1
|
Alkafaas SS, Elsalahaty MI, Ismail DF, Radwan MA, Elkafas SS, Loutfy SA, Elshazli RM, Baazaoui N, Ahmed AE, Hafez W, Diab M, Sakran M, El-Saadony MT, El-Tarabily KA, Kamal HK, Hessien M. The emerging roles of sphingosine 1-phosphate and SphK1 in cancer resistance: a promising therapeutic target. Cancer Cell Int 2024; 24:89. [PMID: 38419070 PMCID: PMC10903003 DOI: 10.1186/s12935-024-03221-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 01/09/2024] [Indexed: 03/02/2024] Open
Abstract
Cancer chemoresistance is a problematic dilemma that significantly restrains numerous cancer management protocols. It can promote cancer recurrence, spreading of cancer, and finally, mortality. Accordingly, enhancing the responsiveness of cancer cells towards chemotherapies could be a vital approach to overcoming cancer chemoresistance. Tumour cells express a high level of sphingosine kinase-1 (SphK1), which acts as a protooncogenic factor and is responsible for the synthesis of sphingosine-1 phosphate (S1P). S1P is released through a Human ATP-binding cassette (ABC) transporter to interact with other phosphosphingolipids components in the interstitial fluid in the tumor microenvironment (TME), provoking communication, progression, invasion, and tumor metastasis. Also, S1P is associated with several impacts, including anti-apoptotic behavior, metastasis, mesenchymal transition (EMT), angiogenesis, and chemotherapy resistance. Recent reports addressed high levels of S1P in several carcinomas, including ovarian, prostate, colorectal, breast, and HCC. Therefore, targeting the S1P/SphK signaling pathway is an emerging therapeutic approach to efficiently attenuate chemoresistance. In this review, we comprehensively discussed S1P functions, metabolism, transport, and signaling. Also, through a bioinformatic framework, we pointed out the alterations of SphK1 gene expression within different cancers with their impact on patient survival, and we demonstrated the protein-protein network of SphK1, elaborating its sparse roles. Furthermore, we made emphasis on different machineries of cancer resistance and the tight link with S1P. We evaluated all publicly available SphK1 inhibitors and their inhibition activity using molecular docking and how SphK1 inhibitors reduce the production of S1P and might reduce chemoresistance, an approach that might be vital in the course of cancer treatment and prognosis.
Collapse
Affiliation(s)
- Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Mohamed I Elsalahaty
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Doha F Ismail
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mustafa Ali Radwan
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Sara Samy Elkafas
- Production Engineering and Mechanical Design Department, Faculty of Engineering, Menofia University, Menofia, Egypt
- Faculty of Control System and Robotics, ITMO University, Saint-Petersburg, 197101, Russia
| | - Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
- Nanotechnology Research Center, British University, Cairo, Egypt
| | - Rami M Elshazli
- Biochemistry and Molecular Genetics Unit, Department of Basic Sciences, Faculty of Physical Therapy, Horus University-Egypt, New Damietta, 34517, Egypt
| | - Narjes Baazaoui
- Biology Department, College of Sciences and Arts Muhayil Assir, King Khalid University, Abha 61421, Saudi Arabia
| | - Ahmed Ezzat Ahmed
- Biology Department, College of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - Wael Hafez
- NMC Royal Hospital, 16th Street, 35233, Khalifa, Abu Dhabi, United Arab Emirates
- Medical Research Division, Department of Internal Medicine, The National Research Centre, Cairo 11511, Egypt
| | - Mohanad Diab
- Burjeel Hospital Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Mohamed Sakran
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
- Biochemistry Department, Faculty of Science, University of Tabuk, Tabuk 47512, Saudi Arabia
| | - Mohamed T El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| | - Khaled A El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Hani K Kamal
- Anatomy and Histology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohamed Hessien
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
2
|
Reed CW, Rodriguez AL, Kalbfleisch JJ, Seto M, Jenkins MT, Blobaum AL, Chang S, Lindsley CW, Niswender CM. Development and profiling of mGlu 7 NAMs with a range of saturable inhibition of agonist responses in vitro. Bioorg Med Chem Lett 2022; 74:128923. [PMID: 35944850 PMCID: PMC10015594 DOI: 10.1016/j.bmcl.2022.128923] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/29/2022] [Accepted: 08/03/2022] [Indexed: 11/02/2022]
Abstract
We describe here a series of metabotropic glutamate receptor 7 (mGlu7) negative allosteric modulators (NAMs) with a saturable range of activity in inhibiting responses to an orthosteric agonist in two distinct in vitro pharmacological assays. The range of inhibition among compounds in this scaffold provides highly structurally related ligands with differential degrees of receptor blockade that can be used to understand inhibitory efficacy profiles in native tissue or in vivo.
Collapse
Affiliation(s)
- Carson W Reed
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, United States
| | - Alice L Rodriguez
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, United States
| | - Jacob J Kalbfleisch
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, United States
| | - Mabel Seto
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, United States
| | - Matthew T Jenkins
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, United States
| | - Anna L Blobaum
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, United States
| | - Sichen Chang
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, United States
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, United States; Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN 37232, United States; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States; Vanderbilt Department of Chemistry, Vanderbilt University, Nashville, TN 37232, United States
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, United States; Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN 37232, United States; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, United States.
| |
Collapse
|
3
|
Revealing 2-Dimethylhydrazino-2-alkyl alkynyl sphingosine derivatives as Sphingosine Kinase 2 inhibitors: some hints on the structural basis for selective inhibition. Bioorg Chem 2022; 121:105668. [DOI: 10.1016/j.bioorg.2022.105668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 02/02/2022] [Accepted: 02/07/2022] [Indexed: 12/29/2022]
|
4
|
Exploiting activity cliffs for building pharmacophore models and comparison with other pharmacophore generation methods: sphingosine kinase 1 as case study. J Comput Aided Mol Des 2022; 36:39-62. [PMID: 35059939 DOI: 10.1007/s10822-021-00435-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 11/24/2021] [Indexed: 12/20/2022]
|
5
|
Hengst JA, Hegde S, Paulson RF, Yun JK. Development of SKI-349, a dual-targeted inhibitor of sphingosine kinase and microtubule polymerization. Bioorg Med Chem Lett 2020; 30:127453. [PMID: 32736077 DOI: 10.1016/j.bmcl.2020.127453] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 11/28/2022]
Abstract
Our sphingosine kinase inhibitor (SKI) optimization studies originated with the optimization of the SKI-I chemotype by replacement of the substituted benzyl rings with substituted phenyl rings giving rise to the discovery of SKI-178. We have recently reported that SKI-178 is a dual-targeted inhibitor of both sphingosine kinase isoforms (SphK1/2) and a microtubule disrupting agent (MDA). In mechanism-of-action studies, we have shown that these two separate actions synergize to induce cancer cell death in acute myeloid leukemia (AML) cell and animal models. Owning to the effectiveness of SKI-178, we sought to further refine the chemotype while maintaining "on-target" SKI and MDA activities. Herein, we modified the "linker region" between the substituted phenyl rings of SKI-178 through a structure guided approach. These studies have yielded the discovery of an SKI-178 congener, SKI-349, with log-fold enhancements in both SphK inhibition and cytotoxic potency. Importantly, SKI-349 also demonstrates log-fold improvements in therapeutic efficacy in a retro-viral transduction model of MLL-AF9 AML as compared to previous studies with SKI-178. Together, our results strengthen the hypothesis that simultaneous targeting of the sphingosine kinases (SphK1/2) and the induction of mitotic spindle assembly checkpoint arrest, via microtubule disruption, might be an effective therapeutic strategy for hematological malignancies including AML.
Collapse
Affiliation(s)
- Jeremy A Hengst
- Department of Pharmacology, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA, USA
| | - Shailaja Hegde
- Hoxworth Blood Center, University of Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Robert F Paulson
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, USA
| | - Jong K Yun
- Department of Pharmacology, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA, USA.
| |
Collapse
|
6
|
Roy S, Mahapatra AD, Mohammad T, Gupta P, Alajmi MF, Hussain A, Rehman MT, Datta B, Hassan MI. Design and Development of Novel Urea, Sulfonyltriurea, and Sulfonamide Derivatives as Potential Inhibitors of Sphingosine Kinase 1. Pharmaceuticals (Basel) 2020; 13:E118. [PMID: 32526899 PMCID: PMC7346089 DOI: 10.3390/ph13060118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/30/2020] [Accepted: 06/01/2020] [Indexed: 12/17/2022] Open
Abstract
Sphingosine kinase 1 (SphK1) is one of the well-studied drug targets for cancer and inflammatory diseases. Recently discovered small-molecule inhibitors of SphK1 have been recommended in cancer therapeutics; however, selectivity and potency of first-generation inhibitors are great challenge. In search of effective SphK1 inhibitors, a set of small molecules have been designed and synthesized bearing urea, sulfonylurea, sulfonamide, and sulfonyltriurea groups. The binding affinity of these inhibitors was measured by fluorescence-binding assay and isothermal titration calorimetry. Compounds 1, 5, 6, and 7 showed an admirable binding affinity to the SphK1 in the sub-micromolar range and significantly inhibited SphK1 activity with admirable IC50 values. Molecular docking studies revealed that these compounds fit well into the sphingosine binding pocket of SphK1 and formed significant number of hydrogen bonds and van der Waals interactions. These molecules may be exploited as potent and selective inhibitors of SphK1 that could be implicated in cancer therapeutics after the required in vivo validation.
Collapse
Affiliation(s)
- Sonam Roy
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (S.R.); (T.M.); (P.G.)
| | - Amarjyoti Das Mahapatra
- Department of Chemistry, Indian Institute of Technology, Palaj, Gandhinagar, Gujarat 382355, India;
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (S.R.); (T.M.); (P.G.)
| | - Preeti Gupta
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (S.R.); (T.M.); (P.G.)
| | - Mohamed F. Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.F.A.); (A.H.); (M.T.R.)
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.F.A.); (A.H.); (M.T.R.)
| | - Md. Tabish Rehman
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.F.A.); (A.H.); (M.T.R.)
| | - Bhaskar Datta
- Department of Chemistry, Indian Institute of Technology, Palaj, Gandhinagar, Gujarat 382355, India;
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (S.R.); (T.M.); (P.G.)
| |
Collapse
|
7
|
Lin SY, Miao YR, Hu FF, Hu H, Zhang Q, Li Q, Chen Z, Guo AY. A 6-Membrane Protein Gene score for prognostic prediction of cytogenetically normal acute myeloid leukemia in multiple cohorts. J Cancer 2020; 11:251-259. [PMID: 31892991 PMCID: PMC6930412 DOI: 10.7150/jca.35382] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 09/27/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Cytogenetically normal acute myeloid leukemia (CN-AML) is a large proportion of AMLs with diverse prognostic outcomes. Identifying membrane protein genes as prognostic factors to stratify CN-AML patients will be critical to improve their outcomes. Purpose: This study aims to identify prognostic factors to stratify CN-AML patients to choose better treatments and improve their outcomes. Methods: CN-AML data were from TCGA cohort (n = 79) and four GEO datasets. We identified independent prognostic genes by Cox regression and Kaplan-Meier methods, and constructed linear regression model using LASSO algorithm. The prediction error curve was calculated using R package “pec”. Results: Based on independent prognostic membrane genes, we constructed a regression model for CN-AML prognosis prediction: score = (0.0492 * CD52) - (0.0018 * CD96) + (0.0131 * EMP1) + (0.2058 * TSPAN2) + (0.0234 * STAB1) - (0.3658 * MBTPS1), which was named as MPG6 (6-Membrane Protein Gene) score. Tested in multiple CN-AML datasets, consistent results showed that CN-AML patients with high MPG6 score had poor survival, higher WBC count and shorter EFS. Comparing with other reported scoring models, the benchmark result of MPG6 achieved better association with survival in multiple cohorts. Moreover, by combining with other clinical indicators in CN-AML, MPG6 could improve the performance of survival prediction and serve as a robust prognostic factor. Conclusions: We identified the MPG6 score as a stable indicator with great potential for clinical application in risk stratification and outcome prediction in CN-AML.
Collapse
Affiliation(s)
- Sheng-Yan Lin
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Ya-Ru Miao
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Fei-Fei Hu
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Hui Hu
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Qiong Zhang
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Qiubai Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhichao Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - An-Yuan Guo
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
8
|
Targeting sphingosine kinase 1 for the treatment of pulmonary arterial hypertension. Future Med Chem 2019; 11:2939-2953. [DOI: 10.4155/fmc-2019-0130] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pulmonary arterial hypertension (PAH), characterized by high morbidity and mortality, is a serious hazard to human life. Until now, the long-term survival of the PAH patients is still suboptimal. Recently, sphingosine kinase 1 (SPHK1) has drawn more and more attention due to its essential role in the pulmonary vasoconstriction, remodeling of pulmonary blood vessels and right cardiac lesions in PAH patients, and this enzyme is regarded as a new target for the treatment of PAH. Here, we discussed the multifarious functions of SPHK1 in PAH physiology and pathogenesis. Moreover, the structural features of SPHK1 and binding modes with different inhibitors were summarized. Finally, recent advances in the medicinal chemistry research of SPHK1 inhibitors are presented.
Collapse
|
9
|
Pang H, Wang Y, Gallou F, Lipshutz BH. Fe-Catalyzed Reductive Couplings of Terminal (Hetero)Aryl Alkenes and Alkyl Halides under Aqueous Micellar Conditions. J Am Chem Soc 2019; 141:17117-17124. [DOI: 10.1021/jacs.9b04510] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Haobo Pang
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Ye Wang
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | | | - Bruce H. Lipshutz
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| |
Collapse
|
10
|
Synthetic analogues of marine cytotoxic jaspine B and its stereoisomers. Carbohydr Res 2019; 482:107737. [PMID: 31325781 DOI: 10.1016/j.carres.2019.06.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 11/23/2022]
Abstract
Synthetic analogues of the cytotoxic jaspine B and its stereochemical congeners have become an attractive target in the synthetic organic community owing to the search for novel therapeutic candidates with more potent anticancer activity, as cancer is the second leading cause of death worldwide. This review article provides insights into the different approaches and strategies available in the literature for the construction of jaspine B-related compounds.
Collapse
|
11
|
Inuki S, Miyagawa T, Oishi S, Ohno H. Introduction of a Polar Functional Group to the Lipid Tail of 4- epi-Jaspine B Affects Sphingosine Kinase Isoform Selectivity. Chem Pharm Bull (Tokyo) 2018; 66:866-872. [DOI: 10.1248/cpb.c18-00366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Shinsuke Inuki
- Graduate School of Pharmaceutical Sciences, Kyoto University
| | | | - Shinya Oishi
- Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Hiroaki Ohno
- Graduate School of Pharmaceutical Sciences, Kyoto University
| |
Collapse
|
12
|
Alshaker H, Srivats S, Monteil D, Wang Q, Low CMR, Pchejetski D. Field template-based design and biological evaluation of new sphingosine kinase 1 inhibitors. Breast Cancer Res Treat 2018; 172:33-43. [PMID: 30043096 PMCID: PMC6208908 DOI: 10.1007/s10549-018-4900-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 07/19/2018] [Indexed: 11/29/2022]
Abstract
Purpose Sphingosine kinase 1 (SK1) is a protooncogenic enzyme expressed in many human tumours and is associated with chemoresistance and poor prognosis. It is a potent therapy target and its inhibition chemosensitises solid tumours. Despite recent advances in SK1 inhibitors synthesis and validation, their clinical safety and chemosensitising options are not well described. In this study, we have designed, synthesised and tested a new specific SK1 inhibitor with a low toxicity profile. Methods Field template molecular modelling was used for compound design. Lead compounds were tested in cell and mouse cancer models. Results Field template analysis of three known SK1 inhibitors, SKI-178, 12aa and SK1-I, was performed and compound screening identified six potential new SK1 inhibitors. SK1 activity assays in both cell-free and in vitro settings showed that two compounds were effective SK1 inhibitors. Compound SK-F has potently decreased cancer cell viability in vitro and sensitised mouse breast tumours to docetaxel (DTX) in vivo, without significant whole-body toxicity. Conclusion Through field template screening, we have identified a new SK1 inhibitor, SK-F, which demonstrated antitumour activity in vitro and in vivo without overt toxicity when combined with DTX. Electronic supplementary material The online version of this article (10.1007/s10549-018-4900-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Heba Alshaker
- School of Medicine, University of East Anglia, 2.53 BCRE, Norwich Research Park, Norwich, NR47UQ, UK. .,Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan.
| | - Shyam Srivats
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Danielle Monteil
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Qi Wang
- School of Medicine, University of East Anglia, 2.53 BCRE, Norwich Research Park, Norwich, NR47UQ, UK
| | | | - Dmitri Pchejetski
- School of Medicine, University of East Anglia, 2.53 BCRE, Norwich Research Park, Norwich, NR47UQ, UK.
| |
Collapse
|
13
|
White C, Alshaker H, Cooper C, Winkler M, Pchejetski D. The emerging role of FTY720 (Fingolimod) in cancer treatment. Oncotarget 2018; 7:23106-27. [PMID: 27036015 PMCID: PMC5029614 DOI: 10.18632/oncotarget.7145] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 01/19/2016] [Indexed: 02/07/2023] Open
Abstract
FTY720 (Fingolimod) is a clinically approved immunomodulating therapy for multiple sclerosis that sequesters T-cells to lymph nodes through functional antagonism of sphingosine-1-phosphate 1 receptor. FTY720 also demonstrates a proven efficacy in multiple in vitro and in vivo cancer models, suggesting a potential therapeutic role in cancer patients. A potential anticancer mechanism of FTY720 is through the inhibition of sphingosine kinase 1, a proto-oncogene with in vitro and clinical cancer association. In addition, FTY720's anticancer properties may be attributable to actions on several other molecular targets. This study focuses on reviewing the emerging evidence regarding the anticancer properties and molecular targets of FTY720. While the clinical transition of FTY720 is currently limited by its immune suppression effects, studies aiming at FTY720 delivery and release together with identifying its key synergetic combinations and relevant patient subsets may lead to its rapid introduction into the clinic.
Collapse
Affiliation(s)
| | - Heba Alshaker
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan.,School of Medicine, University of East Anglia, Norwich, UK
| | - Colin Cooper
- School of Medicine, University of East Anglia, Norwich, UK
| | - Matthias Winkler
- Department of Surgery and Cancer, Imperial College London, London, UK
| | | |
Collapse
|
14
|
The Role of Sphingosine-1-Phosphate and Ceramide-1-Phosphate in Inflammation and Cancer. Mediators Inflamm 2017; 2017:4806541. [PMID: 29269995 PMCID: PMC5705877 DOI: 10.1155/2017/4806541] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/01/2017] [Accepted: 08/30/2017] [Indexed: 01/02/2023] Open
Abstract
Inflammation is part of our body's response to tissue injury and pathogens. It helps to recruit various immune cells to the site of inflammation and activates the production of mediators to mobilize systemic protective processes. However, chronic inflammation can increase the risk of diseases like cancer. Apart from cytokines and chemokines, lipid mediators, particularly sphingosine-1-phosphate (S1P) and ceramide-1-phosphate (C1P), contribute to inflammation and cancer. S1P is an important player in inflammation-associated colon cancer progression. On the other hand, C1P has been recognized to be involved in cancer cell growth, migration, survival, and inflammation. However, whether C1P is involved in inflammation-associated cancer is not yet established. In contrast, few studies have also suggested that S1P and C1P are involved in anti-inflammatory pathways regulated in certain cell types. Ceramide is the substrate for ceramide kinase (CERK) to yield C1P, and sphingosine is phosphorylated to S1P by sphingosine kinases (SphKs). Biological functions of sphingolipid metabolites have been studied extensively. Ceramide is associated with cell growth inhibition and enhancement of apoptosis while S1P and C1P are associated with enhancement of cell growth and survival. Altogether, S1P and C1P are important regulators of ceramide level and cell fate. This review focuses on S1P and C1P involvement in inflammation and cancer with emphasis on recent progress in the field.
Collapse
|
15
|
"Dicing and Splicing" Sphingosine Kinase and Relevance to Cancer. Int J Mol Sci 2017; 18:ijms18091891. [PMID: 28869494 PMCID: PMC5618540 DOI: 10.3390/ijms18091891] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 08/29/2017] [Accepted: 08/29/2017] [Indexed: 02/06/2023] Open
Abstract
Sphingosine kinase (SphK) is a lipid enzyme that maintains cellular lipid homeostasis. Two SphK isozymes, SphK1 and SphK2, are expressed from different chromosomes and several variant isoforms are expressed from each of the isozymes, allowing for the multi-faceted biological diversity of SphK activity. Historically, SphK1 is mainly associated with oncogenicity, however in reality, both SphK1 and SphK2 isozymes possess oncogenic properties and are recognized therapeutic targets. The absence of mutations of SphK in various cancer types has led to the theory that cancer cells develop a dependency on SphK signaling (hyper-SphK signaling) or “non-oncogenic addiction”. Here we discuss additional theories of SphK cellular mislocation and aberrant “dicing and splicing” as contributors to cancer cell biology and as key determinants of the success or failure of SphK/S1P (sphingosine 1 phosphate) based therapeutics.
Collapse
|
16
|
Hengst JA, Dick TE, Sharma A, Doi K, Hegde S, Tan SF, Geffert LM, Fox TE, Sharma AK, Desai D, Amin S, Kester M, Loughran TP, Paulson RF, Claxton DF, Wang HG, Yun JK. SKI-178: A Multitargeted Inhibitor of Sphingosine Kinase and Microtubule Dynamics Demonstrating Therapeutic Efficacy in Acute Myeloid Leukemia Models. CANCER TRANSLATIONAL MEDICINE 2017; 3:109-121. [PMID: 28890935 DOI: 10.4103/ctm.ctm_7_17] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
AIM To further characterize the selectivity, mechanism-of-action and therapeutic efficacy of the novel small molecule inhibitor, SKI-178. METHODS Using the state-of-the-art Cellular Thermal Shift Assay (CETSA) technique to detect "direct target engagement" of proteins intact cells, in vitro and in vivo assays, pharmacological assays and multiple mouse models of acute myeloid leukemia (AML). RESULTS Herein, we demonstrate that SKI-178 directly target engages both Sphingosine Kinase 1 and 2. We also present evidence that, in addition to its actions as a Sphingosine Kinase Inhibitor, SKI-178 functions as a microtubule network disrupting agent both in vitro and in intact cells. Interestingly, we separately demonstrate that simultaneous SphK inhibition and microtubule disruption synergistically induces apoptosis in AML cell lines. Furthermore, we demonstrate that SKI-178 is well tolerated in normal healthy mice. Most importantly, we demonstrate that SKI-178 has therapeutic efficacy in several mouse models of AML. CONCLUSION SKI-178 is a multi-targeted agent that functions both as an inhibitor of the SphKs as well as a disruptor of the microtubule network. SKI-178 induced apoptosis arises from a synergistic interaction of these two activities. SKI-178 is safe and effective in mouse models of AML, supporting its further development as a multi-targeted anti-cancer therapeutic agent.
Collapse
Affiliation(s)
- Jeremy A Hengst
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA, USA.,The Jake Gittlen Laboratories for Cancer Research, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Taryn E Dick
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA, USA.,The Jake Gittlen Laboratories for Cancer Research, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Arati Sharma
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA, USA
| | - Kenichiro Doi
- Department of Pediatrics, Penn State Hershey College of Medicine, Hershey, PA, USA
| | - Shailaja Hegde
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Su-Fern Tan
- University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Laura M Geffert
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA, USA.,The Jake Gittlen Laboratories for Cancer Research, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Todd E Fox
- University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Arun K Sharma
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA, USA
| | - Dhimant Desai
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA, USA
| | - Shantu Amin
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA, USA
| | - Mark Kester
- University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Thomas P Loughran
- University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Robert F Paulson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
| | - David F Claxton
- Department of Hematology, Penn State Hershey Cancer Institute, Hershey, PA, USA
| | - Hong-Gang Wang
- Department of Pediatrics, Penn State Hershey College of Medicine, Hershey, PA, USA
| | - Jong K Yun
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA, USA.,The Jake Gittlen Laboratories for Cancer Research, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
17
|
Wiesenfeldt MP, Nairoukh Z, Li W, Glorius F. Hydrogenation of fluoroarenes: Direct access to all-cis-(multi)fluorinated cycloalkanes. Science 2017; 357:908-912. [DOI: 10.1126/science.aao0270] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 07/28/2017] [Indexed: 01/20/2023]
|
18
|
Highly stereoselective synthesis of 1-cyanocyclopropane-carboxamides from 3-substituted-2-cyanoacrylamides with N -tosylhydrazones under metal-free conditions. Tetrahedron Lett 2017. [DOI: 10.1016/j.tetlet.2017.05.070] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
19
|
Menoctone Resistance in Malaria Parasites Is Conferred by M133I Mutations in Cytochrome b That Are Transmissible through Mosquitoes. Antimicrob Agents Chemother 2017; 61:AAC.00689-17. [PMID: 28584157 DOI: 10.1128/aac.00689-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 05/29/2017] [Indexed: 11/20/2022] Open
Abstract
Malaria-related mortality has slowly decreased over the past decade; however, eradication of malaria requires the development of new antimalarial chemotherapies that target liver stages of the parasite and combat the emergence of drug resistance. The diminishing arsenal of anti-liver-stage compounds sparked our interest in reviving the old and previously abandoned compound menoctone. In support of these studies, we developed a new convergent synthesis method that was facile, required fewer steps, produced better yields, and utilized less expensive reagents than the previously published method. Menoctone proved to be highly potent against liver stages of Plasmodium berghei (50 percent inhibitory concentration [IC50] = 0.41 nM) and erythrocytic stages of Plasmodium falciparum (113 nM). We selected for resistance to menoctone and found M133I mutations in cytochrome b of both P. falciparum and P. berghei The same mutation has been observed previously in atovaquone resistance, and we confirmed cross-resistance between menoctone and atovaquone in vitro (for P. falciparum) and in vivo (for P. berghei). Finally, we assessed the transmission potential of menoctone-resistant P. berghei and found that the M133I mutant parasites were readily transmitted from mouse to mosquitoes and back to mice. In each step, the M133I mutation in cytochrome b, inducing menoctone resistance, was confirmed. In summary, this study is the first to show the mechanism of resistance to menoctone and that menoctone and atovaquone resistance is transmissible through mosquitoes.
Collapse
|
20
|
Hatoum D, Haddadi N, Lin Y, Nassif NT, McGowan EM. Mammalian sphingosine kinase (SphK) isoenzymes and isoform expression: challenges for SphK as an oncotarget. Oncotarget 2017; 8:36898-36929. [PMID: 28415564 PMCID: PMC5482707 DOI: 10.18632/oncotarget.16370] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/02/2017] [Indexed: 12/16/2022] Open
Abstract
The various sphingosine kinase (SphK) isoenzymes (isozymes) and isoforms, key players in normal cellular physiology, are strongly implicated in cancer and other diseases. Mutations in SphKs, that may justify abnormal physiological function, have not been recorded. Nonetheless, there is a large and growing body of evidence demonstrating the contribution of gain or loss of function and the imbalance in the SphK/S1P rheostat to a plethora of pathological conditions including cancer, diabetes and inflammatory diseases. SphK is expressed as two isozymes SphK1 and SphK2, transcribed from genes located on different chromosomes and both isozymes catalyze the phosphorylation of sphingosine to S1P. Expression of each SphK isozyme produces alternately spliced isoforms. In recent years the importance of the contribution of SpK1 expression to treatment resistance in cancer has been highlighted and, additionally, differences in treatment outcome appear to also be dependent upon SphK isoform expression. This review focuses on an exciting emerging area of research involving SphKs functions, expression and subcellular localization, highlighting the complexity of targeting SphK in cancer and also comorbid diseases. This review also covers the SphK isoenzymes and isoforms from a historical perspective, from their first discovery in murine species and then in humans, their role(s) in normal cellular function and in disease processes, to advancement of SphK as an oncotarget.
Collapse
Affiliation(s)
- Diana Hatoum
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| | - Nahal Haddadi
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| | - Yiguang Lin
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| | - Najah T. Nassif
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| | - Eileen M. McGowan
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| |
Collapse
|
21
|
Britten CD, Garrett-Mayer E, Chin SH, Shirai K, Ogretmen B, Bentz TA, Brisendine A, Anderton K, Cusack SL, Maines LW, Zhuang Y, Smith CD, Thomas MB. A Phase I Study of ABC294640, a First-in-Class Sphingosine Kinase-2 Inhibitor, in Patients with Advanced Solid Tumors. Clin Cancer Res 2017; 23:4642-4650. [PMID: 28420720 DOI: 10.1158/1078-0432.ccr-16-2363] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 10/11/2016] [Accepted: 04/11/2017] [Indexed: 01/17/2023]
Abstract
Purpose: Sphingosine kinases (SK1 and SK2) regulate tumor growth by generating the mitogenic and proinflammatory lipid sphingosine 1-phosphate (S1P). This phase I study investigated the safety, pharmacokinetics, pharmacodynamics, and antitumor activity of ABC294640, a first-in-class orally available inhibitor of SK2.Experimental Design: Escalating doses of ABC294640 were administered orally to patients with advanced solid tumors in sequential cohorts at the following dose levels: 250 mg qd, 250 mg bid, 500 mg bid, and 750 mg bid, continuously in cycles of 28 days. Serial blood samples were obtained to measure ABC294640 concentrations and sphingolipid profiles.Results: Twenty-two patients were enrolled, and 21 received ABC294640. The most common drug-related toxicities were nausea, vomiting, and fatigue. Among the 4 patients at 750 mg bid, one had dose-limiting grade 3 nausea and vomiting, and 2 were unable to complete cycle 1 due to diverse drug-related toxicities. The 500 mg bid dose level was established as the recommended phase II dose. ABC294640 administration resulted in decreases in S1P levels over the first 12 hours, with return to baseline at 24 hours. The best response was a partial response in a patient with cholangiocarcinoma at 250 mg qd, and stable disease was observed in 6 patients with various solid tumors across dose levels.Conclusions: At 500 mg bid, ABC294640 is well tolerated and achieves biologically relevant plasma concentrations. Changes in plasma sphingolipid levels may provide a useful pharmacodynamic biomarker for ABC294640. Clin Cancer Res; 23(16); 4642-50. ©2017 AACR.
Collapse
Affiliation(s)
- Carolyn D Britten
- Division of Hematology/Oncology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Elizabeth Garrett-Mayer
- Department of Population Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Steven H Chin
- Division of Hematology/Oncology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Keisuke Shirai
- Division of Hematology/Oncology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Besim Ogretmen
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Tricia A Bentz
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Alan Brisendine
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Kate Anderton
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Susan L Cusack
- Apogee Biotechnology Corporation, Hummelstown, Pennsylvania
| | - Lynn W Maines
- Apogee Biotechnology Corporation, Hummelstown, Pennsylvania
| | - Yan Zhuang
- Apogee Biotechnology Corporation, Hummelstown, Pennsylvania
| | - Charles D Smith
- Apogee Biotechnology Corporation, Hummelstown, Pennsylvania.
| | - Melanie B Thomas
- Division of Hematology/Oncology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
22
|
LeBlanc FR, Liu X, Hengst J, Fox T, Calvert V, Petricoin EF, Yun J, Feith DJ, Loughran TP. Sphingosine kinase inhibitors decrease viability and induce cell death in natural killer-large granular lymphocyte leukemia. Cancer Biol Ther 2016; 16:1830-40. [PMID: 26252351 DOI: 10.1080/15384047.2015.1078949] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Sphingolipid metabolism has been identified as a potential therapeutic target in cancer. Sphingosine-1-phosphate (S1P) is a potent bioactive sphingolipid metabolite produced by sphingosine kinases-1 and -2 (SPHK1 and SPHK2). Elevated SPHK1 has been found in numerous cancer types and been shown to contribute to survival, chemotherapeutic resistance and malignancy. However, its role in large granular Natural Killer (NK) large granular lymphocyte (LGL) leukemia has not been investigated. Here, we examine SPHK1 as a therapeutic target in LGL leukemia. We found that SPHK1 is overexpressed in peripheral blood mononuclear cells (PBMCs) from LGL leukemia patients which results in elevated S1P in the sera. The use of SPHK1 inhibitors, SKI-II or SKI-178, decreased leukemic NK cell viability and induced caspase-dependent apoptosis. SKI-II and SKI-178 restored the sphingolipid balance by increasing ceramide and decreasing S1P in leukemic NKL cells. SKI-II and SKI-178 also induced apoptosis in primary NK-LGLs from leukemia patients. Mechanistic studies in NK-LGL cell lines demonstrated that SKI-178 and SKI-II induced cell cycle arrest at G2/M. We found that SKI-178 induced phosphorylation of Bcl-2 at Ser70, and that this was dependent on CDK1. We further show that SPHK1 inhibition with SKI-178 leads to decreased JAK-STAT signaling. Our data demonstrate that SPHK1 represents a novel therapeutic target for the treatment of NK-LGL leukemia.
Collapse
Affiliation(s)
- Francis R LeBlanc
- a Penn State Hershey Cancer Institute; Penn State College of Medicine ; Hershey , PA USA.,b University of Virginia Cancer Center; University of Virginia ; Charlottesville , VA USA
| | - Xin Liu
- a Penn State Hershey Cancer Institute; Penn State College of Medicine ; Hershey , PA USA
| | - Jeremy Hengst
- c Department of Pharmacology ; Penn State College of Medicine ; Hershey , PA USA
| | - Todd Fox
- b University of Virginia Cancer Center; University of Virginia ; Charlottesville , VA USA
| | - Valerie Calvert
- d Center for Applied Proteomics and Molecular Medicine; George Mason University ; Manassas , VA USA
| | - Emanuel F Petricoin
- d Center for Applied Proteomics and Molecular Medicine; George Mason University ; Manassas , VA USA
| | - Jong Yun
- c Department of Pharmacology ; Penn State College of Medicine ; Hershey , PA USA
| | - David J Feith
- b University of Virginia Cancer Center; University of Virginia ; Charlottesville , VA USA
| | - Thomas P Loughran
- b University of Virginia Cancer Center; University of Virginia ; Charlottesville , VA USA
| |
Collapse
|
23
|
Pitman MR, Costabile M, Pitson SM. Recent advances in the development of sphingosine kinase inhibitors. Cell Signal 2016; 28:1349-1363. [DOI: 10.1016/j.cellsig.2016.06.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/09/2016] [Accepted: 06/09/2016] [Indexed: 12/11/2022]
|
24
|
Lynch KR, Thorpe SB, Santos WL. Sphingosine kinase inhibitors: a review of patent literature (2006-2015). Expert Opin Ther Pat 2016; 26:1409-1416. [PMID: 27539678 DOI: 10.1080/13543776.2016.1226282] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Sphingosine kinase (SphK1 & SphK2) is the sole source of the pleiotropic lipid mediator, sphingosine-1-phosphate (S1P). S1P has been implicated in a variety of diseases such as cancer, Alzheimer's disease, sickle cell disease and fibrosis and thus the biosynthetic route to S1P is a logical target for drug discovery. Areas covered: In this review, the authors consider the SphK inhibitor patent literature from 2006-2016 Q1 with the emphasis on composition of matter utility patents. The Espacenet database was queried with the search term 'sphingosine AND kinase' to identify relevant literature. Expert opinion: Early inhibitor discovery focused on SphK1 with a bias towards oncology indications. Structurally, the reported inhibitors occupy the sphingosine 'J-shaped' binding pocket. The lack of cytotoxicity with improved SphK1 inhibitors raises doubt about the enzyme as an oncology target. SphK2 inhibitors are featured in more recent patent applications. Interestingly, both SphK1 and SphK2 inhibition and gene 'knockout' share opposing effects on circulating S1P levels: SphK1 inhibition/gene ablation decreases, while SphK2 inhibition/gene ablation increases, blood S1P. As understanding of S1P's physiological roles increases and more drug-like SphK inhibitors emerge, inhibiting one or both SphK isotypes could provide unique strategies for treating disease.
Collapse
Affiliation(s)
- Kevin R Lynch
- a Department of Pharmacology , University of Virginia , Charlottesville , VA , USA
| | | | - Webster L Santos
- c Department of Chemistry and Virginia Tech Center for Drug Discovery , Virginia Tech , Blacksburg , VA , USA
| |
Collapse
|
25
|
Vogt D, Stark H. Therapeutic Strategies and Pharmacological Tools Influencing S1P Signaling and Metabolism. Med Res Rev 2016; 37:3-51. [PMID: 27480072 DOI: 10.1002/med.21402] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 06/01/2016] [Accepted: 06/28/2016] [Indexed: 02/06/2023]
Abstract
During the last two decades the study of the sphingolipid anabolic, catabolic, and signaling pathways has attracted enormous interest. Especially the introduction of fingolimod into market as first p.o. therapeutic for the treatment of multiple sclerosis has boosted this effect. Although the complex regulation of sphingosine-1-phosphate (S1P) and other catabolic and anabolic sphingosine-related compounds is not fully understood, the influence on different (patho)physiological states from inflammation to cytotoxicity as well as the availability of versatile pharmacological tools that represent new approaches to study these states are described. Here, we have summarized various aspects concerning the many faces of sphingolipid function modulation by different pharmacological tools up to clinical candidates. Due to the immense heterogeneity of physiological or pharmacological actions and complex cross regulations, it is difficult to predict their role in upcoming therapeutic approaches. Currently, inflammatory, immunological, and/or antitumor aspects are discussed.
Collapse
Affiliation(s)
- Dominik Vogt
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, D-60438, Frankfurt, Germany
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitätsstraße 1, D-40225, Düsseldorf, Germany
| |
Collapse
|
26
|
Arora P, Narang R, Nayak SK, Singh SK, Judge V. 2,4-Disubstituted thiazoles as multitargated bioactive molecules. Med Chem Res 2016. [DOI: 10.1007/s00044-016-1610-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
27
|
Development of hydroxy-based sphingosine kinase inhibitors and anti-inflammation in dextran sodium sulfate induced colitis in mice. Bioorg Med Chem 2016; 24:3218-30. [DOI: 10.1016/j.bmc.2016.05.047] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/20/2016] [Accepted: 05/21/2016] [Indexed: 01/01/2023]
|
28
|
Tiper IV, East JE, Subrahmanyam PB, Webb TJ. Sphingosine 1-phosphate signaling impacts lymphocyte migration, inflammation and infection. Pathog Dis 2016; 74:ftw063. [PMID: 27354294 DOI: 10.1093/femspd/ftw063] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2016] [Indexed: 01/01/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) is a sphingosine containing lipid intermediate obtained from ceramide. S1P is known to be an important signaling molecule and plays multiple roles in the context of immunity. This lysophospholipid binds and activates G-protein-coupled receptors (GPCRs) known as S1P receptors 1-5 (S1P1-5). Once activated, these GPCRs mediate signaling that can lead to alterations in cell proliferation, survival or migration, and can also have other effects such as promoting angiogenesis. In this review, we will present evidence demonstrating a role for S1P in lymphocyte migration, inflammation and infection, as well as in cancer. The therapeutic potential of targeting S1P receptors, kinases and lyase will also be discussed.
Collapse
Affiliation(s)
- Irina V Tiper
- Department of Microbiology and Immunology, University of Maryland School of Medicine and the Marlene and Stewart Greenebaum Cancer Center, 685 W Baltimore St., Baltimore, MD 21201, USA
| | - James E East
- Department of Microbiology and Immunology, University of Maryland School of Medicine and the Marlene and Stewart Greenebaum Cancer Center, 685 W Baltimore St., Baltimore, MD 21201, USA
| | - Priyanka B Subrahmanyam
- Department of Microbiology and Immunology, University of Maryland School of Medicine and the Marlene and Stewart Greenebaum Cancer Center, 685 W Baltimore St., Baltimore, MD 21201, USA
| | - Tonya J Webb
- Department of Microbiology and Immunology, University of Maryland School of Medicine and the Marlene and Stewart Greenebaum Cancer Center, 685 W Baltimore St., Baltimore, MD 21201, USA
| |
Collapse
|
29
|
Houck JD, Dawson TK, Kennedy AJ, Kharel Y, Naimon ND, Field SD, Lynch KR, Macdonald TL. Structural Requirements and Docking Analysis of Amidine-Based Sphingosine Kinase 1 Inhibitors Containing Oxadiazoles. ACS Med Chem Lett 2016; 7:487-92. [PMID: 27190598 DOI: 10.1021/acsmedchemlett.6b00002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/01/2016] [Indexed: 12/18/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) is a potent growth-signaling lipid that has been implicated in cancer progression, inflammation, sickle cell disease, and fibrosis. Two sphingosine kinases (SphK1 and 2) are the source of S1P; thus, inhibitors of the SphKs have potential as targeted cancer therapies and will help to clarify the roles of S1P and the SphKs in other hyperproliferative diseases. Recently, we reported a series of amidine-based inhibitors with high selectivity for SphK1 and potency in the nanomolar range. However, these inhibitors display a short half-life. With the goal of increasing metabolic stability and maintaining efficacy, we designed an analogous series of molecules containing oxadiazole moieties. Generation of a library of molecules resulted in the identification of the most selective inhibitor of SphK1 reported to date (705-fold selectivity over SphK2), and we found that potency and selectivity vary significantly depending on the particular oxadiazole isomer employed. The best inhibitors were subjected to in silico molecular dynamics docking analysis, which revealed key insights into the binding of amidine-based inhibitors by SphK1. Herein, the design, synthesis, biological evaluation, and docking analysis of these molecules are described.
Collapse
Affiliation(s)
- Joseph D. Houck
- Department of Chemistry, University of Virginia, McCormick Road, Charlottesville, Virginia 22904, United States
| | - Thomas K. Dawson
- Department of Chemistry, University of Virginia, McCormick Road, Charlottesville, Virginia 22904, United States
| | - Andrew J. Kennedy
- Department of Chemistry, University of Virginia, McCormick Road, Charlottesville, Virginia 22904, United States
| | - Yugesh Kharel
- Department of Pharmacology, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22904, United States
| | - Niels D. Naimon
- Department of Chemistry, University of Virginia, McCormick Road, Charlottesville, Virginia 22904, United States
| | - Saundra D. Field
- Department of Chemistry, University of Virginia, McCormick Road, Charlottesville, Virginia 22904, United States
| | - Kevin R. Lynch
- Department of Pharmacology, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22904, United States
| | - Timothy L. Macdonald
- Department of Chemistry, University of Virginia, McCormick Road, Charlottesville, Virginia 22904, United States
- Department of Pharmacology, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22904, United States
| |
Collapse
|
30
|
Aurelio L, Scullino CV, Pitman MR, Sexton A, Oliver V, Davies L, Rebello RJ, Furic L, Creek DJ, Pitson SM, Flynn BL. From Sphingosine Kinase to Dihydroceramide Desaturase: A Structure-Activity Relationship (SAR) Study of the Enzyme Inhibitory and Anticancer Activity of 4-((4-(4-Chlorophenyl)thiazol-2-yl)amino)phenol (SKI-II). J Med Chem 2016; 59:965-84. [PMID: 26780304 DOI: 10.1021/acs.jmedchem.5b01439] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The sphingosine kinase (SK) inhibitor, SKI-II, has been employed extensively in biological investigations of the role of SK1 and SK2 in disease and has demonstrated impressive anticancer activity in vitro and in vivo. However, interpretations of results using this pharmacological agent are complicated by several factors: poor SK1/2 selectivity, additional activity as an inducer of SK1-degradation, and off-target effects, including its recently identified capacity to inhibit dihydroceramide desaturase-1 (Des1). In this study, we have delineated the structure-activity relationship (SAR) for these different targets and correlated them to that required for anticancer activity and determined that Des1 inhibition is primarily responsible for the antiproliferative effects of SKI-II and its analogues. In the course of these efforts, a series of novel SK1, SK2, and Des1 inhibitors have been generated, including compounds with significantly greater anticancer activity.
Collapse
Affiliation(s)
- Luigi Aurelio
- Monash Institute of Pharmaceutical Science, Monash University , 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Carmen V Scullino
- Monash Institute of Pharmaceutical Science, Monash University , 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Melissa R Pitman
- Centre for Cancer Biology, University of South Australia and SA Pathology , Frome Road, Adelaide South Australia 5000, Australia
| | - Anna Sexton
- Monash Institute of Pharmaceutical Science, Monash University , 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Victoria Oliver
- Monash Institute of Pharmaceutical Science, Monash University , 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Lorena Davies
- Centre for Cancer Biology, University of South Australia and SA Pathology , Frome Road, Adelaide South Australia 5000, Australia
| | - Richard J Rebello
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Clayton, Victoria 3800, Australia
| | - Luc Furic
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Clayton, Victoria 3800, Australia
| | - Darren J Creek
- Monash Institute of Pharmaceutical Science, Monash University , 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology , Frome Road, Adelaide South Australia 5000, Australia
| | - Bernard L Flynn
- Monash Institute of Pharmaceutical Science, Monash University , 381 Royal Parade, Parkville, Victoria 3052, Australia
| |
Collapse
|
31
|
Kharel Y, Morris EA, Congdon MD, Thorpe SB, Tomsig JL, Santos WL, Lynch KR. Sphingosine Kinase 2 Inhibition and Blood Sphingosine 1-Phosphate Levels. J Pharmacol Exp Ther 2015; 355:23-31. [PMID: 26243740 DOI: 10.1124/jpet.115.225862] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 07/23/2015] [Indexed: 01/14/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) levels are significantly higher in blood and lymph than in tissues. This S1P concentration difference is necessary for proper lymphocyte egress from secondary lymphoid tissue and to maintain endothelial barrier integrity. Studies with mice lacking either sphingosine kinase (SphK) type 1 and 2 indicate that these enzymes are the sole biosynthetic source of S1P, but they play different roles in setting S1P blood levels. We have developed a set of drug-like SphK inhibitors, with differing selectivity for the two isoforms of this enzyme. Although all SphK inhibitors tested decrease S1P when applied to cultured U937 cells, only those inhibitors with a bias for SphK2 drove a substantial increase in blood S1P in mice and this rise was detectable within minutes of administration of the inhibitor. Blood S1P also increased in response to SphK2 inhibitors in rats. Mass-labeled S1P was cleared more slowly after intravenous injection into SphK2 inhibitor-treated mice or mice lacking a functional SphK2 gene; thus, the increased accumulation of S1P in the blood appears to result from the decreased clearance of S1P from the blood. Therefore, SphK2 appears to have a function independent of generating S1P in cells. Our results suggest that differential SphK inhibition with a drug might afford a method to manipulate blood S1P levels in either direction while lowering tissue S1P levels.
Collapse
Affiliation(s)
- Yugesh Kharel
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia (Y.K., J.L.T., K.R.L.); Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia (E.A.M., M.D.C., W.L.S.); and SphynKx Therapeutics LLC, Charlottesville, Virginia (S.B.T.)
| | - Emily A Morris
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia (Y.K., J.L.T., K.R.L.); Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia (E.A.M., M.D.C., W.L.S.); and SphynKx Therapeutics LLC, Charlottesville, Virginia (S.B.T.)
| | - Molly D Congdon
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia (Y.K., J.L.T., K.R.L.); Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia (E.A.M., M.D.C., W.L.S.); and SphynKx Therapeutics LLC, Charlottesville, Virginia (S.B.T.)
| | - Steven B Thorpe
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia (Y.K., J.L.T., K.R.L.); Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia (E.A.M., M.D.C., W.L.S.); and SphynKx Therapeutics LLC, Charlottesville, Virginia (S.B.T.)
| | - Jose L Tomsig
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia (Y.K., J.L.T., K.R.L.); Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia (E.A.M., M.D.C., W.L.S.); and SphynKx Therapeutics LLC, Charlottesville, Virginia (S.B.T.)
| | - Webster L Santos
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia (Y.K., J.L.T., K.R.L.); Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia (E.A.M., M.D.C., W.L.S.); and SphynKx Therapeutics LLC, Charlottesville, Virginia (S.B.T.)
| | - Kevin R Lynch
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia (Y.K., J.L.T., K.R.L.); Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia (E.A.M., M.D.C., W.L.S.); and SphynKx Therapeutics LLC, Charlottesville, Virginia (S.B.T.)
| |
Collapse
|
32
|
Structure-based functional annotation of hypothetical proteins from Candida dubliniensis: a quest for potential drug targets. 3 Biotech 2015; 5:561-576. [PMID: 28324558 PMCID: PMC4522726 DOI: 10.1007/s13205-014-0256-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 09/30/2014] [Indexed: 12/24/2022] Open
Abstract
Candida dubliniensis is an emerging pathogenic yeast in humans and infections are usually restricted to mucosal parts of the body. However, its presence in specimens of immunocompromised individuals, especially in HIV-positive patients, is of major medical concern. There is a large fraction of genomes of C. dubliniensis in the database which are uncharacterized for their biochemical, biophysical, and/or cellular functions, and are identified as hypothetical proteins (HPs). Function annotation of Candida genome is, therefore, essentially required to facilitate the understanding of mechanisms of pathogenesis and biochemical pathways important for selecting novel therapeutic target. Here, we carried out an extensive analysis to explain the functional properties of genome, using available protein structure and function analysis tools. We successfully modeled the structures of eight HPs for which a template with moderate sequence similarity was available in the protein data bank. All modeled structures were analyzed and we found that these proteins may act as transporter, kinase, transferase, ketosteroid, isomerase, hydrolase, oxidoreductase, and binding targets for DNA and RNA. Since these unique HPs of Candida showed no homologs in humans, these proteins are expected to be a potential target for future antifungal therapy.
Collapse
|
33
|
Patwardhan NN, Morris EA, Kharel Y, Raje MR, Gao M, Tomsig JL, Lynch KR, Santos WL. Structure-activity relationship studies and in vivo activity of guanidine-based sphingosine kinase inhibitors: discovery of SphK1- and SphK2-selective inhibitors. J Med Chem 2015; 58:1879-1899. [PMID: 25643074 DOI: 10.1021/jm501760d] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sphingosine 1-phosphate (S1P) is a pleiotropic signaling molecule that acts as a ligand for five G-protein coupled receptors (S1P1-5) whose downstream effects are implicated in a variety of important pathologies including sickle cell disease, cancer, inflammation, and fibrosis. The synthesis of S1P is catalyzed by sphingosine kinase (SphK) isoforms 1 and 2, and hence, inhibitors of this phosphorylation step are pivotal in understanding the physiological functions of SphKs. To date, SphK1 and 2 inhibitors with the potency, selectivity, and in vivo stability necessary to determine the potential of these kinases as therapeutic targets are lacking. Herein, we report the design, synthesis, and structure-activity relationship studies of guanidine-based SphK inhibitors bearing an oxadiazole ring in the scaffold. Our studies demonstrate that SLP120701, a SphK2-selective inhibitor (Ki = 1 μM), decreases S1P levels in histiocytic lymphoma (U937) cells. Surprisingly, homologation with a single methylene unit between the oxadiazole and heterocyclic ring afforded a SphK1-selective inhibitor in SLP7111228 (Ki = 48 nM), which also decreased S1P levels in cultured U937 cells. In vivo application of both compounds, however, resulted in contrasting effect in circulating levels of S1P. Administration of SLP7111228 depressed blood S1P levels while SLP120701 increased levels of S1P. Taken together, these compounds provide an in vivo chemical toolkit to interrogate the effect of increasing or decreasing S1P levels and whether such a maneuver can have implications in disease states.
Collapse
Affiliation(s)
- Neeraj N Patwardhan
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Emily A Morris
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Yugesh Kharel
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Mithun R Raje
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Ming Gao
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Jose L Tomsig
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Kevin R Lynch
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Webster L Santos
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
34
|
Abstract
The transfer of the gamma phosphate from ATP to sphingosine (Sph) to generate a small signaling molecule, sphingosine 1-phosphate (S1P), is catalyzed by sphingosine kinases (SphK), which exist as two isoforms, SphK1 and SphK2. SphK is a key regulator of S1P and the S1P:Sph/ceramide ratio. Increases in S1P levels have been linked to diseases including sickle cell disease, cancer, and fibrosis. Therefore, SphKs are potential targets for drug discovery. However, the current chemical biology toolkit needed to validate these enzymes as drug targets is inadequate. With this review, we survey in vivo active SphK inhibitors and highlight the need for developing more potent and selective inhibitors.
Collapse
Affiliation(s)
- Webster L. Santos
- Department
of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Kevin R. Lynch
- Department
of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| |
Collapse
|
35
|
Dick TE, Hengst JA, Fox TE, Colledge AL, Kale VP, Sung SS, Sharma A, Amin S, Loughran TP, Kester M, Wang HG, Yun JK. The apoptotic mechanism of action of the sphingosine kinase 1 selective inhibitor SKI-178 in human acute myeloid leukemia cell lines. J Pharmacol Exp Ther 2015; 352:494-508. [PMID: 25563902 DOI: 10.1124/jpet.114.219659] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We previously developed SKI-178 (N'-[(1E)-1-(3,4-dimethoxyphenyl)ethylidene]-3-(4-methoxxyphenyl)-1H-pyrazole-5-carbohydrazide) as a novel sphingosine kinase-1 (SphK1) selective inhibitor and, herein, sought to determine the mechanism-of-action of SKI-178-induced cell death. Using human acute myeloid leukemia (AML) cell lines as a model, we present evidence that SKI-178 induces prolonged mitosis followed by apoptotic cell death through the intrinsic apoptotic cascade. Further examination of the mechanism of action of SKI-178 implicated c-Jun NH2-terminal kinase (JNK) and cyclin-dependent protein kinase 1 (CDK1) as critical factors required for SKI-178-induced apoptosis. In cell cycle synchronized human AML cell lines, we demonstrate that entry into mitosis is required for apoptotic induction by SKI-178 and that CDK1, not JNK, is required for SKI-178-induced apoptosis. We further demonstrate that the sustained activation of CDK1 during prolonged mitosis, mediated by SKI-178, leads to the simultaneous phosphorylation of the prosurvival Bcl-2 family members, Bcl-2 and Bcl-xl, as well as the phosphorylation and subsequent degradation of Mcl-1. Moreover, multidrug resistance mediated by multidrug-resistant protein1 and/or prosurvival Bcl-2 family member overexpression did not affect the sensitivity of AML cells to SKI-178. Taken together, these findings highlight the therapeutic potential of SKI-178 targeting SphK1 as a novel therapeutic agent for the treatment of AML, including multidrug-resistant/recurrent AML subtypes.
Collapse
Affiliation(s)
- Taryn E Dick
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Jeremy A Hengst
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Todd E Fox
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Ashley L Colledge
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Vijay P Kale
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Shen-Shu Sung
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Arun Sharma
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Shantu Amin
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Thomas P Loughran
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Mark Kester
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Hong-Gang Wang
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Jong K Yun
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| |
Collapse
|
36
|
Wang X, Sun C, Fang L, Yin D. Discovery of novel sphingosine kinase 1 inhibitorsvia structure-based hierarchical virtual screening. MEDCHEMCOMM 2015. [DOI: 10.1039/c4md00312h] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Hierarchical structure-based virtual screening against the sphingosine kinase 1(SphK1) binding pocket was performed. 25 compounds were selected for biological evaluation. Compound 25 exhibited comparable SphK1 and SphK2 inhibitory activities and anti-proliferative effects on U937 cells to the positive control N,N-dimethylsphingosine (DMS) 1. Further molecule dynamic (MD) simulations revealed the binding mode between SphK1 and 25.
Collapse
Affiliation(s)
- Xiaojian Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines
- Institute of Materia Medica
- Peking Union Medical College
- Chinese Academy of Medical Sciences
- Beijing 100050
| | - Chenbin Sun
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines
- Institute of Materia Medica
- Peking Union Medical College
- Chinese Academy of Medical Sciences
- Beijing 100050
| | - Liang Fang
- Department of Medicinal Chemistry
- Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation
- Institute of Materia Medica
- Peking Union Medical College
- Chinese Academy of Medical Sciences
| | - Dali Yin
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines
- Institute of Materia Medica
- Peking Union Medical College
- Chinese Academy of Medical Sciences
- Beijing 100050
| |
Collapse
|
37
|
Mo DL, Pecak WH, Zhao M, Wink DJ, Anderson LL. Synthesis of N-Styrenyl Amidines from α,β-Unsaturated Nitrones and Isocyanates through CO2 Elimination and Styrenyl Migration. Org Lett 2014; 16:3696-9. [DOI: 10.1021/ol501503a] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Dong-Liang Mo
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Wiktoria H. Pecak
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Meng Zhao
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Donald J. Wink
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Laura L. Anderson
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| |
Collapse
|
38
|
Chemical and genetic tools to explore S1P biology. Curr Top Microbiol Immunol 2014; 378:55-83. [PMID: 24728593 PMCID: PMC7120161 DOI: 10.1007/978-3-319-05879-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The zwitterionic lysophospholipid Sphingosine 1-Phosphate (S1P) is a pleiotropic mediator of physiology and pathology. The synthesis, transport, and degradation of S1P are tightly regulated to ensure that S1P is present in the proper concentrations in the proper location. The binding of S1P to five G protein-coupled S1P receptors regulates many physiological systems, particularly the immune and vascular systems. Our understanding of the functions of S1P has been aided by the tractability of the system to both chemical and genetic manipulation. Chemical modulators have been generated to affect most of the known components of S1P biology, including agonists of S1P receptors and inhibitors of enzymes regulating S1P production and degradation. Genetic knockouts and manipulations have been similarly engineered to disrupt the functions of individual S1P receptors or enzymes involved in S1P metabolism. This chapter will focus on the development and utilization of these chemical and genetic tools to explore the complex biology surrounding S1P and its receptors, with particular attention paid to the in vivo findings that these tools have allowed for.
Collapse
|
39
|
Plano D, Amin S, Sharma AK. Importance of sphingosine kinase (SphK) as a target in developing cancer therapeutics and recent developments in the synthesis of novel SphK inhibitors. J Med Chem 2014; 57:5509-24. [PMID: 24471412 DOI: 10.1021/jm4011687] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Sphingosine kinase (SphK) is an oncogenic lipid kinase that regulates the sphingolipid metabolic pathway that has been shown to play a role in numerous hyperproliferative/inflammatory diseases. The SphK isoforms (SphK1 and SphK2) catalyze the conversion of the proapoptotic substrate d-erythrosphingosine to the promitogenic/migratory product sphingosine 1-phosphate (S1P). Accumulation of S1P has been linked to the development/progression of cancer and various other diseases including, but not limited to, asthma, inflammatory bowel disease, rheumatoid arthritis, and diabetic nephropathy. SphK therefore represents a potential new target for developing novel therapeutics for cancer and other diseases. This finding has stimulated the development and evaluation of numerous SphK inhibitors over the past decade or so. In this review, we highlight the recent advancement in the field of SphK inhibitors including SphK1 and SphK2 specific inhibitors. Both sphingolipid based and nolipidic small molecule inhibitors and their importance in treatment of cancer and other diseases are discussed.
Collapse
Affiliation(s)
- Daniel Plano
- Department of Pharmacology, Penn State Hershey Cancer Institute, CH72, Penn State College of Medicine , 500 University Drive, Hershey, Pennsylvania 17033, United States
| | | | | |
Collapse
|
40
|
Baek DJ, MacRitchie N, Anthony NG, Mackay SP, Pyne S, Pyne NJ, Bittman R. Structure-activity relationships and molecular modeling of sphingosine kinase inhibitors. J Med Chem 2013; 56:9310-27. [PMID: 24164513 PMCID: PMC3848335 DOI: 10.1021/jm401399c] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
![]()
The
design, synthesis, and evaluation of the potency of new isoform-selective
inhibitors of sphingosine kinases 1 and 2 (SK1 and SK2), the enzyme
that catalyzes the phosphorylation of d-erythro-sphingosine to produce the key signaling lipid, sphingosine 1-phosphate,
are described. Recently, we reported that 1-(4-octylphenethyl)piperidin-4-ol
(RB-005) is a selective inhibitor of SK1. Here we report
the synthesis of 43 new analogues of RB-005, in which
the lipophilic tail, polar headgroup, and linker region were modified
to extend the structure–activity relationship profile for this
lead compound, which we explain using modeling studies with the recently
published crystal structure of SK1. We provide a basis for the key
residues targeted by our profiled series and provide further evidence
for the ability to discriminate between the two isoforms using pharmacological
intervention.
Collapse
Affiliation(s)
- Dong Jae Baek
- Department of Chemistry and Biochemistry, Queens College, The City University of New York , Flushing, New York 11367-1597, United States
| | | | | | | | | | | | | |
Collapse
|
41
|
Fleury LM, Wilson EE, Vogt M, Fan TJ, Oliver AG, Ashfeld BL. Amine-free approach toward N-toluenesulfonyl amidine construction: a phosphite-mediated Beckmann-like coupling of oximes and p-toluenesulfonyl azide. Angew Chem Int Ed Engl 2013; 52:11589-93. [PMID: 24038711 DOI: 10.1002/anie.201305141] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Indexed: 11/06/2022]
Abstract
Atom hopping: A chlorophosphite-mediated Beckmann ligation of oximes and p-toluenesulfonyl azide gives access to N-sulfonyl phosphoramidines in good to excellent yields. The reaction proceeds under exceptionally mild conditions and constitutes a bioorthogonal approach toward amidines by avoiding the use of amines and transition-metal catalysts. dmp-ol=3,3-dimethylpropanediol.
Collapse
Affiliation(s)
- Lauren M Fleury
- Department of Chemistry and Biochemistry, University of Notre Dame, 251 Nieuwland Science Hall, Notre Dame, IN 46556 (USA)
| | | | | | | | | | | |
Collapse
|
42
|
Fleury LM, Wilson EE, Vogt M, Fan TJ, Oliver AG, Ashfeld BL. Amine-Free Approach towardN-Toluenesulfonyl Amidine Construction: A Phosphite-Mediated Beckmann-Like Coupling of Oximes andp-Toluenesulfonyl Azide. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201305141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
43
|
Cencetti F, Bernacchioni C, Tonelli F, Roberts E, Donati C, Bruni P. TGFβ1 evokes myoblast apoptotic response via a novel signaling pathway involving S1P4 transactivation upstream of Rho-kinase-2 activation. FASEB J 2013; 27:4532-46. [PMID: 23913862 DOI: 10.1096/fj.13-228528] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In view of its multiple detrimental effects, transforming growth factor β1 (TGFβ1) is recognized as critical negative regulator of skeletal muscle repair. Apoptosis of skeletal muscle precursor cells driven by TGFβ1 contributes to the negative role exerted by the cytokine in tissue repair, although the underlying molecular mechanisms are still elusive. Herein we report the identification of a new signaling pathway, relying on Rho kinase-2 stimulation, subsequent to SMAD-dependent S1P4 up-regulation and transactivation via sphingosine kinase (SK)-2, that accounts for TGFβ1-induced apoptosis in cultured myoblasts. S1P4-specific gene silencing reduced by almost 50% activation of caspase-3 and poly-ADP ribosyl transferase cleavage elicited by TGFβ1. Moreover, the selective S1P4 antagonist CYM50358 also reduced the TGFβ1 proapoptotic effects. By employing pharmacological and molecular biological approaches, the involvement of SK2 and ROCK2 in the transmission of the TGFβ1 apoptotic action was also demonstrated. These results reinforce the notion that the SK/S1P axis plays a fundamental role in TGFβ1 mode of action in skeletal muscle cells and, by disclosing a novel mechanism by which TGFβ1 exerts its harmful action, pinpoint new molecular targets that in principle could be beneficial in the treatment of several skeletal muscle disorders or aging-dependent muscle atrophy.
Collapse
Affiliation(s)
- Francesca Cencetti
- 2Dipartimento di Scienze Biomediche, Sperimentali e Cliniche, Università di Firenze, Viale G.B. Morgagni 50, 50134 Florence, Italy.
| | | | | | | | | | | |
Collapse
|
44
|
Structure guided design of a series of sphingosine kinase (SphK) inhibitors. Bioorg Med Chem Lett 2013; 23:4608-16. [PMID: 23845219 DOI: 10.1016/j.bmcl.2013.06.030] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 06/04/2013] [Accepted: 06/11/2013] [Indexed: 11/21/2022]
Abstract
Sphingosine-1-phosphate (S1P) signaling plays a vital role in mitogenesis, cell migration and angiogenesis. Sphingosine kinases (SphKs) catalyze a key step in sphingomyelin metabolism that leads to the production of S1P. There are two isoforms of SphK and observations made with SphK deficient mice show the two isoforms can compensate for each other's loss. Thus, inhibition of both isoforms is likely required to block SphK dependent angiogenesis. A structure based approach was used to design and synthesize a series of SphK inhibitors resulting in the identification of the first potent inhibitors of both isoforms of human SphK. Additionally, to our knowledge, this series of inhibitors contains the only sufficiently potent inhibitors of murine SphK1 with suitable physico-chemical properties to pharmacologically interrogate the role of SphK1 in rodent models and to reproduce the phenotype of SphK1 (-/-) mice.
Collapse
|
45
|
Alshaker H, Sauer L, Monteil D, Ottaviani S, Srivats S, Böhler T, Pchejetski D. Therapeutic potential of targeting SK1 in human cancers. Adv Cancer Res 2013; 117:143-200. [PMID: 23290780 DOI: 10.1016/b978-0-12-394274-6.00006-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Sphingosine kinase 1 (SK1) is a lipid enzyme with oncogenic properties that converts the proapoptotic lipids ceramide and sphingosine into the antiapoptotic lipid sphingosine-1-phosphate and activates the signal transduction pathways that lead to cell proliferation, migration, the activation of the inflammatory response, and the impairment of apoptosis. There is compelling evidence that SK1 activation contributes to cancer progression leading to increased oncogenic transformation, tumor growth, resistance to therapies, tumor neovascularization, and metastatic spread. High levels of SK1 expression or activity have been associated with a poor prognosis in several human cancers. Recent studies using cancer cell and mouse models demonstrate a significant potential for SK1-targeting therapies to synergize with the effects of chemotherapy and radiotherapy; however, until recently the absence of clinically applicable SK1 inhibitors has limited the translation of these findings into patients. With the recent discovery of SK1 inhibiting properties of a clinically approved drug FTY720 (Fingolimod), SK1 has gained significant attention from both clinicians and the pharmaceutical industry and it is hoped that trials of newly developed SK1 inhibitors may follow soon. This review provides an overview of the SK1 signaling, its relevance to cancer progression, and the potential clinical significance of targeting SK1 for improved local or systemic control of human cancers.
Collapse
Affiliation(s)
- Heba Alshaker
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
The role of sphingolipids as bioactive signaling molecules that can regulate cell fate decisions puts them at center stage for cancer treatment and prevention. While ceramide and sphingosine have been established as antigrowth molecules, sphingosine-1-phosphate (S1P) offers a progrowth message to cells. The enzymes responsible for maintaining the balance between these "stop" or "go" signals are the sphingosine kinases (SK), SK1 and SK2. While the relative contribution of SK2 is still being elucidated and may involve an intranuclear role, a substantial amount of evidence suggests that regulation of sphingolipid levels by SK1 is an important component of carcinogenesis. Here, we review the literature regarding the role of SK1 as an oncogene that can function to enhance cancer cell viability and promote tumor growth and metastasis; highlighting the importance of developing specific SK1 inhibitors to supplement current cancer therapies.
Collapse
Affiliation(s)
- Linda A Heffernan-Stroud
- Molecular and Cellular Biology and Pathobiology Program, Medical University of South Carolina, Charleston, SC, USA
| | | |
Collapse
|
47
|
Wallington-Beddoe CT, Bradstock KF, Bendall LJ. Oncogenic properties of sphingosine kinases in haematological malignancies. Br J Haematol 2013; 161:623-638. [PMID: 23521541 DOI: 10.1111/bjh.12302] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The sphingosine kinases (SphKs) have relatively recently been implicated in contributing to malignant cellular processes with particular interest in the oncogenic properties of SPHK1. Whilst SPHK1 has received considerable attention as a putative oncoprotein, SPHK2 has been much more difficult to study, with often conflicting data surrounding its role in cancer. Initial studies focused on non-haemopoietic malignancies, however a growing body of literature on the role of sphingolipid metabolism in haemopoietic malignancies is now emerging. This review provides an overview of the current state of knowledge of the SphKs and the bioactive lipid sphingosine 1-phosphate (S1P), the product of the reaction they catalyse. It then reviews the current literature regarding the roles of S1P and the SphKs in haemopoietic malignancies and discusses the compounds currently available that modulate sphingolipid metabolism and their potential and shortcomings as therapeutic agents for the treatment of haematological malignancies.
Collapse
Affiliation(s)
- Craig T Wallington-Beddoe
- Westmead Institute for Cancer Research, Westmead Millennium Institute, The University of Sydney, Sydney, NSW, Australia
| | | | - Linda J Bendall
- Westmead Institute for Cancer Research, Westmead Millennium Institute, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
48
|
Liu K, Guo TL, Hait NC, Allegood J, Parikh HI, Xu W, Kellogg GE, Grant S, Spiegel S, Zhang S. Biological characterization of 3-(2-amino-ethyl)-5-[3-(4-butoxyl-phenyl)-propylidene]-thiazolidine-2,4-dione (K145) as a selective sphingosine kinase-2 inhibitor and anticancer agent. PLoS One 2013; 8:e56471. [PMID: 23437140 PMCID: PMC3577900 DOI: 10.1371/journal.pone.0056471] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 01/08/2013] [Indexed: 12/20/2022] Open
Abstract
In our effort to develop selective sphingosine kinase-2 (SphK2) inhibitors as pharmacological tools, a thiazolidine-2,4-dione analogue, 3-(2-amino-ethyl)-5-[3-(4-butoxyl-phenyl)-propylidene]-thiazolidine-2,4-dione (K145), was synthesized and biologically characterized. Biochemical assay results indicate that K145 is a selective SphK2 inhibitor. Molecular modeling studies also support this notion. In vitro studies using human leukemia U937 cells demonstrated that K145 accumulates in U937 cells, suppresses the S1P level, and inhibits SphK2. K145 also exhibited inhibitory effects on the growth of U937 cells as well as apoptotic effects in U937 cells, and that these effects may be through the inhibition of down-stream ERK and Akt signaling pathways. K145 also significantly inhibited the growth of U937 tumors in nude mice by both intraperitoneal and oral administration, thus demonstrating its in vivo efficacy as a potential lead anticancer agent. The antitumor activity of K145 was also confirmed in a syngeneic mouse model by implanting murine breast cancer JC cells in BALB/c mice. Collectively, these results strongly encourage further optimization of K145 as a novel lead compound for development of more potent and selective SphK2 inhibitors.
Collapse
Affiliation(s)
- Kai Liu
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Tai L. Guo
- Departments of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Nitai C. Hait
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Jeremy Allegood
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Hardik I. Parikh
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Wenfang Xu
- School of Pharmaceutical Sciences, Shandong University, Shandong, People’s Republic of China
| | - Glen E. Kellogg
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Steven Grant
- Department of Internal Medicine and Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Shijun Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia, United States of America
- * E-mail:
| |
Collapse
|
49
|
Zheng S, Zhong Q, Jiang Q, Mottamal M, Zhang Q, Zhu N, Burow ME, Worthylake RA, Wang G. Discovery of a Series of Thiazole Derivatives as Novel Inhibitors of Metastatic Cancer Cell Migration and Invasion. ACS Med Chem Lett 2013; 4:191-196. [PMID: 23526571 DOI: 10.1021/ml300322n] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Effective inhibitors of cancer cell migration and invasion can potentially lead to clinical applications as therapy to block tumor metastasis, the primary cause of death in cancer patients. To this end we have designed and synthesized a series of thiazole derivatives that showed potent efficacy against cell migration and invasion in metastatic cancer cells. The most effective compound, 5k, was found to have an IC50 value of 176 nM in the dose-dependent transwell migration assays in MDA-MB-231cells. At the dose of 10 μM, 5k also blocked about 80% of migration in HeLa and A549 cells and 60% of invasion of MDA-MB-231 cells. Importantly, the majority of the derivatives exhibited no apparent cytotoxicity in the clonogenic assays. The low to negligible inhibition of cell proliferation is a desirable property of these anti-migration derivatives because they hold promise of low toxicity to healthy cells as potential therapeutic agents. Mechanistic studies analyzing the actin cytoskeleton by microscopy demonstrate that compound 5k substantially reduced cellular f-actin, and prevented localization of fascin to actin-rich membrane protrusions. These results suggest that the anti-migration activity may result from impaired actin structures in protrusions that are necessary to drive migration.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Matthew E. Burow
- Department of Medicine, Section of Hematology and Medical Oncology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Rebecca A. Worthylake
- Department of Oral Biology, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70119, United States
| | | |
Collapse
|
50
|
Tripathi SK, Singh VP, Gupta KC, Kumar P. Hydrophobic and membrane permeable polyethylenimine nanoparticles efficiently deliver nucleic acids in vitro and in vivo. J Mater Chem B 2013; 1:2515-2524. [DOI: 10.1039/c3tb00481c] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|