Gung University, 333, Taoyuan, Taiwan. .,Liver Research Center, Chang Gung Memorial Hospital, 333, Linko, Taoyuan, Taiwan. .,Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, 333, Taoyuan, Taiwan.
| |
Collapse
|
59
|
Wang K, Deng X, Shen Z, Jia Y, Ding R, Li R, Liao X, Wang S, Ha Y, Kong Y, Wu Y, Guo J, Jie W. High glucose promotes vascular smooth muscle cell proliferation by upregulating proto-oncogene serine/threonine-protein kinase Pim-1 expression. Oncotarget 2017; 8:88320-88331. [PMID: 29179437 PMCID: PMC5687607 DOI: 10.18632/oncotarget.19368] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 06/28/2017] [Indexed: 02/06/2023] Open
Abstract
Serine/threonine kinase proviral integration site for Moloney murine leukemia virus 1 (Pim-1) plays an essential role in arterial wall cell proliferation and associated vascular diseases, including pulmonary arterial hypertension and aortic wall neointima formation. Here we tested a role of Pim-1 in high-glucose (HG)-mediated vascular smooth muscle cell (VSMC) proliferation. Pim-1 and proliferating cell nuclear antigen (PCNA) expression levels in arterial samples from streptozotocin-induced hyperglycemia rats were increased, compared with their weak expression in normoglycemic groups. In cultured rat VSMCs, HG led to transient Pim-1 expression decline, followed by sustained expression increase at both transcriptional and translational levels. Immunoblot analysis demonstrated that HG increased the expression of the 33-kDa isoform of Pim-1, but at much less extent to its 44-kDa plasma membrane isoform. D-glucose at a concentration of 25 mmol/L showed highest activity in stimulating Pim-1 expression. Both Pim-1 inhibitor quercetagetin and STAT3 inhibitor stattic significantly attenuated HG-induced VSMC proliferation and arrested cell cycle progression at the G1 phase. Quercetagetin showed no effect on Pim-1 expression but decreased the phosphorylated-Bad (T112)/Bad ratio in HG-treated VSMCs. However, stattic decreased phosphorylated-STAT3 (Y705) levels and caused transcriptional and translational down-regulation of Pim-1 in HG-treated VSMCs. Our findings suggest HG-mediated Pim-1 expression contributes to VSMC proliferation, which may be partly due to the activation of STAT3/Pim-1 signaling.
Collapse
Affiliation(s)
- Keke Wang
- Department of Pathology, School of Basic medicine Sciences, Guangdong Medical University, Zhanjiang, P.R. China
| | - Xiaojiang Deng
- Department of Cardiovascular, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Zhihua Shen
- Department of Pathology, School of Basic medicine Sciences, Guangdong Medical University, Zhanjiang, P.R. China
| | - Yanan Jia
- Department of Pathology, School of Basic medicine Sciences, Guangdong Medical University, Zhanjiang, P.R. China
| | - Ranran Ding
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Rujia Li
- Department of Pathology, School of Basic medicine Sciences, Guangdong Medical University, Zhanjiang, P.R. China
| | - Xiaomin Liao
- Department of Pathology, School of Basic medicine Sciences, Guangdong Medical University, Zhanjiang, P.R. China
| | - Sisi Wang
- Department of Pathology, School of Basic medicine Sciences, Guangdong Medical University, Zhanjiang, P.R. China
| | - Yanping Ha
- Department of Pathology, School of Basic medicine Sciences, Guangdong Medical University, Zhanjiang, P.R. China
| | - Yueqiong Kong
- Cardiovascular Institute of 1st Affiliated Hospital & Key Laboratory of Tropical Diseases and Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, P.R. China
| | - Yuyou Wu
- Cardiovascular Institute of 1st Affiliated Hospital & Key Laboratory of Tropical Diseases and Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, P.R. China
| | - Junli Guo
- Cardiovascular Institute of 1st Affiliated Hospital & Key Laboratory of Tropical Diseases and Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, P.R. China
| | - Wei Jie
- Department of Pathology, School of Basic medicine Sciences, Guangdong Medical University, Zhanjiang, P.R. China
| |
Collapse
|
60
|
Abnous K, Manavi H, Mehri S, Alibolandi M, Kamali H, Ghandadi M, Hadizadeh F. In vitro evaluation of dihydropyridine-3-carbonitriles as potential cytotoxic agents through PIM-1 protein kinase inhibition. Res Pharm Sci 2017. [PMID: 28626477 PMCID: PMC5465828 DOI: 10.4103/1735-5362.207200] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
PIM-1 protein kinase inhibitor belongs to a novel class of serine/threonine kinases. As PIM-1 is overexpressed in cancer cells and possesses oncogenic functions, its inhibition provides a new option in cancer therapy. In this study, in vitro inhibitory effects of seven analogues of 1, 2-dihydropyridine-3-carbonitrile derivatives Ia-c, IIa-d on the activity of recombinant PIM-1 were evaluated using dimethylthiazol diphenyltetrazolium bromide (MTT) assay. The PIM-1 protein kinase inhibitory potencies and the cytotoxicity effects of tested compounds were respectively as follows: Ic > IIa > Ia > IIb > Ib > IId > IIc and IIb > IIa > Ia > IIc > Ic > Ib > IId, respectively. The compound Ic with methylthio imidazole substituent at C-3 position and benzodioxole substituent at C-6 position of 2-imino-1, 2-dihydropyridine-3- carbonitrile structure showed the strongest PIM-1 inhibitory effect (IC50 = 111.01 nM), while the compound IIc with methythio imidazole substituent at C-3 position and benzodioxole substituent at C-6 position of 2-oxo-1, 2-dihydropyridine-3- carbonitrile structure exhibited the least inhibition activity (IC50 = 433.71 nM). The docking results showed that all tested compounds localized appropriately in the middle of binding cavity after docking procedure, demonstrating suitable interactions between ligands and protein. This study demonstrated that the PIM-1 inhibitory potencies of newly synthesized compounds were in submicromolar concentrations (IC50 < 150 nM) while they exhibited low cytotoxicity on HT-29 cell line (IC50> 130 μM). Altogether, our data indicated that compounds Ic, IIa, Ia could be considered as new potent non-toxic PIM-1 inhibitors which could be used in combination with routine anti-proliferative drugs.
Collapse
Affiliation(s)
- Khalil Abnous
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, I.R. Iran
| | - Hesam Manavi
- Student Research Committee, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, I.R. Iran
| | - Soghra Mehri
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, I.R. Iran.,Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, I.R. Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, I.R. Iran
| | - Hossein Kamali
- Targeted Drug Delivery Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, I.R. Iran
| | - Morteza Ghandadi
- Biotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, I.R. Iran
| | - Farzin Hadizadeh
- Biotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, I.R. Iran
| |
Collapse
|
61
|
Naguib BH, El-Nassan HB, Abdelghany TM. Synthesis of new pyridothienopyrimidinone derivatives as Pim-1 inhibitors. J Enzyme Inhib Med Chem 2017; 32:457-467. [PMID: 28097906 PMCID: PMC6010143 DOI: 10.1080/14756366.2016.1261130] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Four series of pyridothienopyrimidin-4-one derivatives were designed and prepared to improve the pim-1 inhibitory activity of the previously reported thieno[2,3-b]pyridines. Significant improvement in the pim-1 inhibition and cytotoxic activity was achieved using structure rigidification strategy via ring closure. Six compounds (6c, 7a, 7c, 7d, 8b and 9) showed highly potent pim-1 inhibitory activity with IC50 of 4.62, 1.18, 1.38, 1.97, 8.83 and 4.18 μM, respectively. Four other compounds (6b, 6d, 7b and 8a) showed moderate pim-1 inhibition. The most active compounds were tested for their cytotoxic activity on three cell lines [MCF7, HCT116 and PC3]. Compounds 7a [the 2-(2-chlorophenyl)-2,3-dihydro derivative] and 7d [the 2-(2-(trifluoromethyl)-phenyl)-2,3-dihydro derivative] displayed the most potent cytotoxic effect on the three cell lines tested consistent with their highest estimated pim-1 IC50 values.
Collapse
Affiliation(s)
- Bassem H Naguib
- a Pharmaceutical Chemistry Department, Faculty of Pharmacy , The British University in Egypt , Cairo , Egypt.,b Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy , Cairo University , Cairo , Egypt
| | - Hala B El-Nassan
- b Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy , Cairo University , Cairo , Egypt
| | - Tamer M Abdelghany
- c Department of Pharmacology and Toxicology, Faculty of Pharmacy , Al-Azhar University , Cairo , Egypt
| |
Collapse
|
62
|
Bataille CJR, Brennan MB, Byrne S, Davies SG, Durbin M, Fedorov O, Huber KVM, Jones AM, Knapp S, Liu G, Nadali A, Quevedo CE, Russell AJ, Walker RG, Westwood R, Wynne GM. Thiazolidine derivatives as potent and selective inhibitors of the PIM kinase family. Bioorg Med Chem 2017; 25:2657-2665. [PMID: 28341403 DOI: 10.1016/j.bmc.2017.02.056] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/23/2017] [Accepted: 02/25/2017] [Indexed: 12/31/2022]
Abstract
The PIM family of serine/threonine kinases have become an attractive target for anti-cancer drug development, particularly for certain hematological malignancies. Here, we describe the discovery of a series of inhibitors of the PIM kinase family using a high throughput screening strategy. Through a combination of molecular modeling and optimization studies, the intrinsic potencies and molecular properties of this series of compounds was significantly improved. An excellent pan-PIM isoform inhibition profile was observed across the series, while optimized examples show good selectivity over other kinases. Two PIM-expressing leukemic cancer cell lines, MV4-11 and K562, were employed to evaluate the in vitro anti-proliferative effects of selected inhibitors. Encouraging activities were observed for many examples, with the best example (44) giving an IC50 of 0.75μM against the K562 cell line. These data provide a promising starting point for further development of this series as a new cancer therapy through PIM kinase inhibition.
Collapse
Affiliation(s)
- Carole J R Bataille
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| | - Méabh B Brennan
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| | - Simon Byrne
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| | - Stephen G Davies
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK.
| | - Matthew Durbin
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| | - Oleg Fedorov
- Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, UK
| | - Kilian V M Huber
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| | - Alan M Jones
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| | - Stefan Knapp
- Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, UK
| | - Gu Liu
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| | - Anna Nadali
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Camilo E Quevedo
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| | - Angela J Russell
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK; Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.
| | - Roderick G Walker
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Robert Westwood
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| | - Graham M Wynne
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| |
Collapse
|
63
|
Wang XX, Liu J, Tang YM, Hong L, Zeng Z, Tan GH. MicroRNA-638 inhibits cell proliferation by targeting suppress PIM1 expression in human osteosarcoma. Tumour Biol 2016; 37:16367–16375. [PMID: 28050866 DOI: 10.1007/s13277-016-5379-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 09/09/2016] [Indexed: 12/31/2022] Open
Abstract
MicroRNAs (miRNAs) are a type of small noncoding RNAs that often play important roles in carcinogenesis, but the carcinogenic mechanism of miRNAs is still unclear. This study will investigate the functions and the mechanism of miR-638 in osteosarcoma (OS). The expression of miR-638 in OS and the DNA copy number of miR-638 were detected by real-time PCR. The effect of miR-638 on cell proliferation was measured by CCK8 assay. Different assays, including bioinformatics algorithms, luciferase report assay, and Western blotting, were used to identify the target gene proviral integration site for Moloney murine leukemia virus 1 (PIM1) of miR-638 in OS. The expression of PIM1 in clinical OS tissues was also validated by immunohistochemical assay. From this research, we found that miR-638 was downregulated in OS tissues compared with corresponding noncancerous tissues (NCTs), and the DNA copy number of miR-638 was lower in OS than in NCTs, which may induce the corresponding downregulation of miR-638 in OS. Ectopic expression of miR-638 inhibited OS cell growth in vitro. Subsequently, we identified that PIM1 is the downstream target gene of miR-638 in OS cells, and silencing PIM1 expression phenocopied the inhibitory effect of miR-638 on OS cell proliferation. Furthermore, we observed that PIM1 was overexpressed in OS tissues, and high expression of PIM1 in OS predicted poor overall survival. In summary, we revealed that miR-638 functions as a tumor suppressor through inhibiting PIM1 expression in OS.
Collapse
Affiliation(s)
- Xiao-Xu Wang
- Department of Joint Surgery, the Second Affiliated Hospital, University of South China, 35 Jiefang Road, Hengyang, Hunan, People's Republic of China
| | - Jue Liu
- Department of Dobstertics and Gynecology, the Second Affiliated Hospital, University of South China, Hengyang, Hunan, People's Republic of China
| | - Yi-Min Tang
- Department of Nursing, the First Affiliated Hospital, University of South China, Hengyang, Hunan, People's Republic of China
| | - Liang Hong
- Department of Joint Surgery, the Second Affiliated Hospital, University of South China, 35 Jiefang Road, Hengyang, Hunan, People's Republic of China
| | - Zhi Zeng
- Department of Joint Surgery, the Second Affiliated Hospital, University of South China, 35 Jiefang Road, Hengyang, Hunan, People's Republic of China
| | - Guang-Hua Tan
- Department of Joint Surgery, the Second Affiliated Hospital, University of South China, 35 Jiefang Road, Hengyang, Hunan, People's Republic of China.
| |
Collapse
|
64
|
Jiménez-García MP, Lucena-Cacace A, Robles-Frías MJ, Narlik-Grassow M, Blanco-Aparicio C, Carnero A. The role of PIM1/PIM2 kinases in tumors of the male reproductive system. Sci Rep 2016; 6:38079. [PMID: 27901106 PMCID: PMC5128923 DOI: 10.1038/srep38079] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 11/03/2016] [Indexed: 12/18/2022] Open
Abstract
The PIM family of serine/threonine kinases has three highly conserved isoforms (PIM1, PIM2 and PIM3). PIM proteins are regulated through transcription and stability by JAK/STAT pathways and are overexpressed in hematological malignancies and solid tumors. The PIM kinases possess weak oncogenic abilities, but enhance other genes or chemical carcinogens to induce tumors. We generated conditional transgenic mice that overexpress PIM1 or PIM2 in male reproductive organs and analyzed their contribution to tumorigenesis. We found an increase in alterations of sexual organs and hyperplasia in the transgenic mice correlating with inflammation. We also found that PIM1/2 are overexpressed in a subset of human male germ cells and prostate tumors correlating with inflammatory features and stem cell markers. Our data suggest that PIM1/2 kinase overexpression is a common feature of male reproductive organs tumors, which provoke tissue alterations and a large inflammatory response that may act synergistically during the process of tumorigenesis. There is also a correlation with markers of cancer stem cells, which may contribute to the therapy resistance found in tumors overexpressing PIM kinases.
Collapse
Affiliation(s)
- Manuel Pedro Jiménez-García
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocío/Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n 41013, Seville, Spain
| | - Antonio Lucena-Cacace
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocío/Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n 41013, Seville, Spain
| | - María José Robles-Frías
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocío/Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n 41013, Seville, Spain
| | - Maja Narlik-Grassow
- Experimental Therapeutics Programme, Spanish National Cancer Centre (CNIO), C/Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Carmen Blanco-Aparicio
- Experimental Therapeutics Programme, Spanish National Cancer Centre (CNIO), C/Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocío/Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n 41013, Seville, Spain
| |
Collapse
|
65
|
Wurz RP, Sastri C, D'Amico DC, Herberich B, Jackson CLM, Pettus LH, Tasker AS, Wu B, Guerrero N, Lipford JR, Winston JT, Yang Y, Wang P, Nguyen Y, Andrews KL, Huang X, Lee MR, Mohr C, Zhang JD, Reid DL, Xu Y, Zhou Y, Wang HL. Discovery of imidazopyridazines as potent Pim-1/2 kinase inhibitors. Bioorg Med Chem Lett 2016; 26:5580-5590. [PMID: 27769621 DOI: 10.1016/j.bmcl.2016.09.067] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/23/2016] [Accepted: 09/27/2016] [Indexed: 10/20/2022]
Abstract
High levels of Pim expression have been implicated in several hematopoietic and solid tumor cancers, suggesting that inhibition of Pim signaling could provide patients with therapeutic benefit. Herein, we describe our progress towards this goal using a screening hit (rac-1) as a starting point. Modification of the indazole ring resulted in the discovery of a series of imidazopyridazine-based Pim inhibitors exemplified by compound 22m, which was found to be a subnanomolar inhibitor of the Pim-1 and Pim-2 isoforms (IC50 values of 0.024nM and 0.095nM, respectively) and to potently inhibit the phosphorylation of BAD in a cell line that expresses high levels of all Pim isoforms, KMS-12-BM (IC50=28nM). Profiling of Pim-1 and Pim-2 expression levels in a panel of multiple myeloma cell lines and correlation of these data with the potency of compound 22m in a proliferation assay suggests that Pim-2 inhibition would be advantageous for this indication.
Collapse
Affiliation(s)
- Ryan P Wurz
- Department of Therapeutic Discovery, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA.
| | - Christine Sastri
- Department of Oncology Research, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA.
| | - Derin C D'Amico
- Department of Therapeutic Discovery, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - Brad Herberich
- Department of Therapeutic Discovery, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - Claire L M Jackson
- Department of Therapeutic Discovery, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - Liping H Pettus
- Department of Therapeutic Discovery, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - Andrew S Tasker
- Department of Therapeutic Discovery, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - Bin Wu
- Department of Therapeutic Discovery, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - Nadia Guerrero
- Department of Oncology Research, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - J Russell Lipford
- Department of Oncology Research, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - Jeffrey T Winston
- Department of Oncology Research, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - Yajing Yang
- Department of Oncology Research, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - Paul Wang
- Department of Discovery Technologies, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - Yen Nguyen
- Department of Discovery Attribute Sciences, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - Kristin L Andrews
- Department of Molecular Engineering, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - Xin Huang
- Department of Molecular Engineering, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - Matthew R Lee
- Department of Molecular Engineering, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - Christopher Mohr
- Department of Molecular Engineering, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - J D Zhang
- Department of Molecular Engineering, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - Darren L Reid
- Department of Pre-pivotal Drug Product, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - Yang Xu
- Department of Clinical Pharmacology, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - Yihong Zhou
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| | - Hui-Ling Wang
- Department of Therapeutic Discovery, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
| |
Collapse
|
66
|
Mazzacurati L, Lambert QT, Pradhan A, Griner LN, Huszar D, Reuther GW. The PIM inhibitor AZD1208 synergizes with ruxolitinib to induce apoptosis of ruxolitinib sensitive and resistant JAK2-V617F-driven cells and inhibit colony formation of primary MPN cells. Oncotarget 2016; 6:40141-57. [PMID: 26472029 PMCID: PMC4741885 DOI: 10.18632/oncotarget.5653] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/30/2015] [Indexed: 01/22/2023] Open
Abstract
Classical myeloproliferative neoplasms (MPNs) are hematopoietic stem cell disorders that exhibit excess mature myeloid cells, bone marrow fibrosis, and risk of leukemic transformation. Aberrant JAK2 signaling plays an etiological role in MPN formation. Because neoplastic cells in patients are largely insensitive to current anti-JAK2 therapies, effective therapies remain needed. Members of the PIM family of serine/threonine kinases are induced by JAK/STAT signaling, regulate hematopoietic stem cell growth, protect hematopoietic cells from apoptosis, and exhibit hematopoietic cell transforming properties. We hypothesized that PIM kinases may offer a therapeutic target for MPNs. We treated JAK2-V617F-dependent MPN model cells as well as primary MPN patient cells with the PIM kinase inhibitors SGI-1776 and AZD1208 and the JAK2 inhibitor ruxolitinib. While MPN model cells were rather insensitive to PIM inhibitors, combination of PIM inhibitors with ruxolitinib led to a synergistic effect on MPN cell growth due to enhanced apoptosis. Importantly, PIM inhibitor mono-therapy inhibited, and AZD1208/ruxolitinib combination therapy synergistically suppressed, colony formation of primary MPN cells. Enhanced apoptosis by combination therapy was associated with activation of BAD, inhibition of downstream components of the mTOR pathway, including p70S6K and S6 protein, and activation of 4EBP1. Importantly, PIM inhibitors re-sensitized ruxolitinib-resistant MPN cells to ruxolitinib by inducing apoptosis. Finally, exogenous expression of PIM1 induced ruxolitinib resistance in MPN model cells. These data indicate that PIMs may play a role in MPNs and that combining PIM and JAK2 kinase inhibitors may offer a more efficacious therapeutic approach for MPNs over JAK2 inhibitor mono-therapy.
Collapse
Affiliation(s)
- Lucia Mazzacurati
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Que T Lambert
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Anuradha Pradhan
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Lori N Griner
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Dennis Huszar
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL, USA.,Oncology iMed, AstraZeneca, Waltham, MA, USA
| | - Gary W Reuther
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
67
|
Pettus LH, Andrews KL, Booker SK, Chen J, Cee VJ, Chavez F, Chen Y, Eastwood H, Guerrero N, Herberich B, Hickman D, Lanman BA, Laszlo J, Lee MR, Lipford JR, Mattson B, Mohr C, Nguyen Y, Norman MH, Powers D, Reed AB, Rex K, Sastri C, Tamayo N, Wang P, Winston JT, Wu B, Wu T, Wurz RP, Xu Y, Zhou Y, Tasker AS, Wang HL. Discovery and Optimization of Quinazolinone-pyrrolopyrrolones as Potent and Orally Bioavailable Pan-Pim Kinase Inhibitors. J Med Chem 2016; 59:6407-30. [DOI: 10.1021/acs.jmedchem.6b00610] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Liping H. Pettus
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Kristin L. Andrews
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Shon K. Booker
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Jie Chen
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Victor J. Cee
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Frank Chavez
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Yuping Chen
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Heather Eastwood
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Nadia Guerrero
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Bradley Herberich
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Dean Hickman
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Brian A. Lanman
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Jimmy Laszlo
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Matthew R. Lee
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - J. Russell Lipford
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Bethany Mattson
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Christopher Mohr
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Yen Nguyen
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Mark H. Norman
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - David Powers
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Anthony B. Reed
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Karen Rex
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Christine Sastri
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Nuria Tamayo
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Paul Wang
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Jeffrey T. Winston
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Bin Wu
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Tian Wu
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Ryan P. Wurz
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Yang Xu
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Yihong Zhou
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Andrew S. Tasker
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| | - Hui-Ling Wang
- Department of Therapeutic Discovery—Medicinal
Chemistry, ‡Molecular Structure, §Pharmacokinetics and Drug Metabolism, ∥Oncology Research, ⊥Pharmaceutics, #Discovery Technologies, Amgen Inc., One Amgen
Center Drive, Thousand Oaks, California 91320, United States
| |
Collapse
|
68
|
Synthesis and biological evaluation of quinoline derivatives as potential anti-prostate cancer agents and Pim-1 kinase inhibitors. Bioorg Med Chem 2016; 24:1889-97. [DOI: 10.1016/j.bmc.2016.03.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 03/05/2016] [Accepted: 03/07/2016] [Indexed: 11/17/2022]
|
69
|
Naguib BH, El-Nassan HB. Synthesis of new thieno[2,3-b]pyridine derivatives as pim-1 inhibitors. J Enzyme Inhib Med Chem 2016; 31:1718-25. [PMID: 27541740 DOI: 10.3109/14756366.2016.1158711] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Three series of 5-bromo-thieno[2,3-b]pyridines bearing amide or benzoyl groups at position 2 were prepared as pim-1 inhibitors. All the prepared compounds were tested for their pim-1 enzyme inhibitory activity. Two compounds (3c and 5b) showed moderate pim-1 inhibitory activity with IC50 of 35.7 and 12.71 μM, respectively. Three other compounds (3d, 3g and 6d) showed poor pim-1 inhibition. The most active compounds were tested for their cytotoxic activity on five cell lines [MCF7, HEPG2, HCT116, A549 and PC3]. Compound 3g was the most potent cytotoxic agent on almost all the cell lines tested.
Collapse
Affiliation(s)
- Bassem H Naguib
- a Pharmaceutical Chemistry Department , Faculty of Pharmacy, The British University in Egypt , Cairo , Egypt and.,b Pharmaceutical Organic Chemistry Department , Faculty of Pharmacy, Cairo University , Cairo , Egypt
| | - Hala B El-Nassan
- b Pharmaceutical Organic Chemistry Department , Faculty of Pharmacy, Cairo University , Cairo , Egypt
| |
Collapse
|
70
|
Synthesis of novel S-acyl and S-alkylpyrimidinone derivatives as potential cytotoxic agents. RESEARCH ON CHEMICAL INTERMEDIATES 2016. [DOI: 10.1007/s11164-016-2487-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
71
|
Fan YB, Li K, Huang M, Cao Y, Li Y, Jin SY, Liu WB, Wen JC, Liu D, Zhao LX. Design and synthesis of substituted pyrido[3,2-d]-1,2,3-triazines as potential Pim-1 inhibitors. Bioorg Med Chem Lett 2016; 26:1224-8. [PMID: 26804231 DOI: 10.1016/j.bmcl.2016.01.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 12/24/2015] [Accepted: 01/12/2016] [Indexed: 12/31/2022]
Abstract
A novel series of substituted pyrido[3,2-d]-1,2,3-triazines were designed and synthesized as Pim-1 inhibitors through scaffold hopping. Most of the derivatives showed potent in vitro Pim-1 inhibitory activities and anti-proliferative effects toward prostate cancer cells. Among them, 6b, 6h and 6m showed the best Pim-1 inhibitory activity with IC50 values of 0.69, 0.60 and 0.80 μM, respectively. Furthermore, compounds 6b, 6i, 6j and 6m showed strong inhibitory activity to human prostate cancer LNcap and PC-3 cell lines with IC50 values at low micromolar level. Structure-activity relationship analysis revealed that appropriate substitutions at C-6 positions contributed to the kinase inhibition and antiproliferative effects. Moreover, western blot assay suggested that 6j could decrease the levels of p-BAD and p-4E-BP1 in a dose-dependent manner in PC-3 cells. Docking studies showed that 3-N of the scaffold formed a hydrogen bond with Lys67, aromatic 4-aniline formed a key π-π stack with Phe49. Taken together, this study might provide the first sight for developing the pyrido[3,2-d]-1,2,3-triazine scaffold as novel Pim-1 inhibitors.
Collapse
Affiliation(s)
- Yin-Bo Fan
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Kun Li
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Min Huang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yu Cao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ying Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shu-Yu Jin
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Wen-Bing Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jia-Chen Wen
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Lin-Xiang Zhao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
72
|
Xie Y, Bayakhmetov S. PIM1 kinase as a promise of targeted therapy in prostate cancer stem cells. Mol Clin Oncol 2015; 4:13-17. [PMID: 26835011 DOI: 10.3892/mco.2015.673] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/20/2015] [Indexed: 11/05/2022] Open
Abstract
Since the last decade, the PIM family serine/threonine kinases have become a focus in cancer research. Numerous clinical data supports that overexpression of PIM1 is associated with tumor formation in various tissues. However, little is known regarding the function of PIM1 in cancer stem cells. In cancer cells, PIM1 has essential roles in the regulation of the cell cycle, cell proliferation, cell survival and multiple drug resistance. In stem cells, PIM1 kinase exhibits a significant function in stem cell proliferation, self-renewal and expansion. Thus, PIM1 shows a great promise in cancer therapy by targeting stem cells. Furthermore, it is imperative to investigate Pim-1 targeting in cancer stem cells by applicable inhibitors for improving future outcomes. The present review investigated the potential of PIM1 as a therapy target in prostate cancer stem cells.
Collapse
Affiliation(s)
- Yingqiu Xie
- Department of Biology, Nazarbayev University School of Science and Technology, Astana 010000, Republic of Kazakhstan
| | - Samat Bayakhmetov
- Department of Biology, Nazarbayev University School of Science and Technology, Astana 010000, Republic of Kazakhstan
| |
Collapse
|
73
|
Romero-Canelón I, Phoenix B, Pitto-Barry A, Tran J, Soldevila-Barreda JJ, Kirby N, Green S, Sadler PJ, Barry NP. Arene ruthenium dithiolato–carborane complexes for boron neutron capture therapy (BNCT). J Organomet Chem 2015. [DOI: 10.1016/j.jorganchem.2015.05.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
74
|
Discovery of 3,5-substituted 6-azaindazoles as potent pan-Pim inhibitors. Bioorg Med Chem Lett 2015; 25:5258-64. [DOI: 10.1016/j.bmcl.2015.09.052] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/10/2015] [Accepted: 09/22/2015] [Indexed: 11/19/2022]
|
75
|
Different Persistence of the Cellular Effects Promoted by Protein Kinase CK2 Inhibitors CX-4945 and TDB. BIOMED RESEARCH INTERNATIONAL 2015; 2015:185736. [PMID: 26558259 PMCID: PMC4628960 DOI: 10.1155/2015/185736] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/06/2015] [Indexed: 01/31/2023]
Abstract
We compare the cellular efficacy of two selective and cell permeable inhibitors of the antiapoptotic kinase CK2. One inhibitor, CX-4945, is already in clinical trials as antitumor drug, while the other, TDB, has been recently successfully employed to demonstrate the implication of CK2 in cellular (dis)regulation. We found that, upon treatment of cancer cells with these compounds, the extent of inhibition of endocellular CK2 is initially comparable but becomes significantly different after the inhibitors are removed from the cellular medium: while in CX-4945 treated cells CK2 activity is restored to control level after 24 h, in the case of TDB it is still strongly reduced after 4 days from removal. The biological effects of the two inhibitors have been analyzed by performing clonogenic, spheroid formation, and wound-healing assays: we observed a permanent inhibition of cell survival and migration in TDB-treated cells even after the inhibitor removal, while in the case of CX-4945 only its maintenance for the whole duration of the assay insured a persisting effect. We suggest that the superiority of TDB in maintaining kinase activity inhibited and perpetuating the consequent effects is an added value to be considered when planning new therapies based on CK2 targeting.
Collapse
|
76
|
Rajaratnam R, Martin EK, Dörr M, Harms K, Casini A, Meggers E. Correlation between the Stereochemistry and Bioactivity in Octahedral Rhodium Prolinato Complexes. Inorg Chem 2015; 54:8111-20. [DOI: 10.1021/acs.inorgchem.5b01349] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Rajathees Rajaratnam
- Fachbereich Chemie, Philipps-Universität Marburg, Hans-Meerwein-Strasse
4, 35043 Marburg, Germany
| | - Elisabeth K. Martin
- Fachbereich Chemie, Philipps-Universität Marburg, Hans-Meerwein-Strasse
4, 35043 Marburg, Germany
| | - Markus Dörr
- Fachbereich Chemie, Philipps-Universität Marburg, Hans-Meerwein-Strasse
4, 35043 Marburg, Germany
| | - Klaus Harms
- Fachbereich Chemie, Philipps-Universität Marburg, Hans-Meerwein-Strasse
4, 35043 Marburg, Germany
| | - Angela Casini
- Department of Pharmacokinetics, Toxicology
and Targeting, Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, The Netherlands
- Cardiff School
of Chemistry, University of Cardiff, Park Place, Cardiff CF10 3A, U.K
| | - Eric Meggers
- Fachbereich Chemie, Philipps-Universität Marburg, Hans-Meerwein-Strasse
4, 35043 Marburg, Germany
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, People’s Republic of China
| |
Collapse
|
77
|
Hünten S, Kaller M, Drepper F, Oeljeklaus S, Bonfert T, Erhard F, Dueck A, Eichner N, Friedel CC, Meister G, Zimmer R, Warscheid B, Hermeking H. p53-Regulated Networks of Protein, mRNA, miRNA, and lncRNA Expression Revealed by Integrated Pulsed Stable Isotope Labeling With Amino Acids in Cell Culture (pSILAC) and Next Generation Sequencing (NGS) Analyses. Mol Cell Proteomics 2015; 14:2609-29. [PMID: 26183718 DOI: 10.1074/mcp.m115.050237] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Indexed: 12/20/2022] Open
Abstract
We determined the effect of p53 activation on de novo protein synthesis using quantitative proteomics (pulsed stable isotope labeling with amino acids in cell culture/pSILAC) in the colorectal cancer cell line SW480. This was combined with mRNA and noncoding RNA expression analyses by next generation sequencing (RNA-, miR-Seq). Furthermore, genome-wide DNA binding of p53 was analyzed by chromatin-immunoprecipitation (ChIP-Seq). Thereby, we identified differentially regulated proteins (542 up, 569 down), mRNAs (1258 up, 415 down), miRNAs (111 up, 95 down) and lncRNAs (270 up, 123 down). Changes in protein and mRNA expression levels showed a positive correlation (r = 0.50, p < 0.0001). In total, we detected 133 direct p53 target genes that were differentially expressed and displayed p53 occupancy in the vicinity of their promoter. More transcriptionally induced genes displayed occupied p53 binding sites (4.3% mRNAs, 7.2% miRNAs, 6.3% lncRNAs, 5.9% proteins) than repressed genes (2.4% mRNAs, 3.2% miRNAs, 0.8% lncRNAs, 1.9% proteins), suggesting indirect mechanisms of repression. Around 50% of the down-regulated proteins displayed seed-matching sequences of p53-induced miRNAs in the corresponding 3'-UTRs. Moreover, proteins repressed by p53 significantly overlapped with those previously shown to be repressed by miR-34a. We confirmed up-regulation of the novel direct p53 target genes LINC01021, MDFI, ST14 and miR-486 and showed that ectopic LINC01021 expression inhibits proliferation in SW480 cells. Furthermore, KLF12, HMGB1 and CIT mRNAs were confirmed as direct targets of the p53-induced miR-34a, miR-205 and miR-486-5p, respectively. In line with the loss of p53 function during tumor progression, elevated expression of KLF12, HMGB1 and CIT was detected in advanced stages of cancer. In conclusion, the integration of multiple omics methods allowed the comprehensive identification of direct and indirect effectors of p53 that provide new insights and leads into the mechanisms of p53-mediated tumor suppression.
Collapse
Affiliation(s)
- Sabine Hünten
- From the ‡Experimental and Molecular Pathology, Institute of Pathology, Ludwig-Maximilians-University Munich, Thalkirchner Straβe 36, 80337 Munich, Germany
| | - Markus Kaller
- From the ‡Experimental and Molecular Pathology, Institute of Pathology, Ludwig-Maximilians-University Munich, Thalkirchner Straβe 36, 80337 Munich, Germany
| | - Friedel Drepper
- ‖Department of Biochemistry and Functional Proteomics, Faculty of Biology and BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Silke Oeljeklaus
- ‖Department of Biochemistry and Functional Proteomics, Faculty of Biology and BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Thomas Bonfert
- ‡‡Institute for Informatics, Ludwig-Maximilians-University Munich, 80337 Munich, Germany
| | - Florian Erhard
- ‡‡Institute for Informatics, Ludwig-Maximilians-University Munich, 80337 Munich, Germany
| | - Anne Dueck
- §§Biochemistry Center Regensburg (BZR), Laboratory for RNA Biology, University of Regensburg, 93053 Regensburg, Germany
| | - Norbert Eichner
- §§Biochemistry Center Regensburg (BZR), Laboratory for RNA Biology, University of Regensburg, 93053 Regensburg, Germany
| | - Caroline C Friedel
- ‡‡Institute for Informatics, Ludwig-Maximilians-University Munich, 80337 Munich, Germany
| | - Gunter Meister
- §§Biochemistry Center Regensburg (BZR), Laboratory for RNA Biology, University of Regensburg, 93053 Regensburg, Germany
| | - Ralf Zimmer
- ‡‡Institute for Informatics, Ludwig-Maximilians-University Munich, 80337 Munich, Germany
| | - Bettina Warscheid
- ‖Department of Biochemistry and Functional Proteomics, Faculty of Biology and BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany; **Center for Biological Systems Analysis (ZBSA), University of Freiburg, 79104 Freiburg, Germany
| | - Heiko Hermeking
- From the ‡Experimental and Molecular Pathology, Institute of Pathology, Ludwig-Maximilians-University Munich, Thalkirchner Straβe 36, 80337 Munich, Germany; §German Cancer Consortium (DKTK), D-69120 Heidelberg, Germany; ¶German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany;
| |
Collapse
|
78
|
RNA Seq profiling reveals a novel expression pattern of TGF-β target genes in human blood eosinophils. Immunol Lett 2015; 167:1-10. [PMID: 26112417 DOI: 10.1016/j.imlet.2015.06.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/05/2015] [Accepted: 06/15/2015] [Indexed: 12/16/2022]
Abstract
Despite major advances in our understanding of TGF-β signaling in multiple cell types, little is known about the direct target genes of this pathway in human eosinophils. These cells constitute the major inflammatory component present in the sputum and lung of active asthmatics and their numbers correlate well with disease severity. During the transition from acute to chronic asthma, TGF-β levels rise several fold in the lung which drives fibroblasts to produce extracellular matrix (ECM) and participate in airway and parenchymal remodeling. In this report, we use purified blood eosinophils from healthy donors and analyze baseline and TGF-β responsive genes by RNA Seq, and demonstrate that eosinophils (PBE) express 7981 protein-coding genes of which 178 genes are up-regulated and 199 genes are down-regulated by TGF-β. While 18 target genes have been previously associated with asthma and eosinophilic disorders, the vast majority have been implicated in cell death and survival, differentiation, and cellular function. Ingenuity pathway analysis revealed that 126 canonical pathways are activated by TGF-β including iNOS, TREM1, p53, IL-8 and IL-10 signaling. As TGF-β is an important cytokine for eosinophil function and survival, and pulmonary inflammation and fibrosis, our results represent a significant step toward the identification of novel TGF-β responsive genes and provide a potential therapeutic opportunity by selectively targeting relevant genes and pathways.
Collapse
|
79
|
Wurz RP, Pettus LH, Jackson C, Wu B, Wang HL, Herberich B, Cee V, Lanman BA, Reed AB, Chavez F, Nixey T, Laszlo J, Wang P, Nguyen Y, Sastri C, Guerrero N, Winston J, Lipford JR, Lee MR, Andrews KL, Mohr C, Xu Y, Zhou Y, Reid DL, Tasker AS. The discovery and optimization of aminooxadiazoles as potent Pim kinase inhibitors. Bioorg Med Chem Lett 2015; 25:847-55. [DOI: 10.1016/j.bmcl.2014.12.067] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 12/16/2014] [Accepted: 12/19/2014] [Indexed: 11/26/2022]
|
80
|
Huang SMA, Wang A, Greco R, Li Z, Barberis C, Tabart M, Patel V, Schio L, Hurley R, Chen B, Cheng H, Lengauer C, Pollard J, Watters J, Garcia-Echeverria C, Wiederschain D, Adrian F, Zhang J. Combination of PIM and JAK2 inhibitors synergistically suppresses MPN cell proliferation and overcomes drug resistance. Oncotarget 2015; 5:3362-74. [PMID: 24830942 PMCID: PMC4102815 DOI: 10.18632/oncotarget.1951] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Inhibitors of JAK2 kinase are emerging as an important treatment modality for myeloproliferative neoplasms (MPN). However, similar to other kinase inhibitors, resistance to JAK2 inhibitors may eventually emerge through a variety of mechanisms. Effective drug combination is one way to enhance therapeutic efficacy and combat resistance against JAK2 inhibitors. To identify potential combination partners for JAK2 compounds in MPN cell lines, we performed pooled shRNA screen targeting 5,000 genes in the presence or absence of JAK2 blockade. One of the top hits identified was MYC, an oncogenic transcription factor that is difficult to inhibit directly, but could be targeted by modulation of upstream regulatory elements such as kinases. We demonstrate herein that PIM kinase inhibitors efficiently suppress MYC protein levels in MPN cell lines. Overexpression of MYC restores the viability of PIM inhibitor-treated cells, revealing causal relationship between MYC down-regulation and cell growth inhibition by PIM compounds. Combination of various PIM inhibitors with a JAK2 inhibitor results in significant synergistic growth inhibition of multiple MPN cancer cell lines and induction of apoptosis. Mechanistic studies revealed strong downregulation of phosphorylated forms of S6 and 4EBP1 by JAK2/PIM inhibitor combination treatment. Finally, such combination was effective in eradicating in vitro JAK2 inhibitor-resistant MPN clones, where MYC is consistently up-regulated. These findings demonstrate that simultaneous suppression of JAK2 and PIM kinase activity by small molecule inhibitors is more effective than either agent alone in suppressing MPN cell growth. Our data suggest that JAK2 and PIM combination might warrant further investigation for the treatment of JAK2-driven hematologic malignancies.
Collapse
|
81
|
Kalatova B, Jesenska R, Hlinka D, Dudas M. Tripolar mitosis in human cells and embryos: occurrence, pathophysiology and medical implications. Acta Histochem 2015; 117:111-25. [PMID: 25554607 DOI: 10.1016/j.acthis.2014.11.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 11/26/2014] [Accepted: 11/27/2014] [Indexed: 01/08/2023]
Abstract
Tripolar mitosis is a specific case of cell division driven by typical molecular mechanisms of mitosis, but resulting in three daughter cells instead of the usual count of two. Other variants of multipolar mitosis show even more mitotic poles and are relatively rare. In nature, this phenomenon was frequently observed or suspected in multiple common cancers, infected cells, the placenta, and in early human embryos with impaired pregnancy-yielding potential. Artificial causes include radiation and various toxins. Here we combine several pieces of the most recent evidence for the existence of different types of multipolar mitosis in preimplantation embryos together with a detailed review of the literature. The related molecular and cellular mechanisms are discussed, including the regulation of centriole duplication, mitotic spindle biology, centromere functions, cell cycle checkpoints, mitotic autocorrection mechanisms, and the related complicating factors in healthy and affected cells, including post-mitotic cell-cell fusion often associated with multipolar cell division. Clinical relevance for oncology and embryo selection in assisted reproduction is also briefly discussed in this context.
Collapse
|
82
|
Abstract
Pim oncogenes are highly expressed in many types of hematological and solid cancers. Pim kinases regulate the network of signaling pathways that are critical for tumorigenesis and development, making Pim kinases the attractive drug targets. Currently, two approaches have been employed in designing Pim kinase inhibitors: ATP-mimetics and non-ATP mimetics; but all target the ATP-binding pocket and are ATP-competitive. In this review, we summarize the current progress in understanding the Pim-related structure and biology, and provide insights into the binding modes of some prototypical Pim-1 inhibitors. The challenges as well as opportunities are highlighted for development of Pim kinase inhibitors as potential anticancer agents.
Collapse
|
83
|
Xu J, Zhang T, Wang T, You L, Zhao Y. PIM kinases: an overview in tumors and recent advances in pancreatic cancer. Future Oncol 2014; 10:865-76. [PMID: 24799066 DOI: 10.2217/fon.13.229] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The PIM kinases represent a family of serine/threonine kinases, which is composed of three different members (PIM1, PIM2 and PIM3). Aberrant expression of PIM kinases is observed in variety of tumors, including pancreatic cancer. The PIM kinases play pivotal roles in the regulation of cell cycle, apoptosis, properties of stem cells, metabolism, autophagy, drug resistance and targeted therapy. The roles of PIM kinases in pancreatic cancer include the regulation of proliferation, apoptosis, cell cycle, formation, angiogenesis and prediction prognosis. Blocking the activities of PIM kinases could prevent pancreatic cancer development. PIM kinases may be a novel target for cancer therapy.
Collapse
Affiliation(s)
- Jianwei Xu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | | | | | | | | |
Collapse
|
84
|
Wade MA, Jones D, Wilson L, Stockley J, Coffey K, Robson CN, Gaughan L. The histone demethylase enzyme KDM3A is a key estrogen receptor regulator in breast cancer. Nucleic Acids Res 2014; 43:196-207. [PMID: 25488809 PMCID: PMC4288188 DOI: 10.1093/nar/gku1298] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Endocrine therapy has successfully been used to treat estrogen receptor (ER)-positive breast cancer, but this invariably fails with cancers becoming refractory to treatment. Emerging evidence has suggested that fluctuations in ER co-regulatory protein expression may facilitate resistance to therapy and be involved in breast cancer progression. To date, a small number of enzymes that control methylation status of histones have been identified as co-regulators of ER signalling. We have identified the histone H3 lysine 9 mono- and di-methyl demethylase enzyme KDM3A as a positive regulator of ER activity. Here, we demonstrate that depletion of KDM3A by RNAi abrogates the recruitment of the ER to cis-regulatory elements within target gene promoters, thereby inhibiting estrogen-induced gene expression changes. Global gene expression analysis of KDM3A-depleted cells identified gene clusters associated with cell growth. Consistent with this, we show that knockdown of KDM3A reduces ER-positive cell proliferation and demonstrate that KDM3A is required for growth in a model of endocrine therapy-resistant disease. Crucially, we show that KDM3A catalytic activity is required for both ER-target gene expression and cell growth, demonstrating that developing compounds which target demethylase enzymatic activity may be efficacious in treating both ER-positive and endocrine therapy-resistant disease.
Collapse
Affiliation(s)
- Mark A Wade
- Northen Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Dominic Jones
- Northen Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Laura Wilson
- Northen Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Jacqueline Stockley
- The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Kelly Coffey
- Northen Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Craig N Robson
- Northen Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Luke Gaughan
- Northen Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
85
|
Identification of novel inhibitors of human Chk1 using pharmacophore-based virtual screening and their evaluation as potential anti-cancer agents. J Comput Aided Mol Des 2014; 28:1247-56. [DOI: 10.1007/s10822-014-9800-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 10/03/2014] [Indexed: 12/22/2022]
|
86
|
Liu Y, Zhao B, Mao G, Fang X, Liu Y, Huang Y, Wang N. Epigallocatechin-3-O-gallate, a green tea polyphenol, induces expression of pim-1 kinase via PPARγ in human vascular endothelial cells. Cardiovasc Toxicol 2014; 13:391-5. [PMID: 23990052 DOI: 10.1007/s12012-013-9220-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Pim-1 is a serine/threonine kinase and involved in cell survival and proliferation. Recently, it has been shown that pim-1 signaling pathway plays an important role in cardiovascular protection and differentiation. In this study, we sought to explore the expression of pim-1 in human vascular endothelial cells (ECs) and its regulation by epigallocatechin-3-O-gallate (EGCG), a green tea polyphenol which has anti-oxidant, anti-inflammatory and vascular protective effects. By using quantitative reverse transcriptase PCR (qRT-PCR) and Western blotting, we showed that EGCG dose-dependently increased the expression of pim-1 in cultured umbilical vein endothelial cells. Next, we showed that EGCG activated a luciferase reporter driven by peroxisome proliferators-activated receptor (PPAR)-responsive elements. The induced expression of pim-1 was inhibited in ECs pretreated with GW9662, a specific antagonist of PPARγ. In addition, pim-1 was also up-regulated in endothelial cells treated with rosiglitazone, a specific agonist for PPARγ, or those infected with the adenovirus expressing a constitutively active PPARγ. Collectively, our results provided new evidence that pim-1 can be up-regulated by EGCG via a PPARγ-mediated mechanism and may mediate its vascular protective effects.
Collapse
Affiliation(s)
- Yan Liu
- Institute of Cardiovascular Science, Peking University Health Science Center, 38 Xueyuan Rd., Beijing, 100191, China
| | | | | | | | | | | | | |
Collapse
|
87
|
Lee J, More KN, Yang SA, Hong VS. 3,5-Bis(aminopyrimidinyl)indole Derivatives: Synthesis and Evaluation of Pim Kinase Inhibitory Activities. B KOREAN CHEM SOC 2014. [DOI: 10.5012/bkcs.2014.35.7.2123] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
88
|
Kidd M, Modlin IM, Drozdov I. Gene network-based analysis identifies two potential subtypes of small intestinal neuroendocrine tumors. BMC Genomics 2014; 15:595. [PMID: 25023465 PMCID: PMC4124138 DOI: 10.1186/1471-2164-15-595] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 07/07/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tumor transcriptomes contain information of critical value to understanding the different capacities of a cell at both a physiological and pathological level. In terms of clinical relevance, they provide information regarding the cellular "toolbox" e.g., pathways associated with malignancy and metastasis or drug dependency. Exploration of this resource can therefore be leveraged as a translational tool to better manage and assess neoplastic behavior. The availability of public genome-wide expression datasets, provide an opportunity to reassess neuroendocrine tumors at a more fundamental level. We hypothesized that stringent analysis of expression profiles as well as regulatory networks of the neoplastic cell would provide novel information that facilitates further delineation of the genomic basis of small intestinal neuroendocrine tumors. RESULTS We re-analyzed two publically available small intestinal tumor transcriptomes using stringent quality control parameters and network-based approaches and validated expression of core secretory regulatory elements e.g., CPE, PCSK1, secretogranins, including genes involved in depolarization e.g., SCN3A, as well as transcription factors associated with neurodevelopment (NKX2-2, NeuroD1, INSM1) and glucose homeostasis (APLP1). The candidate metastasis-associated transcription factor, ST18, was highly expressed (>14-fold, p < 0.004). Genes previously associated with neoplasia, CEBPA and SDHD, were decreased in expression (-1.5 - -2, p < 0.02). Genomic interrogation indicated that intestinal tumors may consist of two different subtypes, serotonin-producing neoplasms and serotonin/substance P/tachykinin lesions. QPCR validation in an independent dataset (n = 13 neuroendocrine tumors), confirmed up-regulated expression of 87% of genes (13/15). CONCLUSIONS An integrated cellular transcriptomic analysis of small intestinal neuroendocrine tumors identified that they are regulated at a developmental level, have key activation of hypoxic pathways (a known regulator of malignant stem cell phenotypes) as well as activation of genes involved in apoptosis and proliferation. Further refinement of these analyses by RNAseq studies of large-scale databases will enable definition of individual master regulators and facilitate the development of novel tissue and blood-based tools to better understand diagnose and treat tumors.
Collapse
Affiliation(s)
- Mark Kidd
- Yale University School of Medicine, New Haven, CT 06510, USA.
| | | | | |
Collapse
|
89
|
Abstract
The central role of phosphoinositide 3-kinase (PI3K) activation in tumour cell biology has prompted a sizeable effort to target PI3K and/or downstream kinases such as AKT and mammalian target of rapamycin (mTOR) in cancer. However, emerging clinical data show limited single-agent activity of inhibitors targeting PI3K, AKT or mTOR at tolerated doses. One exception is the response to PI3Kδ inhibitors in chronic lymphocytic leukaemia, where a combination of cell-intrinsic and -extrinsic activities drive efficacy. Here, we review key challenges and opportunities for the clinical development of inhibitors targeting the PI3K-AKT-mTOR pathway. Through a greater focus on patient selection, increased understanding of immune modulation and strategic application of rational combinations, it should be possible to realize the potential of this promising class of targeted anticancer agents.
Collapse
|
90
|
Sun H, Zhao L, Peng S, Huang N. Incorporating replacement free energy of binding-site waters in molecular docking. Proteins 2014; 82:1765-76. [DOI: 10.1002/prot.24530] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 01/17/2014] [Accepted: 01/28/2014] [Indexed: 12/24/2022]
Affiliation(s)
- Hanzi Sun
- College of Life Sciences; Beijing Normal University; Beijing 100875 China
- National Institute of Biological Sciences, Beijing, Zhongguancun Life Science Park; Beijing 102206 China
| | - Lifeng Zhao
- National Institute of Biological Sciences, Beijing, Zhongguancun Life Science Park; Beijing 102206 China
| | - Shiming Peng
- National Institute of Biological Sciences, Beijing, Zhongguancun Life Science Park; Beijing 102206 China
| | - Niu Huang
- College of Life Sciences; Beijing Normal University; Beijing 100875 China
- National Institute of Biological Sciences, Beijing, Zhongguancun Life Science Park; Beijing 102206 China
| |
Collapse
|
91
|
Discovery and identification of PIM-1 kinase inhibitors through a hybrid screening approach. Mol Divers 2014; 18:335-44. [DOI: 10.1007/s11030-014-9504-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 01/13/2014] [Indexed: 10/25/2022]
|
92
|
Wähler K, Kräling K, Steuber H, Meggers E. Non-ATP-Mimetic Organometallic Protein Kinase Inhibitor. ChemistryOpen 2013; 2:180-5. [PMID: 24551564 PMCID: PMC3892196 DOI: 10.1002/open.201300031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Indexed: 11/09/2022] Open
Affiliation(s)
- Kathrin Wähler
- Fachbereich Chemie, Philipps-Universität Marburg, Hans-Meerwein-Straße 35043 Marburg (Germany) E-mail:
| | - Katja Kräling
- Fachbereich Chemie, Philipps-Universität Marburg, Hans-Meerwein-Straße 35043 Marburg (Germany) E-mail:
| | - Holger Steuber
- LOEWE-Zentrum für Synthetische Mikrobiologie, Philipps-Universität Marburg, Hans-Meerwein-Straße 35043 Marburg (Germany)
| | - Eric Meggers
- Fachbereich Chemie, Philipps-Universität Marburg, Hans-Meerwein-Straße 35043 Marburg (Germany) E-mail: ; College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 (P. R. China)
| |
Collapse
|
93
|
Arunesh GM, Shanthi E, Krishna MH, Sooriya Kumar J, Viswanadhan VN. Small molecule inhibitors of PIM1 kinase: July 2009 to February 2013 patent update. Expert Opin Ther Pat 2013; 24:5-17. [PMID: 24131033 DOI: 10.1517/13543776.2014.848196] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION The proviral insertion in murine (PIM) lymphoma proteins for which three isoforms, PIM1, PIM2 and PIM3 have been identified, belonging to the family of serine/threonine kinases has emerged recently as an important therapeutic target for the development of selective inhibitors as the new drugs for treating hematological malignancies and solid tumors. The small molecules developed by academia and the pharmaceutical industry have steadily increased in the last few years. Several drug discovery groups focus on treating disorders, such as cancer mediated by PIM kinase, have provided preclinical evidence suggesting that PIM inhibitor provides anti-apoptotic activity, inhibit cell survival and cell proliferation. AREAS COVERED This article discloses recent reviews on research and advances published in the patent literature and scientific publications from July 2009 to February 2013, highlighting discoveries on PIM1 kinase. EXPERT OPINION Several PIM1 kinase small molecule inhibitors are now at the pre-clinical research stage, development and testing. Though nearly 40 patents emerged in the last 3 years, greater efforts towards additional designs and medicinal chemistry continues for developing clinically efficacious PIM1 inhibitors, due to the significance of the target for cancer and the potential for novel and diverse inhibitors as drug candidates.
Collapse
Affiliation(s)
- Gubbi M Arunesh
- Department of Computational Chemistry and Informatics, Jubilant Biosys Ltd, Industrial Suburb , 96, Industrial Suburb, 2nd Stage, Yeshwanthpur, Bangalore 560 022, Karnataka , India +91 80 6662 8908 ; +91 80 66628333 ;
| | | | | | | | | |
Collapse
|
94
|
Casuscelli F, Ardini E, Avanzi N, Casale E, Cervi G, D'Anello M, Donati D, Faiardi D, Ferguson RD, Fogliatto G, Galvani A, Marsiglio A, Mirizzi DG, Montemartini M, Orrenius C, Papeo G, Piutti C, Salom B, Felder ER. Discovery and optimization of pyrrolo[1,2-a]pyrazinones leads to novel and selective inhibitors of PIM kinases. Bioorg Med Chem 2013; 21:7364-80. [PMID: 24139169 DOI: 10.1016/j.bmc.2013.09.054] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 09/13/2013] [Accepted: 09/20/2013] [Indexed: 11/25/2022]
Abstract
A novel series of PIM inhibitors was derived from a combined effort in natural product-inspired library generation and screening. The novel pyrrolo[1,2-a]pyrazinones initial hits are inhibitors of PIM isoforms with IC50 values in the low micromolar range. The application of a rational optimization strategy, guided by the determination of the crystal structure of the complex in the kinase domain of PIM1 with compound 1, led to the discovery of compound 15a, which is a potent PIM kinases inhibitor exhibiting excellent selectivity against a large panel of kinases, representative of each family. The synthesis, structure-activity relationship studies, and pharmacokinetic data of compounds from this inhibitor class are presented herein. Furthermore, the cellular activities including inhibition of cell growth and modulation of downstream targets are also described.
Collapse
Affiliation(s)
- Francesco Casuscelli
- Oncology, Nerviano Medical Sciences, viale Pasteur 10, 20014 Nerviano (MI), Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Mori M, Tintori C, Christopher RSA, Radi M, Schenone S, Musumeci F, Brullo C, Sanità P, Delle Monache S, Angelucci A, Kissova M, Crespan E, Maga G, Botta M. A combination strategy to inhibit Pim-1: synergism between noncompetitive and ATP-competitive inhibitors. ChemMedChem 2013; 8:484-96. [PMID: 23436791 DOI: 10.1002/cmdc.201200480] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Indexed: 12/30/2022]
Abstract
Pim-1 is a serine/threonine kinase critically involved in the initiation and progression of various types of cancer, especially leukemia, lymphomas and solid tumors such as prostate, pancreas and colon, and is considered a potential drug target against these malignancies. In an effort to discover new potent Pim-1 inhibitors, a previously identified ATP-competitive indolyl-pyrrolone scaffold was expanded to derive structure-activity relationship data. A virtual screening campaign was also performed, which led to the discovery of additional ATP-competitive inhibitors as well as a series of 2-aminothiazole derivatives, which are noncompetitive with respect to both ATP and peptide substrate. This mechanism of action, which resembles allosteric inhibition, has not previously been characterized for Pim-1. Notably, further evaluation of the 2-aminothiazoles indicated a synergistic inhibitory effect in enzymatic assays when tested in combination with ATP-competitive inhibitors. A synergistic effect in the inhibition of cell proliferation by ATP-competitive and ATP-noncompetitive compounds was also observed in prostate cancer cell lines (PC3), where all Pim-1 inhibitors tested in showed synergism with the known anticancer agent, paclitaxel. These results further establish Pim-1 as a target in cancer therapy, and highlight the potential of these agents for use as adjuvant agents in the treatment of cancer diseases in which Pim-1 is associated with chemotherapeutic resistance.
Collapse
Affiliation(s)
- Mattia Mori
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Synthesis of (Z)-2-((1H-indazol-3-yl)methylene)-6-[11C]methoxy-7-(piperazin-1-ylmethyl)benzofuran-3(2H)-one as a new potential PET probe for imaging of the enzyme PIM1. Bioorg Med Chem Lett 2013; 23:4342-6. [DOI: 10.1016/j.bmcl.2013.05.091] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 05/22/2013] [Accepted: 05/28/2013] [Indexed: 12/23/2022]
|
97
|
Lee SJ, Han BG, Cho JW, Choi JS, Lee J, Song HJ, Koh JS, Lee BI. Crystal structure of pim1 kinase in complex with a pyrido[4,3-d]pyrimidine derivative suggests a unique binding mode. PLoS One 2013; 8:e70358. [PMID: 23936194 PMCID: PMC3729456 DOI: 10.1371/journal.pone.0070358] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 06/18/2013] [Indexed: 02/07/2023] Open
Abstract
Human Pim1 kinase is a serine/threonine protein kinase that plays important biological roles in cell survival, apoptosis, proliferation, and differentiation. Moreover, Pim1 is up-regulated in various hematopoietic malignancies and solid tumors. Thus, Pim1 is an attractive target for cancer therapeutics, and there has been growing interest in developing small molecule inhibitors for Pim1. Here, we describe the crystal structure of Pim1 in complex with a newly developed pyrido[4,3-d]pyrimidine-derivative inhibitor (SKI-O-068). Our inhibitor exhibits a half maximum inhibitory concentration (IC50) of 123 (±14) nM and has an unusual binding mode in complex with Pim1 kinase. The interactions between SKI-O-068 and the Pim1 active site pocket residue are different from those of other scaffold inhibitor-bound structures. The binding mode analysis suggests that the SKI-O-068 inhibitor can be improved by introducing functional groups that facilitate direct interaction with Lys67, which aid in the design of an optimized inhibitor.
Collapse
Affiliation(s)
- Sang Jae Lee
- Biomolecular Function Research Branch, Research Institute, National Cancer Center, Goyang, Gyeonggi, Republic of Korea
- The Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Byeong-Gu Han
- Biomolecular Function Research Branch, Research Institute, National Cancer Center, Goyang, Gyeonggi, Republic of Korea
| | - Jea-Won Cho
- Biomolecular Function Research Branch, Research Institute, National Cancer Center, Goyang, Gyeonggi, Republic of Korea
| | | | - Jaekyoo Lee
- Genosco, Cambridge, Massachusetts, United States of America
| | - Ho-Juhn Song
- Genosco, Cambridge, Massachusetts, United States of America
| | - Jong Sung Koh
- Oscotec Inc., Seongnam, Gyeonggi, Republic of Korea
- Genosco, Cambridge, Massachusetts, United States of America
- * E-mail: (JSK); (BIL)
| | - Byung Il Lee
- Biomolecular Function Research Branch, Research Institute, National Cancer Center, Goyang, Gyeonggi, Republic of Korea
- * E-mail: (JSK); (BIL)
| |
Collapse
|
98
|
Barry NPE, Sadler PJ. Exploration of the medical periodic table: towards new targets. Chem Commun (Camb) 2013; 49:5106-5131. [PMID: 23636600 DOI: 10.1039/c3cc41143e] [Citation(s) in RCA: 573] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Metallodrugs offer potential for unique mechanisms of drug action based on the choice of the metal, its oxidation state, the types and number of coordinated ligands and the coordination geometry. We discuss recent progress in identifying new target sites and elucidating the mechanisms of action of anti-cancer, anti-bacterial, anti-viral, anti-parasitic, anti-inflammatory, and anti-neurodegenerative agents, as well as in the design of metal-based diagnostic agents. Progress in identifying and defining target sites has been accelerated recently by advances in proteomics, genomics and metal speciation analysis. Examples of metal compounds and chelating agents (enzyme inhibitors) currently in clinical use, clinical trials or preclinical development are highlighted.
Collapse
Affiliation(s)
- Nicolas P E Barry
- University of Warwick, Department of Chemistry, Gibbet Hill Road, Warwick, UK.
| | | |
Collapse
|
99
|
Wang X, Magnuson S, Pastor R, Fan E, Hu H, Tsui V, Deng W, Murray J, Steffek M, Wallweber H, Moffat J, Drummond J, Chan G, Harstad E, Ebens AJ. Discovery of novel pyrazolo[1,5-a]pyrimidines as potent pan-Pim inhibitors by structure- and property-based drug design. Bioorg Med Chem Lett 2013; 23:3149-53. [DOI: 10.1016/j.bmcl.2013.04.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 04/05/2013] [Accepted: 04/08/2013] [Indexed: 10/27/2022]
|
100
|
Kim W, Youn H, Kwon T, Kang J, Kim E, Son B, Yang HJ, Jung Y, Youn B. PIM1 kinase inhibitors induce radiosensitization in non-small cell lung cancer cells. Pharmacol Res 2013; 70:90-101. [PMID: 23352980 DOI: 10.1016/j.phrs.2013.01.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 01/07/2013] [Accepted: 01/14/2013] [Indexed: 01/28/2023]
Abstract
Radiotherapy plays a critical role in the treatment of non-small cell lung cancer (NSCLC). However, radioresistance is a major barrier against increasing the efficiency of radiotherapy for NSCLC. To understand the mechanisms underlying NSCLC radioresistance, we previously focused on the potential involvement of PIM1, PRAS40, FOXO3a, 14-3-3, and protein phosphatases. Among these proteins, PIM1 functioned as an oncogene and was found to act as a crucial mediator in radioresistant NSCLC cells. Therefore, we investigated the use of PIM1-specific inhibitors as novel therapeutic drugs to regulate radiosensitivity in NSCLC. After structure-based drug selection, SGI-1776, ETP-45299, and tryptanthrin were selected as candidates of PIM1 inhibitors that act as radiosensitizers. With irradiation, these drugs inhibited only PIM1 kinase activity without affecting PIM1 mRNA/protein levels or cellular localization. When PIM1 kinase activity was suppressed by these inhibitors, PRAS40 was not phosphorylated. Consequently, unphosphorylated PRAS40 did not form trimeric complexes with 14-3-3 and FOXO3a, leading to increased nuclear localization of FOXO3a. Nuclear FOXO3a promoted the expression of pro-apoptotic proteins such as Bim and FasL, resulting in a radiosensitizing effect on radioresistant NSCLC cells. Moreover, an in vivo xenograft mouse model confirmed this radiosensitizing effect induced by PIM1 inhibitors. In these model systems, tumor volume was significantly reduced by a combinational treatment with irradiation and PIM1 inhibitors compared to irradiation alone. Taken together, our findings provided evidence that PIM1-specific inhibitors, SGI-1776, ETP-45299, and tryptanthrin, can act as novel radiosensitizers to enhance the efficacy of radiotherapy by inhibiting irradiation-induced signaling pathway associated with radioresistance.
Collapse
Affiliation(s)
- Wanyeon Kim
- Department of Biological Sciences, Pusan National University, Busandaehak-ro 63, Geumjeong-gu, Busan, 609-735, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|